Вы находитесь на странице: 1из 743

Principles of Cancer Biotherapy

5th Edition
Principles of Cancer
Biotherapy
5th Edition

Edited by
Robert K. Oldham
Hematology-Oncology Associates of Southeast Missouri Hospital
Southeast Medical Plaza
Cape Girardeau, MO
USA

Robert O. Dillman
Hoag Cancer Center
Newport Beach, CA
USA
University of California
Irvine, CA
USA
Robert K. Oldham Robert O. Dillman
Hematology-Oncology Associates Hoag Cancer Center
of Southeast Missouri Hospital Newport Beach, CA
Southeast Medical Plaza USA
Cape Girardeau, MO University of California
USA Irvine, CA
USA

ISBN 978-90-481-2277-6 e-ISBN 978-90-481-2289-9


DOI 10.1007/978-90-481-229-9
Springer Dordrecht Heidelberg London New York

Library of Congress Control Number: 2009929387

First edition published 1987 by Raven Press


Second edition published 1993 by Williams & Wilkins
Third and Fourth edition published 1998 Kluwer Academic Publishers
© Springer Science+Business Media B.V. 2009
No part of this work may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic, mechanical,
photocopying, microfilming, recording or otherwise, without written permission from the Publisher, with the exception of any material sup-
plied specifically for the purpose of being entered and executed on a computer system, for exclusive use by the purchaser of the work.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


Preface

The idea for the first edition of Principles of Cancer those mediated by their active sites. There is increasing
Biotherapy was formulated in the early 1980s. As the evidence that drug development will focus on the inter-
founding director of the Biological Response Modifiers action between the smaller active regions of these large
Program for the National Cancer Institute from 1980- biological molecules and their receptors. This opens up
1984, one of us (rko) envisioned a textbook that would a broad field of molecular design for extending and
embody the principles of the then fledgling fourth modal- improving the therapeutic activities of natural biological
ity of cancer treatment - biotherapy. Contributing authors molecules. This has recently been extended to small
were solicited in 1985, and the first edition came off the molecules interacting with DNA/RNA (anti-sense), with
presses in 1987. Principles represented the first compre- enzymes such as tyrosine kinase and with growth and
hensive textbook on the use of cancer biotherapy and vascular factor receptors. The last decade has been
summarized the work done in this field through 1986. extraordinarily productive in the development of new
The second edition of Principles was published in anticancer drugs through chemical and biological manip-
1991 about the time biotherapy was more broadly recog- ulation of these natural molecules. Thus, the body itself
nized as the fourth major cancer treatment modality. has become the “medicine cabinet” of the future.
Subsequent textbooks by DeVita, Hellman and Rosenberg In the 1990s and beyond into this millennium, medi-
[1] in 1991, Mitchell [2] in 1993 and Rosenberg [3] in cine will face extraordinary demands. While technol-
2000 validated the importance of this modality in ogy brings us tremendous opportunity, it also highlights
cancer care. problems in our medical care system. Most new tech-
This third edition was published in 1998 and con- nology is expensive and, as it comes from the laboratory
firmed the tremendous progress that had been made in to the clinic, is by its very nature untried and unproven.
the previous five years using biologicals in cancer treat- Our medical care system involves a private and govern-
ment. It was generally agreed that biopharmaceuticals ment insurance reimbursement system that favors pay-
were producing major opportunities for new cancer ing for marginally effective medical care of the past
therapies. Cancer biotherapy was emerging as a more rather than innovative medical treatments of the future.
specific and targeted form of systemic cancer treatment. Such a system is inhibitory to the development of effec-
Cancer growth control was also becoming an effective tive new anti-cancer medicines.
method of treatment complementing cancer destruction To more rapidly and efficiently exploit the opportuni-
as mechanisms of cancer treatment and “cure”. ties in cancer biotherapy in the future, patients, employ-
The fourth edition of Principles was published in ers, insurers, universities, and government must come
2003. It was apparent that biotherapy and the use of bio- together and redefine the system of reimbursement to
pharmaceuticals had not only become recognized as the maximize the patient’s opportunity for access to new
fourth modality of cancer treatment, it was clear that and potentially effective cancer therapies. To simply
biopharmaceuticals had become the dominant form of reimburse for old ineffective or marginally effective
new cancer therapeutics which in the future will replace treatment is not the answer. Provisions must be made to
less selective, more toxic forms of therapy. fund clinical research and afford these new approaches
For years, the chemical manipulation of small mole- broader use at the bedside. We must develop methods to
cules has been pursued in drug development. We now allow our patients access to the opportunities of the
have all the tools for the biological manipulation of natu- future, while maintaining solid support for effective
ral substances for therapeutic use. In fact, as we better therapies of the past. No longer is it acceptable to pay
understand the interaction between biological molecules only for medical care that utilizes old technology, such
and their receptors, it is clear that biological manipula- as chemotherapy, that is approved but only marginally
tion and chemical manipulation are coming together to effective. Across the broad spectrum of human malig-
bring molecular medicine to the bedside. Many biologi- nancies, most chemotherapeutic drugs are toxic and of
cal molecules are large and have functions other than limited medical value. We must support clinical research

v
vi Preface

in its efforts to bring newer methods of cancer treatment These chapters illustrate some of these new methods of
to the clinic, methods that are less toxic and more thinking and illustrate new strategies for the treatment
effective. and control of cancer. It is always difficult to move from
We believe cancer biotherapy will ultimately replace past dogmas to future opportunities, but this fifth edi-
much of what we utilize today in cancer treatment. In tion of Principles of Cancer Biotherapy illustrates why it
light of this view, we want to thank all the authors for is so important for researchers, regulators and clinicians
their dedication to purpose in writing this fifth edition of to explore and apply these new opportunities in cancer
Principles. This book summarizes an evolving science biotherapy to the benefit of our patients.
and a rapidly changing medical practice. As we progress
into the millennium, it now becomes possible to envision Robert K. Oldham, M.D. Robert O. Dillman MD
a much more diversified system of cancer research and
treatment that will afford greater opportunities for our
patients. As indicated in some of the chapters in Principles,
there is increasing evidence that our historical “kill and
References
cure” outlook in cancer treatment is in need of modifica- 1. DeVita VT Jr, Hellman S, Rosenberg SA. Biologic Therapy of
Cancer. J.B. Lippincott, 1991.
tion. Some forms of cancer biotherapy use the strategy 2. Mitchell MS. Biological Approaches to Cancer Treatment.
of tumor growth stabilization and control through con- McGraw-Hill, 1993.
tinued biological therapy over a longer period of time, 3. Rosenberg SA. Principles and Practices of the Biologic Therapy of
akin to the use of insulin in the treatment of diabetes. Cancer. J.B. Lippincott, 2000.
Contents

Preface v
Robert K. Oldham

Contributors ix

1. Cancer biotherapy: general principles 1


Robert K. Oldham

2. The pathogenesis of cancer metastasis: relevance to therapy 17


Sun-Jin Kim, Cheryl Hunt Baker, Yasuhiko Kitadai, Toru Nakamura,
Toshio Kuwai, Takamitsu Sasaki, Robert Langley, and Isaiah J. Fidler

3. Developmental therapeutics and the design of clinical trials 41


Robert K. Oldham

4. Recombinant proteins and genomics in cancer therapy 53


Kapil Mehta, Bulent Ozpolat, Kishorchandra Gohil, and Bharat B. Aggarwal

5. Current concepts in immunology 85


Robert K. Oldham

6. Therapeutic approaches to cancer-associated immune suppression 101


Robert K. Oldham

7. Cancer vaccines 147


Kenneth A. Foon and Malek M. Safa

8. Cytokines 155
Walter M. Lewko and Robert K. Oldham

9. Interferons: therapy for cancer 277


David Goldstein, Robert Jones, Richard V. Smalley, and Ernest C. Borden

10. Monoclonal antibody therapy 303


Robert O. Dillman

11. Immunotoxins 407


Arthur E. Frankel, Jung-Hee Woo, and David M. Neville

12. Drug Immunoconjugates 451


Malek Safa, Kenneth A. Foon, and Robert K. Oldham

13. Targeted radionuclide therapy of cancer 463


John M. Pagel, Otto C. Boerman, Hazel B. Breitz, and Ruby Meredith

vii
viii Contents

14. Stem cell/bone marrow transplantation as biotherapy 497


Robert K. Oldham

15. Cellular immunotherapy (CI), where have we been and where are we going? 505
John R. Yannelli

16. Growth and differentiation factors as cancer therapeutics 527


Kapil Mehta, Robert K. Oldham, and Bulent Ozpolat

17. Granulocyte colony-stimulating factor: biology and clinical potential 569


MaryAnn Foote and George Morstyn

18. Granulocyte-macrophage colony-stimulating factor 581


Maryann Foote and George Morstyn

19. Cancer gene therapy 589


Donald J. Buchsbaum, C. Ryan Miller, Lacey R. Mcnally, and Sergey A. Kaliberov

20. Regulatory process for approval of biologicals for cancer therapy 613
Antonio J. Grillo-López

21.1. Cancer biotherapy: 2009 disease-related activity 631


Robert K. Oldham and Robert O. Dillman

21.2. Biological therapy of melanoma 633


Robert K. Oldham

21.3. Biological therapy of genitourinary cancer 645


Robert K. Oldham

21.4. Biological therapy of colon cancer 659


Robert O. Dillman

21.5. Biological therapy of breast cancer 669


Robert O. Dillman

21.6. Biological therapy of lung cancer 679


Robert O. Dillman

21.7. Biological therapy of B and T cell lymphoproliferative disorders 693


Robert O. Dillman

21.8. Biological therapy of multiple myeloma 711


Robert K. Oldham

21.9. Biological therapy of squamous cell cancers of the head and neck 713
Robert O. Dillman

21.10. Biological therapy of glioblastoma 723


Robert O. Dillman

22. Speculations for 2009 and beyond 733


Robert K. Oldham

Index 739
Contributors

Robert K. Oldham Toshio Kuwai


Hematology-Oncology Associates of Southeast Department of Cancer Biology
Missouri Hospital M. D. Anderson Cancer Center
Southeast Medical Plaza The University of Texas
Cape Girardeau, MO Houston, TX
USA USA

Robert O. Dillman Takamitsu Sasaki


Hoag Cancer Center Department of Cancer Biology
Newport Beach, CA M. D. Anderson Cancer Center
USA The University of Texas
University of California Houston, TX
Irvine, CA USA
USA
Robert Langley
Sun-Jin Kim Department of Cancer Biology
Department of Cancer Biology M. D. Anderson Cancer Center
M. D. Anderson Cancer Center The University of Texas
The University of Texas Houston, TX
Houston, TX USA
USA
Isaiah J. Fidler
Cheryl Hunt Baker Department of Cancer Biology
Department of Cancer Biology M. D. Anderson Cancer Center
M. D. Anderson Cancer Center The University of Texas
The University of Texas Houston, TX
Houston, TX USA
USA
Kapil Mehta
Yasuhiko Kitadai Department of Experimental Therapeutics
Department of Cancer Biology M. D. Anderson Cancer Center
M. D. Anderson Cancer Center The University of Texas
The University of Texas Houston, TX
Houston, TX USA
USA
Bulent Ozpolat
Toru Nakamura Department of Experimental Therapeutics
Department of Cancer Biology M. D. Anderson Cancer Center
M. D. Anderson Cancer Center The University of Texas
The University of Texas Houston, TX
Houston, TX USA
USA

ix
x Contributors

Kishorchandra Gohil Jung-Hee Woo


Department of Internal Medicine Cancer Research Institute of Scott &White
The University of California Temple, TX
Davis, CA USA
USA
David M. Neville
Bharat B. Aggarwal Angimmune LLC,
Department of Experimental Therapeutics Bethesda, MD
M. D. Anderson Cancer Center USA
The University of Texas
Houston, TX John M. Pagel
USA The Fred Hutchinson Cancer
Research Center
Kenneth A. Foon Seattle, WA
Department of Hematological Malignancies USA
Nevada Cancer Institute
Las Vegas, NV Otto C. Boerman
USA University Medical Center
Nijmegen
Malek M. Safa The Netherlands
Division of Hematology / Oncology
University of Cincinnati Hazel B. Breitz
Cincinnati, OH Poniard Pharmaceuticals, Inc.
USA Seattle, WA
USA
Walter M. Lewko
Cancer Cellular Therapeutics, Inc. Ruby F. Meredith
Cape Girardeau, MO Department of Radiation Oncology
USA University of Alabama Birmingham
Birmingham, Alabama
David Goldstein USA
Department of Medical Oncology
Prince of Wales Hospital John R. Yannelli
Randwick, Sydney Department of Microbiology,
Australia Immunology and Molecular Genetics
Markey Cancer Center
Robert Jones University of Kentucky,
Centre for Oncology and Applied Pharmacology School of Medicine
CRUK Beatson Laboratories Lexington, Kentucky
Glasgow, Scotland USA
UK
MaryAnn Foote
Richard V. Smalley (deceased) Thousand Oaks, CA
Synertron Inc. USA
Madison, WI
USA Dr George Morstyn
Department of Microbiology
Arthur E. Frankel Monash University
Cancer Research Institute of Scott &White Victoria
Temple, TX Australia
USA
Contributors xi

Donald J. Buchsbaum C. Ryan Miller


Department of Radiation Oncology Department of Pathology
University of Alabama at Birmingham University of North Carolina
Birmingham, AL Chapel Hill, NC
USA USA

Lacey R. McNally Antonio J. Grillo-López


Department of Radiology Rancho Santa Fé, CA
University of Alabama at Birmingham USA
Birmingham, AL
USA
Sergey A. Kaliberov
Department of Radiation Oncology
University of Alabama at Birmingham
Birmingham, AL
USA
1 Cancer biotherapy: general principles
ROBERT K. OLDHAM

The term biotherapy encompasses the therapeutic use of ception associated with immunotherapy: biotherapy can
any biological substance, but more specifically, it con- be effective against clinically apparent, even bulky, can-
notes the use of products of the mammalian genome. cer, and treatment should not be restricted to situations
With modern techniques of genetic engineering, the where the tumor cell mass is imperceptible [64, 71].
mammalian genome represents the new “medicine cabi- Thus, the clinical trial designs for biotherapy can be
net.” Biological response modifiers (BRM) are agents similar to those used previously for other modalities of
and approaches whose mechanisms of action involve the cancer treatment, as long as one measures both pharma-
individual’s own biological responses. Biologicals and cokinetics and the biological responses affected by these
BRM work through diverse mechanisms in the biother- approaches [65]. Testing is continuing for biotherapy
apy of cancer. They may (a) augment the host’s defenses using the interferons, lymphokines, chemokines and
through the administration of cells, natural biologicals, or cytokines, growth and maturation factors, angiostatic
the synthetic derivatives thereof as effectors or mediators factors, monoclonal antibodies and their immunoconju-
(direct or indirect) of an antitumor response; (b) increase gates, vaccines, and cellular therapy [68].
the individual’s antitumor responses through augmenta-
tion or restoration of effector mechanisms, or decrease a
component of the host’s reaction that is deleterious; (c)
augment the individual’s responses using modified tumor
Historical Perspectives
cells or vaccines to stimulate a greater response, or The use of chemical and biological compounds to mod-
increase tumor cell sensitivity to an existing biological ulate biological responses has been under active investi-
response; (d) decrease transformation and/or increase dif- gation for over 30 years. Although various chemicals,
ferentiation or maturation of tumor cells; (e) interfere bacterial extracts, and viruses have been found to modu-
with growth-promoting factors and angiogenesis induc- late immune responses in experimental animals, and, to
ing factors produced by tumor cells; (f) decrease or arrest a more limited extent, in humans, these “nonspecific”
the tendency of tumor cells to metastasize to other sites; immunomodulators have not been highly effective in
(g) increase the ability of the patient to tolerate damage clinical trials [71]. Molecular biologists have recently
by cytotoxic modalities of cancer treatment; and/or (h) developed techniques for the isolation of genes and their
use biological molecules to target and bind to cancer cells subsequent translation into appropriate production sys-
and induce more effective cytostatic or cytocidial antitu- tems. These methods make available virtually unlimited
mor activity. quantities of highly purified biological compounds for
While several of these approaches involve the aug- experimental and therapeutic use. As a result, several
mentation or use of biological responses, an understand- classes of biologicals are being evaluated in preclinical
ing of the biological properties of immune response models and clinical trials (Table 1).
molecules, growth and maturation factors, and other The continued investigation of nonspecific immuno-
biological substances will assist in the development of modulators and adjuvants, as well as the recent advent
specific molecular entities that can act on biological of genetically engineered biologicals, makes the need
responses and/or act directly on tumor cells. Thus, one for predictive preclinical assays of biological activity
can visualize the development of biological approaches and efficacy apparent [33]. In vitro assays of biological
with response-modifying as well as direct cytolytic, activity (bioassays) are generally used to define and
cytostatic, growth-inhibiting (antiproliferative), or mat- quantitate the activity of a given biological substance.
urational effects on tumor cells. Subsequently, flow cytometry, immunoperoxidase staining,
Biotherapy is the fourth modality of cancer therapy enzyme-linked immunosorbent assays (ELISA), tetramer
and can be effective alone or in association with surgery, assays, radioimmunoassays (RIA), and variations of these
radiotherapy, and chemotherapy. To put biotherapy into methods allow the precise determination of levels of these
perspective, it is important to dispel a historical miscon- molecules and activities in appropriate fluids and tissues.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 1


© Springer Science + Business Media B.V. 2009
2 Cancer biotherapy: general principles

Table 1. Biologicals and BRM cancer and the conclusions drawn from them are unlikely
Immunomodulators (chemicals, bacterial extracts, viruses, etc.) to be broadly applicable to human cancers [33].
Lymphokines/cytokines α, β, γ-interferon; IL-1–32, tumor The modern era of cancer treatment began in the
necrosis factor (TNF); etc. 1950s with the recognition that most cancers were sys-
Growth/maturation factors (CSF, IL-2, EPO, etc.) temic problems. It became obvious that lymphatic and
Effector cells (cytotoxic and helper T cells, NK, and LAK cells,
blood-borne metastases often occurred simultaneously
gene-engineered cells, etc.)
Tumor-associated antigens and gene-engineered cellular with local growth and regional spread. The early success
vaccines of chemotherapy in leukemia and lymphoma prompted a
Monoclonal antibodies massive and enthusiastic search for chemicals that might
Immunoconjugates have cytolytic or cytostatic effects on cancer cells.
Millions of compounds have been “screened” for antitu-
Finally, there is the need to assess the in vivo activity of mor activity [11], and this effort has given clinicians less
these materials in preclinical models to develop predic- than 75 “approved and active” anticancer chemotherapy
tive assays for clinical efficacy and provide information drugs [84]. In addition, there are about 15 approved hor-
useful in the rational selection of agents and the design of monal agents, ten targeted small molecules, and 25 bio-
clinical trials [33, 54]. logical drugs. There is now widespread recognition that
Given the variability in the biological behavior of only a few drugs in cancer treatment can effectively pal-
cancer and its interface with the human outbred host, it liate and sometimes cure [19]. The development of three
is not surprising that trials of nonspecific and specific modalities (surgery, radiotherapy and chemotherapy)
immunotherapy, as translated from artificially con- and their subsequent integration into what is now multi-
structed animal models, have not been uniformly suc- modal cancer treatment has been summarized [18, 20].
cessful in cancer treatment [4, 33]. Naturally occurring Between 1975 and 1990 a plateau was reached in cancer
cancers arise in a particular organ from one cell or a few treatment. New surgical techniques (e.g., debulking,
cells under some carcinogenic stimulus. In humans, intra-operative methods for radiotherapy, and catheter
these initial foci of cancer cells may grow – and some- isolation/infusion for chemotherapy) and new methods
times lie dormant – over very long periods of time (from of radiotherapy (e.g., neutrons, protons, interstitial ther-
1% to 30% of the human life span) before there is clini- apy, isotopes) continue to be developed, but these two
cal evidence of cancer. Dissemination of cells from the modalities are primarily useful in local and regional can-
initial focus may occur at any time during the develop- cer treatment. There has been continued, but slow, prog-
ment of the primary tumor. Subsequently, growth and ress in the treatment of highly replicative, drug-sensitive
metastasis occur over periods of months to years from malignancies over the past 15 years. It is now apparent
primary and secondary foci, causing complex biological that further progress with chemotherapy will depend on
interactions to occur. a greater understanding of the metabolic and enzymatic
In contrast, experimentally induced cancer is an arti- processes of cancer cells and the differences between
ficial (even artifactual) situation. A short-duration, high- these and normal cells. In addition, there are the prob-
dose carcinogen may be used to induce cancer quickly lems of drug resistance, selectivity of action, and drug
so that experiments can proceed rapidly. In transplant- delivery. Cancer cells are more like than unlike normal
able models, the tumor cells are injected into young, cells with respect to chemotherapy sensitivity but they
normal, syngeneic animals, thereby circumventing the are more likely to become chemotherapy resistant with
influences of environmental or genetic factors that may drug exposure. Many chemotherapeutic agents are
be operative in the natural host during tumor develop- highly cytolytic, but the problems of normal tissue tox-
ment. Many of the experimental models are transplant- icity, drug delivery, and tumor-cell resistance remain
able tumors that have been maintained for decades and [19, 46]. Thus, cancer remains a systemic problem, and
have only the most remote relevance to cancer in further systemic but more selective approaches are
humans. The injection represents a single, instantaneous required for more effective treatment [20].
point source for a defined tumor load that has been In addition to standard chemotherapy drugs that are
manipulated in vitro. Regardless of whether that tumor approved and in clinical use, there are approximately 15
load is 10 or 106 cells, it is being placed artificially in a hormonal drugs available. These are primarily used in
single site and allowed to grow and metastasize from breast cancer and prostate cancer, and many are varia-
that selected and artificial, single site. Thus, these trans- tions on the theme of blocking hormonal receptors. The
plantable cancers are simply not analogous to clinical evolution in this area has been primarily testing new
Robert K. Oldham 3

hormonal agents for superiority in therapy and for lesser negative results. Thus, immunotherapy had a poor image
degrees of toxicity when used in large-scale trials against by the end of the 1970s [71, 109].
hormonally sensitive tumors. Immunotherapy failed to establish itself as a major
Then newest area of drug development is in targeted mode of cancer treatment for several reasons. One impor-
therapy. Here, small molecules are tested for the ability to tant factor was the lack of definition and purity of immu-
block a particular receptor, enzyme, or target which may notherapeutic agents. Many of the nonspecific approaches
be involved in cell replication or cell metabolism. Eight involved the use of complex chemicals, bacteria, viruses,
targeted agents are currently available in the clinic, and a and poorly defined extracts in an attempt to “stimulate”
very large number of these small molecules are currently the immune response. Thus, molecular definition of the
in clinical trials, most of which are variations on the actual stimulating entity was not available. Given the lack
theme of blocking a particular receptor or enzyme system of analogy between model systems and humans, the
with many of these drugs being multifunctional in that poorly characterized reagents, and the problems of vari-
they can block more than one receptor or system. ability in experimental procedures, the lack of demon-
The scientific basis for biotherapy as the fourth modal- strable clinical efficacy was predictable [71].
ity of cancer treatment is now firm [21–23, 68, 70, 81, 82, Immunotherapy is not an appropriate term for the
94, 97]. Historically, there was an attempt to establish modern use of biologicals and BRM in medicine.
immunotherapy in this role. Whereas immunotherapy Biological control mechanisms should be envisioned on
was reproducible under specific experimental protocols, a much broader basis than the immune system. While
it was not strikingly effective in animals bearing palpable immunotherapy remains a subcategory of biotherapy,
tumors and did not translate well to patients. Given the growth and differentiation factors, chemokines and
observation that immunotherapy was more effective with angiostatic factors, the use of synthetically derived
small tumor burdens, investigators began to study both molecular analogs, and the pharmacological exploita-
“specific” and “nonspecific” immunotherapy as treatment tion of biological molecules is much broader than
for minimal residual disease. Although it became widely immunotherapy (Table 1) [68].
accepted that the treatment of animals with minimal Certain specific developments led to biotherapy
residual disease was analogous to the postsurgical treat- becoming the fourth modality of cancer therapy.
ment of cancer in humans, this analogy was often Advances in molecular biology have given scientists the
stretched beyond reason. Immunotherapy in young, nor- capability to clone individual genes and produce signifi-
mal, syngeneic animals was often begun on the day of the cant quantities of highly purified genomic products as
tumor transplant (or within 1 or 2 days), using a trans- medicines. Unlike extracted and purified biological
plant of a very small number of tumor cells (1–1,000) to molecules, available in small quantities as semi-purified
a single site. In many of these studies, and in studies in mixtures, the products of cloned genes have a level of
which the tumor was surgically resected and no evident purity on a par with drugs. They can be analyzed alone
disease remained, the effects of immunotherapy were rea- or in combination as to their effects in cancer biology. In
sonably reproducible; the therapy was most beneficial addition, progress in nucleic acid sequencing and trans-
when the tumor mass was less than 106 cells. lation, protein sequencing and synthesis, the isolation
These experimental results produced a dogma that and purification of biological products, and mass cell
immunological manipulation or immunotherapy could culture has given the scientific community the opportu-
work only when the tumor cell mass was imperceptible nity to alter nucleic acids and proteins at the nucleotide
[4, 71]. This posed real problems for clinical immuno- or amino acid level to manipulate then optimize their
therapy, since the tumor cell mass at clinical diagnosis biological activity [42]. The elucidation of the human
or after surgery is usually three orders of magnitude (or genome and the encoded products broadened the oppor-
more) greater than 106 cells. Despite the obvious diffi- tunities for advancements in biotherapy.
culties with the experimental models and the translation As a result of gene cloning, a major approach in can-
to humans, clinicians began larger scale immunotherapy cer treatment has evolved. Interleukin-2 can be used to
trials in the 1970s. The results of initial, small, uncon- stimulate the growth of a broad range of lymphocytes
trolled trials were often reported as positive. Larger, ran- (T, NK, and LAK cells). This has given clinicians the
domized, controlled studies were done to confirm the ability to have large quantities of specific subclasses of
efficacy of a particular immunotherapeutic regimen in a effector cells for cancer treatment. Emerging evidence
particular type of cancer. Although some of the con- suggests that these effector cells can be helpful in the
trolled studies were positive, most yielded marginal or regional and systemic treatment of advanced, bulky
4 Cancer biotherapy: general principles

cancer. It was the availability of Interleukin-2 that neoplasms [33]. There may exist in animals and in
allowed this technology to prosper [83]. In addition, humans specific or nonspecific defects important in the
IL-2 is now being used as the T cell growth factor for development of their autochthonous tumors. Corrections
gene engineered lymphocytes containing new genes to of such defects may require a form of biotherapy totally
enhance their cancer treatment capacity [80, 103]. different from that required to assist the normal host in
Another major technical advance was the discovery controlling a transplanted cancer.
of hybridomas. A major limitation on the use of anti-
bodies had been the inability to make reproducible high-
titer, specific antisera and to define these preparations
Model Screening Criteria
on a molecular basis. Immunoglobulin reagents can now Theoretically, an ideal procedure for screening new bio-
be produced with the same level of molecular purity as logicals should employ a system of sequential and pro-
cloned gene products and drugs. Processes to easily gressively more demanding protocols designed to select
“humanize” antibodies have produced therapeutic anti- a maximum number of effective agents.
bodies with excellent specificity, low immunogenicity The term screening denotes a series of sequential
and optimal pharmacokinetics. These antibodies are assays through which promising agents are tested for
also powerful tools in the isolation and purification of therapeutic potential. For some biotherapies, a general
tumor-associated antigens, lymphokines/cytokines, and screening procedure may be inappropriate. For exam-
other biologicals, which can then be used in biotherapy. ple, the activity of a monoclonal antibody with antitu-
The advances in molecular biology and hybridoma tech- mor specificity would not be detected by use of the
nology have eclipsed previous techniques for the isola- general screen of biological activity. Design consider-
tion and purification of biological molecules [63, 77]. ations for general screening in biotherapy have been
Technical advances in instrumentation, computers, extensively reviewed [33, 54, 56, 61]. A step-by-step
and computer software have been critically important approach to the screening of potential BRM was devel-
in the isolation and purification of biological molecules. oped to define their effects on T-cell, B-cell, NK-cell,
The construction of nucleotide or amino acid sequences and macrophage functions. A progressive in vitro and
to fit any biological message is now possible. While then in vivo sequence allows the variables of dose,
this synthetic capability is currently limited to smaller schedule, route, duration and maintenance of activity,
gene products, techniques by which analysis and con- adjuvanticity, and synergistic potential to be explored
struction of nucleotide sequences will occur in an auto- in an orderly fashion [33]. Unfortunately, the screening
mated way, making enormously complex molecules program was used only briefly by the NCI and no
possible to synthesize and manufacture, are rapidly replacement program is now in use.
becoming available.
Evaluation of Screening Programs
Screening programs for chemotherapeutic agents were
Preclinical Models initiated in the mid-1950s, and attempts have been
made to randomly examine thousands of compounds
Biological Activity in Preclinical Models for antitumor activity [11]. Such large screening pro-
Central to the identification of biotherapy that might be grams are empirically rather than rationally based, and
useful in cancer patients is the recognition that, in the are no longer appropriate [4, 5, 20, 33, 71]. Whether
main, the challenge in humans is the eradication of induced or transplantable animal tumor systems are
metastases. Metastases can result from the dissemina- valid models for testing therapeutic approaches for
tion of different subpopulations of cells within the pri- human cancer has been a controversial issue [33].
mary neoplasm [32]. This may explain the observation In patients, therapy successful against one type of can-
that cells within a metastasis can be antigenically dis- cer may not be successful against another type, or even
tinct from those that predominate in the parental tumor for another patient with the same histological type of
[32]. Metastases may also emanate from other metasta- cancer. Unlike the model systems, in which treatment
ses. The implications of cellular heterogeneity as it can be given with precise timing relative to the meta-
relates to the outcome of the specific immunotherapy static phase of a resected tumor or injected tumor cells,
are obvious. In addition, normal animals are not com- cancer diagnosis in humans is generally late, and micro-
parable to animals or humans bearing autochthonous metastases (and often macro-metastases) have become
Robert K. Oldham 5

well established before treatment can be initiated. Thus, lants of the immune response. Bacillus Calmette-Guerin
screening programs can only provide tentative indica- (BCG) and other whole organisms were used early in
tions on agents and approaches of interest. immunotherapy. The use of a purified derivative of bacte-
rial components, such as muramyl di- or tripeptide, “pack-
aged” in liposomes as a method to stimulate macrophages
Biotherapy: Specific Agents to greater anticancer activity has also been tried. Such
adjuvants may prove useful with genetically engineered or
and Approaches synthetic tumor-associated antigens, active specific
immunotherapy, or immunoprophylaxis.
Nonspecific Immunomodulators Multiple agents that appear capable of augmenting
Since the early 1900s, immunotherapy with bacterial or one or more immune functions already exist. Several of
viral products has been utilized with the hope of “non- these have been associated with prolongation of sur-
specifically” stimulating the host’s immune response vival in prospectively randomized trials involving
[71]. These agents had been useful as adjuvants and as patients with a wide variety of malignancies (Table 2).
nonspecific stimulants in animal tumor models, but Although some of these agents have produced modest
human trials have been disappointing. clinical benefits, and do represent a potential method of
Perhaps purified viruses or specific chemicals will lead immune augmentation, it is doubtful that they will have
to the development of more effective adjuvants or stimu- a major role in future cancer therapy. Great problems

Table 2. Biologicals and biological response modifiers


Immunomodulating agents Lymphokines, cytokines, growth/maturation factors
Alkyl lysophospholipids (ALP) Antigrowth factors
Azimexon Chalones
BCG Colony-stimulating factors (CSF)
Bestatin Growth factors (transforming growth factor, TGF)
Brucella abortus Lymphocyte activation factor (LAF-interleukin 1 [IL-1])
Cornyebacterium parvum Lymphotoxins (TNF, α, β, LT)
Cimetidine Macrophage activation factors (MAF)
Sodium diethyldithiocarbamate (DTC) Macrophage chemotactic factor
Endotoxin Macrophage cytotoxic factor (MCF)
Glucan Macrophage growth factor (MGF)
‘Immune’ RNAs Migration inhibitory factor (MIF)
Krestin Maturation factors
Lentinan Interleukin 3–18, etc.
T-cell growth factors (‘TCGF’ – interleukin 2 [IL-2]) Thymocyte mitogenic factor (TMF)
Levamisole Transfer factor
Muramyldipeptide (MDP), tripeptide (MTP) Transforming growth factor (TFR, α, β)
Maleic anhydride-divinyl ether (MVE-2)
Mixed bacterial vaccines (MBV) Antigens
Nocardia rubra cell wall skeletons (CWS) Tumor-associated antigens
Picibanil (OK432) Molecular vaccines
Prostaglandin inhibitors (aspirin, indomethacin) Cell-engineered cellular vaccines
Thiobendazole Effector cells
Tuftsin Macrophages
Interferons and interferon inducers NK cells
Interferons (α, β, γ) Cytotoxic T cells
Poly IC-LC LAK cells
Poly A-U T helper cells
GE-132 Miscellaneous approaches
Brucella abortus Allogeneic immunization
Tilorone Liposome-encapsulated biologicals
Viruses Bone-marrow transplantation and reconstitution
Pyrimidinones Plasmapheresis and ex vivo treatments (activation
Thymosins columns, immunoabsorbents, and ultrafiltration)
Thymosin alpha-1 Virus infection of cells (oncolysates)
Thymosin fraction 5
Other thymic factors
6 Cancer biotherapy: general principles

exist for most of the agents, including lack of chemical Table 3. Studies of immunotherapy with random designs
definition, low purity, and poor reproducibility from Specific cancer
one lot to another. An additional problem has been the and type of Positive Equivocal Negative
inability to define clearly a mechanism of action for immunotherapy studies studies studies
these agents in humans. The preclinical screening Leukemia
established by the Biological Response Modifiers BCG/AML +
Program (BRMP) of the National Cancer Institute was BCG/Cells/AML + +
one mechanism to do this [31]. Data from this type of MER/AML +
screening with subsequent phase I and phase II clinical CP/Cells/AML +
data could have provided interesting insights and cor- All/Hodgkin’s
relations [98, 104, 105]. Unfortunately, this approach NHL/MM
has been abandoned. Lev/ALL +
The ability to specifically control and manipulate BDG/NHL +
immune responses with highly purified, defined mole- Lev/MM +
cules obtained by genetic engineering is the future. Lung cancer
Thus, it seems probable that nonspecific immunotherapy IT BCG + +
as a sole modality has become obsolete, although as IP BCG + +
adjuvants some may find a role in active specific immu- IP BCG & Lev +
notherapy (Chapter 6). IP CP +
Lev + +
Thy Fr V +
Active Specific Immunotherapy BCG/Cells +
TAA/Freund’s
There has been a substantial effort to produce active Adjuvant +
immunization of autochthonous or syngeneic hosts with
Breast cancer
irradiated or chemically modified tumor cells in an
Poly A/Poly U +
attempt to use active specific immunotherapy (AST) [46]. BCG + +
Inherent is the assumption that tumor cells express immu- Lev +
nogenic tumor-associated antigens (TAA). Treatment of
Colon cancer
tumor cells with a variety of unrelated agents, such as
BCG +
irradiation, mitomycin, lipophilic agents, neurominidase, MER +
viruses, or admixtures of cells with bacterial or chemical CP +
adjuvants, has produced non-tumoroigenic tumor cell Lev +
preparations that are immunogenic upon injection into
Melanoma
syngeneic hosts (Table 3). IL/BCG +
AST using BCG-tumor cell (“antigens”) vaccines has BCG/BCG + Cells + +
been reevaluated using a syngeneic guinea pig hepato- BCG + +
carcinoma. The definition of several variables of vac- CP +
cine preparation, such as a ratio of bacterial organisms Lev +
to viable, metabolically active tumor cells, the proce- Genitourinary cancer
dures of tumor cell dissociation and cryobiologic pres- IC BCG/bladder +
ervation, and the irradiation attenuation of cells, has BCG/prostate +
resulted in the development of an effective non-tumori- Gynecological cancer
genic vaccine. It has proven effective in both micro- and CP/cervix +
macro-metastatic disease [47]. CP/ovary +
The nature of the anatomic alteration in metastatic BCG/ovary +
nodules after AST was explored using a specific Other cancers
monoclonal antibody to assess vascular permeability Lev/H & N +
within these tumor nodules [50, 51]. Immunohistologic CP/H & N +
analysis demonstrated significantly more antibody in BCG/Cells/Sarcoma +
tumors from vaccinated animals than in comparable MER/Neuroblastoma +
tumors from unvaccinated guinea pigs. These data sup-
port the hypothesis that the anatomic characteristics of tecting tumors not only from immunotherapy but from
tumor foci restrict drug and antibody access, thus pro- other forms of treatment as well [45].
Robert K. Oldham 7

The regulation of the blood supply to neoplastic tissue are now undergoing extensive preclinical and clinical
may be unique in comparison to normal tissue. Tumor evaluation (see Chapter 7).
metastatic nodules may have a vascular “barrier,” which
contributes to a limitation in delivery for chemothera-
peutic agents, monoclonal antibodies, and immune
Thymic Factors
effector cells. Such vascular barriers may provide an It has been known for years that thymic extracts have
environment in which some tumor cells survive immunological activity [40, 41]. Thymosin fraction 5 and
blood-borne chemotherapeutic and biologic agents. thymosin alpha-1 have received the most attention in the
Thus, solid tumor nodules may serve as “pharmacologic laboratory and the clinic. Thymosin fraction 5 is an extract
sanctuaries,” allowing even drug-sensitive tumor cells containing a variety of thymic polypeptides, and alpha-1 is
to continue to grow [45–47]. a synthetic polypeptide component present in many thy-
Hanna and co-workers [47] demonstrated that strategi- mic extracts. Thymic preparations have been shown to
cally timed chemotherapy subsequent to immunotherapy enhance and suppress immune responses in both intact
can effectively double the number of survivors attainable and thymectomized animals. Many investigators have
with immunotherapy alone. This effect was not drug spe- reported that the thymosins can correct selected immuno-
cific. These results suggest a new basis for AST in the treat- deficiency states, both natural and experimentally induced.
ment of solid tumors. Inflammatory disruption of anatomic There have also been reports that thymic factors can aug-
barriers of metastatic nodules combined with strategically ment suppressed or depressed T-cell responses in patients
administered chemotherapy or biotherapy may prove use- with cancer. Studies in preclinical screening have demon-
ful in the design of future biotherapy trials in humans. strated stimulation of T-cell activity [99], but clinical stud-
Another approach used more recently involves the deliv- ies have not shown striking effects [25, 98, 99], and none
ery of lymphokines and cytokines, such as interleukins, have been approved for clinical use.
colony stimulating factors, tumor necrosis factor, lympho-
toxins, macrophage cytotoxic factors, and activated com-
plexes (such as those generated by plasma perfusion over
Recombinant DNA Technology
protein A columns) to the tumor and its vascular bed. These Recombinant DNA technology, commonly referred to as
substances are known to have powerful effects on tumor genetic engineering, has provided us with the tool for the
vasculature and cellular infiltrations, something leading to biosynthesis and subsequent mass production of a sig-
tumor necrosis. This approach may, in addition, increase nificant number of biologicals [8, 29, 39]. This is highly
the access of antibody, immunoconjugates, drugs, and acti- relevant to lymphokines/cytokines as well as growth and
vated cells to the cancer nodule [45]. maturation factors, and should revolutionize the treat-
A more recent method to influence the blood supply ment of cancer over the next 10 years. The process
of tumor nodules has been the use of a monoclonal anti- involves the incorporation (recombination) of a segment
body, Bevacizumab, which targets the vascular endothe- of a DNA molecule containing a desired gene into a vec-
lial growth factor (VEGF) receptor. Bevacizumab is tor, usually a plasmid, which in turn is inserted into a
primarily used with chemotherapy, and it may be there host organism, usually an Escherichia coli, although
are similar mechanisms at play with regard to increasing other bacteria, yeasts, insects, and mammalian cells have
chemotherapy drug access to tumor nodules by the use been utilized. The cells are cloned and the cells produc-
of a monoclonal antibody targeting the VEGF receptor ing the desired protein or polypeptide are selected. This
[38, 49, 52]. clone is mass produced using fermentation techniques,
A major limitation of AST has been the availability of and the protein molecule is harvested and purified. The
purified TAA. Whereas whole cell vaccine preparations resultant product is a highly purified (95–99%) protein
contain viable, non-tumorigenic cells prepared from solution, and has a high specific activity (i.e., biological
individual tumors, purified TAA is now prompting large- activity per weight of protein).
scale immunization. TAA purification and characteriza- The relevant DNA can be obtained by a variety of
tion followed by genetic engineering of the antigen for methods. Once messenger RNA (mRNA) is isolated,
vaccine production is underway and several purified complementary DNA (cDNA) can be produced through
antigens are in clinical trials, especially in melanoma. the use of reverse transcriptase. Alternatively, an artificial
Alternatively, synthetic peptide sequences of the active DNA molecule can be constructed once the nucleotide or
portion of TAA may prove useful in the near future. amino acid sequence is known. This can be used to isolate
Even the combining site of antibody to TAA has recently the complementary sequence, which is then isolated and
been suggested as a potential vaccine. These technologies cloned. RNA molecules can also be used in cell-free systems
8 Cancer biotherapy: general principles

Table 4. Clinical lymphokines/cytokines and subsequent cloning in vitro, T-cell lines with spe-
Colony-stimulating factors cific cytotoxic and helper capabilities can be obtained
Erythropoietin and utilized in autologous and allogeneic adoptive
Interferons: α, β, γ immunotherapy [16, 27, 93, 103, 112].
Interleukins 1–32, etc. Many investigators have held the rather simplistic
Lymphotoxincs: α, β (TNF)
view that the immune system (Fig. 1) might be manipu-
Macrophage-activating factors
Thymosins lated in vivo and corrected to better deal with the cancer
Transfer factor(s) problem. Evidence to date suggests that the pharmaco-
logic use of biologicals, in rather high doses, is a more
effective method for cancer treatment, with immunoac-
tivation and immunomodulation, playing the dominant
to produce these biologicals. There are over 200 restric- roles [69, 70].
tion enzymes that can cut desired fragments of DNA and Another specific use of lymphokines may be in the
lead to their isolation. These enzymes can uniquely cleave pharmacological regulation of tumors of the lymphoid sys-
a DNA molecule at specific, predictable sites relative to tem. Although many of these tumors are considered to be
the nucleotide sequence. These fragments are then incor- generally unresponsive to normal growth-controlling
porated into the plasmid, and combine with plasmid mechanisms mediated by lymphokines, it is possible that
DNA. Plasmids have a symbiotic relationship with large quantities of pure lymphokines administered as
selected bacteria inducing resistance to a variety of anti- medicinals, or the use of certain molecular analogs of
biotics, which allows for selection of engineered clones. these naturally occurring lymphokines, may be useful in
A number of alpha interferons, as well as beta and gamma the treatment of lymphoid malignancies.
interferon, have been genetically engineered [24, 39, 106, The use of certain lymphokines/cytokines has been
111]; multiple interleukins, colony-stimulating factors, extended to other cancers, in that in vitro observations
and tumor necrosis factor have been cloned [107]. The suggest an antiproliferative activity in some solid
number of cloned biological products increases yearly tumors. These antiproliferative effects might be maxi-
(see Chapter 4). These biological products and their mized by testing the tumor cells of each patient to “cus-
receptors (Table 4) are rapidly being translated into high- tom tailor” the treatment rather than giving these
quality pharmaceuticals for clinical testing and approval biologicals as general treatment, as has been done with
for general use. anticancer drugs [70].
IL-1, originally known as lymphocyte activating fac-
tor, is a macrophage-derived cytokine that has an
Lymphokines/Cytokines enhancing effect on murine thymocyte proliferation.
Lymphokines and cytokines are molecules secreted by a Both IL-1 and viable macrophages are necessary for the
variety of cells (Table 4). They provide one means initial step in activation of IL-2 (Fig. 2). Cloning of IL-1
through which the cells involved in the immune process and IL-2 has made large quantities of highly purified
communicate with one another and direct the overall materials available for clinical studies.
process [40]. Lymphokines/cytokines with specific Preclinical studies with IL-2 have been oriented around
effects on cell proliferation have been identified and in vitro cell production protocols and induction or mainte-
may prove useful as anticancer agents. Interleukins (IL) nance of antitumor T-cell effects in vivo [15, 16, 67, 70].
1–32 are among the multiple lymphokines that appear to IL-2 has been used to activate peripheral blood cells and
be involved in a cascade phenomenon leading to the initially stimulated much interest [93, 104]. These acti-
induction of a variety of immune responses [86]. Other vated cells are generally more cytotoxic against cancer
examples include multiple subclasses of colony-stimu- cells than normal cells; however, their lineage can be T
lating factors (CSF) chemokines and ligands. The list of cell or NK cell (LAK cells) depending on the technique
lymphokines/cytokines is long, and the potential for employed. This approach, though expensive and techni-
therapeutic manipulation is great [73, 74]. The identifi- cally demanding, illustrates that cellular therapy is feasible
cation of these biological activities is the start of a pro- and can be effective [67, 70, 85, 103].
cess that should ultimately provide us with the knowledge A lymphotoxic product of antigen/mitogen-stimulated
and the tools to identify more accurately and control a leukocytes was called lymphotoxin [92]. Lymphotoxin
number of immune responses. Physicians may then be may be a principle effector of delayed hypersensitivity
able to manipulate the immune system (Fig. 1) intelli- and, although conflicting data have been reported, may
gently, in favor of the host. Further, by selective activation also be involved in the cytoxic reactions of T-cell-mediated
Robert K. Oldham 9

Figure 1. Immune response

lysis and NK- or K-cell lysis. Depending upon the type site [60, 28]. Thus, intratumoral and regional perfusion
of tumor cell involved, the in vitro effect of lymphoto- studies with TNF have yielded positive results in patients
xin may be either cytolytic or cytostatic. Mouse tumor with melanoma and sarcoma [17, 53, 59, 60, 90].
cells are frequently killed by homologous and heterolo- There is now evidence that the combined use of various
gous lymphotoxins, whereas in other species reversible lymphokines may produce enhanced antiproliferative
inhibition of tumor cell proliferation is more common effects. Selective assays for lymphokine antiproliferative
[48]. cocktails may prove useful in “tailoring” such preparations
Human lymphotoxin is of at least two species, alpha for individual patients [6, 95, 97, 102, 100, 110].
and beta [30, 37, 60, 88, 108]. Alpha lymphotoxin is More than 100 biological molecules have already been
tumor necrosis factor (TNF). Both have been cloned, and described and named as lymphokines/cytokines (Table 5).
TNF has undergone rather extensive clinical trials [31, Biologicals such as the interferons, lymphotoxins, TNF,
91, 101]. While some antitumor responses have been CSF, IL-1–22 are now under evaluation (see Chapter 8).
seen, the lymphotoxins have not been highly efficacious The studies require quantities of material obtained
as single agents in the treatment of advanced human through genetic engineering, using sensitive and rapid
tumors and the toxicity of systemic administration has assay procedures to monitor production, purification, and
been unacceptable. Continued trials are underway com- bioavailability.
bining lymphotoxins such as TNF with other lym-
phokine/cytokines and with chemotherapy. Targeted
delivery of these molecules may prove more efficacious
Interferons
since they have high toxicity administered intravenously Interferons are small, biologically active proteins with
with what is probably minimal delivery to the tumor cell antiviral, antiproliferative, and immunomodulatory
10 Cancer biotherapy: general principles

Monocytic Macrophage these functions. Low to moderate doses may inhibit


delayed hypersensitivity while enhancing macrophage
phagocytosis and cytotoxicity, natural killer activity,
and surface antigen expression. Interferons prolong and
inhibit cell division (both transformed and normal cells).
Ag Stimulated
T Cell In addition, interferons stimulate the induction of sev-
eral intracellular enzyme systems with resultant pro-
found effect on macromolecular activities and protein
? CSF synthesis.
We can draw some preliminary conclusions about
interferon therapy for human cancer [9, 36, 43, 44, 57,
Activated Macrophage 69, 96] (Tables 6 and 7). One is that the Cantell, lym-
phoblastoid, and recombinant alpha-interferons are
surprisingly similar, both quantitatively and qualita-
IL-1
tively, in their toxicity, antitumor efficacy, and other
biologic effects. Second, objectively defined antitumor
responses in phase I alpha-interferon trials (mostly
Activated T cells Ag Stimulated T cell involving lymphoma, myeloma, Kaposi’s sarcoma,
melanoma, and renal cancer) were observed in approx-
imately 10% of all patients treated. That level of activ-
ity may not seem impressive, but it does exceed the
IL -2 average response rate of 1–2% in phase I trials of che-
motherapeutic agents. We should also note that very
few responses in patients with tumors of the breast,
Effector T cell
colon, lung, or lower genitourinary system have been
Mature Effector T Cells
Precursor seen with alpha-interferon as a single agent. Overall,
even though interferons have toxicities, they were
Figure 2. Model for interleukin stimulation of T-cell immune more tolerable and less permanent than those observed
responses in early-phase chemotherapy testing.
A third impression, suggested by increased response
rates with higher alpha-interferon doses, is that
interferons may produce their acute antitumor effect by
activities (see Chapter 9). Each interferon has a direct cytostatic action, rather than an indirect immu-
distinctive capabilities in altering a variety of nomodulatory mechanism [12, 23, 57, 66, 69, 72, 98].
immunological and biological responses. As a class, Finally, clinical experience with beta- and gamma-
the interferons appear to have some growth-regulating interferons indicated that both produce response rates
capacity in that antiproliferative effects are measur- and response patterns similar to those obtained with
able with in vitro assays and in animal model systems. alpha-interferon, although beta-interferon is only
The relative efficacy of the mixtures of natural inter- approved for multiple sclerosis and gamma only for
ferons that occur after virus stimulation as compared chronic granulomatous disease. What is clear from the
to the cloned interferons remains to be precisely deter- current preclinical and clinical studies is that the inter-
mined. There are more than 20 interferon molecules ferons have antitumor activity even in bulky, drug-resis-
(and theoretically hundreds of recombinant hybrids tant cancers [102]. Clinical activity has been seen most
there from), and efforts are underway altering individ- reproducibly in several lymphomas and leukemias
ual interferon molecules in specific ways, so the range (Table 6), but responses in many other tumor types with
of biological activities of the interferons as antiviral approval in melanoma and renal cancer [36].
agents, as immunomodulating agents, and as antipro-
liferative agents may be very broad [66, 69].
In addition to antiviral and antiproliferative activity, Growth and Maturation Factors
the interferons have profound effects on the immune Using technology similar to that employed for
system. Low doses enhance antibody formation and lymphokine/cytokines, scientists have cloned and pro-
lymphocyte blastogenesis, while higher doses inhibit duced a variety of growth and maturation factors. The
Robert K. Oldham 11

Table 5. Antigen-nonspecific mediators,a unrestricted by MHC


Helper factors ESP (eosinophil stimulation factor)
LAF (lymphocyte-activating factor, IL-1) LCF (lymphocyte chemotactic factor)
NMF (normal macrophage factor) LTF (lymphocyte trapping factor)
BAF (B-cell-activating factor)
TRF (T-cell-replacing factor) Factors acting on vascular endothelium
MP (mitogenic protein) SRF (skin reactive factor)
TDF (thymus differentiation factor) TPF (thymic permeability factor)
Transferrin LNPF (lymph node permeability factor)
MF (mitogenic [blastogenic] factor) LNAF (lymph node activating factor)
NSF (nonspecific factor) AIPF (anaphylactoid inflammation promoting factor)
TDEF (T-cell-derived enhancing factor) IVPF (increased vascular permeability factor)
TEF (thymus extracted factor) Factors acting on other cells
Complement components Interferons
DSRF (deficient serum-restoring factor) TMIF (tumor cell migration inhibition factor)
Suppressor factors OAF (osteoclast activating factor)
Inhibitor(s) of DNA synthesis Fibroblast chemotactic factor
AIM (antibody-inhibitory material) Pyrogens
IDA (inhibitor of DNA synthesis) FAF (fibroblast activating factor)
LTF (lymphoblastogenesis inhibition factor) Growth-stimulating factors
FIF (feedback inhibition factor) BCGF (B-cell growth factor)
MIFIF (Mif inhibition factor) BCDF (B-cell differentiation factor)
SIRS (soluble immune response suppressor) MGF (macrophage growth factor)
IRF (immunoregulatory gamma-globulin) MF (mitogeni [blastogenic] factor)
Chalones LIAF (lymphocyte-induced angiogenesis factor)
IF (interferons) CSF (colony-stimulating factor)
AFP (alpha-fetoprotein) TDF (thymus differentiation factor)
LDL (low-density lipoproteins) Thymopoietin, thymosin
CRP (C-reactive protein) TCGF (T-cell growth factor, IL-2)
Fibrinogen degradation products IL-3 (interleukin 3)
NIP (normal immunosuppressive protein) EGF (epidermal growth factor)
LMWS (low-molecular-weight suppressor)
HSF (histamine-induced suppressor factor) Direct-acting factors
TCSF (T-cell suppressive factor) Lysosomal enzymes
CTF (cytotoxic factors)
Factors acting on inflammatory cells MTF or MCF (macrophage toxic [cytotoxic] factor)
MIF (migration inhibitory factor) SMC (specific macrophage cytotoxin)
MCF (macrophage chemotactic factor) MCF (macrophage cytolytic factor)
MSF (macrophage slowing factor) ACT (adherent cell toxin)
MEF (migration enhancement factor) Chromosomal breakage factors
MAF (macrophage activating factor) Microbicidal factors
MFF (macrophage fusion factor) LT (lymphotoxin)
PRS (pyrogen-releasing substance) PIF (proliferation inhibitory factor)
LIF (leukocyte inhibition factor) CIF (cloning inhibition factor)
NCF (neutrophil chemotactic factor) IDS (inhibitor of DNA synthesis)
PAR (products of antigenic recognition) Transforming factors
BCF (basophil chemotactic factor) TNF (tumor necrosis factor)
ECF (eosinophil chemotactic factor)
a
These names/factors are based on biological activity. Several may represent the activity of a single molecule.

clinical trials have focused mainly on erythropoietin ration factors (See Chapter 16). It is now clear that a
and the colony-stimulating factors. The former is a drug variety of biological substances up- and down-regulate
now approved for use in refractory anemia, and GM-CSF growth of both normal and neoplastic cells. These sub-
and GCSF are approved for the treatment of bone marrow stances may stimulate or inhibit growth and may change
dysplasia and chemotherapy-induced marrow suppres- the maturation cycle of various normal and neoplastic
sion. Oprelvekim to stimulate platelet production is now cells. Contained within this broad category of factors
approved. These factors are reviewed in later chapters, are the tumor growth factors, colony-stimulating fac-
but it should be noted that they represent only the early tors, and a variety of still to be defined factors important
beginnings of the very broad field of growth and matu- in the regulation of cell growth and maturation. Future
12 Cancer biotherapy: general principles

Table 6. Apha-interferon activitya antigens on the cell surfaces, which will improve our
Active understanding of cell differentiation and of cancer biol-
Hairy-cell leukemia ogy. Major problems in understanding the biology of the
Chronic myelogenous leukemia cancer cell have been the difficulties of isolating, purify-
Myeloproliferative disorders ing, and characterizing tumor-associated antigens (TAA).
Non-Hodgkin’s lymphoma
The use of monoclonal antibody technology will improve
Cutaneous T-cell lymphoma
Kaposi’s sarcoma the definition of the neoplastic cell surface and identify
Multiple myeloma its differences from the normal counterpart. This will be
Melanoma of great value in cancer diagnosis and histopathologic
Renal cell carcinoma classification, and will be useful in the imaging of tumor
Bladder cancer (intravesical)
cell masses and in the therapy of cancer [1, 7, 13, 14, 32,
Inactive 33, 34, 35, 50, 51, 55, 63, 75, 84, 89]. Finally, antibodies
Breast cancer may be a useful reagent in treating certain immune defi-
Colon cancer
Lung cancer ciencies and in altering immune responses. The removal
Prostate cancer of T cells from bone marrow to improve bone marrow
Acute myelogenous leukemia transplantation techniques is an example of using anti-
a
body as a BRM [63]. A more recent example is the use
As a single agent.
of anti-CTLA-4 antibodies to regulate the immune
response. Two monoclonal antibodies are in develop-
Table 7. Malignancies: Summary of responses to alpha- ment that have influence on T-regulatory cells, which
interferon with date of FDA approval influence the immune response to cancer. It is already
Tumor type Response rate (%) Approved clear that this class of monoclonal antibodies can influ-
Multiple myeloma 18–27 ence T-regulatory cells in a yet undefined manner, such
Hairy cell leukemia 80–90 1986 that regression of bulky melanoma can occur. At the
Low-grade lymphoma 38–73 1997 same time, they give a range of interesting toxicities with
High-grade lymphoma 0–10 regard to uveitis, colitis, and depigmentation syndromes
Kaposi’s sarcoma 25–40 1988 relative to their use in melanoma [26].
Chronic myelogenous 80–90 1995 In spite of encouraging data from the use of anti-
leukemia
bodies and, especially, immunoconjugates to target
Melanoma 20–30 1995
Renal cancer 10–20
toxic substances to cancer, the heterogeneity of cancer
is an important consideration [2, 71–78, 87]. If a sin-
gle antibody or a fixed combination of a few antibod-
ies covering only a portion of the tumor cells is used,
therapeutic use of these factors may regulate growth and and if that preparation does not eliminate the true rep-
spread of cancer. Such a chronic growth restraining licating cell population (stem cell) from the patient’s
strategy may not “cure” cancer; rather treatment may be tumor population, eventual outgrowth of viable, per-
more analogous to using insulin in diabetes. This is a haps resistant cells is inevitable. Therefore, it seems
field that is undergoing explosive growth and should be logical to proceed with attempts to type human tumors
watched carefully over the coming decade for molecules and to deliver toxic substances to them utilizing “cock-
with therapeutic activity. tails” of antibodies sufficient to cover all the tumor
cells suspected of replication in each patient. This
type of approach may require a considerable amount
Monoclonal Antibodies of testing for each patient and a “typing” of one or
The advent of hybridoma technology in the late 1970s more tumors from each patient [3, 58, 62, 78, 79, 87].
made available an important tool for the production of Such approaches may be more individually designed
monoclonal antibodies for therapeutic trials [10] (see than is easily approachable through the product devel-
Chapter 10). These antibodies are now being produced in opment paradigm that has been used in the develop-
huge quantities and in highly purified form for cancer ment of new anticancer drugs. If however, the spectrum
treatment. The humanization of murine antibody combin- of human tumor heterogeneity is great, the goal of the
ing sites has yielded a whole new class of therapeutic anti- ideal antibody conjugated to the ideal toxic agent may
bodies [84]. They will undoubtedly define a new range of not be achievable.
Robert K. Oldham 13

Future Perspectives association with, the immune system. The tremendous


diversity of this system is best understood by clinical
How rapidly will biotherapy develop and what role will immunologists and cell biologists who are well suited to
it have in cancer treatment in the next decade? It is cer- assist in the translation of biotherapeutic approaches to
tain that we now have much more powerful tools for the clinic.
improving cancer therapy in the future. We now have Given these new techniques and new approaches, we
the techniques to decipher the major problems in cancer must redesign many of the mechanisms for develop-
biology down to the genetic level. These techniques, mental therapeutics [59]. It may well be that the speci-
along with the recognition that biotherapy can provide ficity of biologicals will require that biotherapy be
increased selectivity in cancer treatment, support the developed in an individualistic fashion and applied to
belief that new and highly effective approaches are near. each patient in a specific, personalized way. This con-
Biotherapy provides an additional technique, which cept was conceptualized in 1977 by the Nobel laureate
may work effectively in combination with surgery or Sir Peter Medawar:
radiotherapy (to decrease the local and regional tumor)
or with chemotherapy (to reduce the systemic tumor The cure of cancer is never going to be found. It is far more likely
burden). It may work very effectively via antibodies in that each tumor in each patient is going to present a unique research
problem for which laboratory workers and clinicians between them
directing radioisotopes selectively to the tumor site, and will have to work out a unique solution.
with chemotherapy, toxins, and other cytostatic or cyto-
toxic molecules in directing the agent to the tumor bed,
enhancing activity and selectivity.
The use of biotherapy is at an early stage. We have References
already seen that highly purified biologicals can be effec- 1. Abrams P, Oldham RK. Monoclonal antibody therapy of solid
tive in patients with clinically apparent, even advanced, tumors. In: Foon KA, Morgan AC, Jr., eds. Monoclonal antibody
bulky cancer. Clinical studies with alpha-interferon have therapy of human cancer. The Hague: Martinus Nijihoff, 1985;
103–120.
demonstrated the responsiveness of radiation- and drug- 2. Abrams PG, Morgan AC, Schroff RW, et al. Monoclonal antibod-
resistant lymphoma, melanoma, and renal carcinoma. ies in cancer therapy: Drug-antibody conjugates. In: Reisfeld RA,
IL-2 with effector cells or alone (in high doses) produces Sell S, eds. Localization and biodistribution studies of a monoclo-
partial and complete remissions in melanoma and kidney nal antibody in patients with melanoma. New York: Alan R. Liss,
1985; 27:233–236.
cancer. These results, along with the clinical results using
3. Avner BP, Liao SK, Avner B, et al. Therapeutic murine monoclo-
monoclonal antibody alone and conjugated to toxic sub- nal antibodies developed for individual cancer patients. J Biol
stances in selected cancers (lymphoma, melanoma, gas- Response Modif 1989; 8(1):25–36.
trointestinal, leukemia and breast cancer), confirm the 4. Baldwin, RW. Relevant animal models for immunotherapy. Cancer
concept that we need not think of biotherapy as a tool that Immunol Immunother 1976; 1:97–206.
5. Balkwill FR, Burke F. The cytokine network. Immunol Today
can be used only in patients with undetectable and mini-
1989; 10(9):29–304.
mal residual tumor burdens [84]. While this modality 6. Balkwill FR, Lee A, Aldam G, et al. Human tumor xenografts
may work best with minimal tumor burdens (a situation treated with recombinant human tumor necrosis factor alone or in
that is also true for chemotherapy), biotherapy can be combination with interferons. Cancer Res 1986; 46:3990–3993.
used as a single modality in clinically apparent disease. It 7. Bernhard MI, Foon KA, Oeltmann TN, et al. Guinea pig line 10
hepatocarcinoma model: characterization of monoclonal antibody
may be even more effective in multimodality treatment and in vivo effect of unconjugated antibody and antibody conjugated
regimens. Biotherapy offers the hope for selective treat- to diphtheria toxin A chain. Cancer Res 1983; 43:4420–4428.
ment to enhance the therapeutic/toxic ratio and lessen the 8. Bollon AP, Barron EA, Berent SL, et al. Recombinant DNA tech-
problem of nonspecific toxicity, a major impediment to niques: Isolation, cloning and expression of genes. In: Bollon AP,
the development of more effective cancer treatment. ed. Recombinant DNA products: Insulin interferon and growth
hormone. Boca Raton, Florida: CRC Press, 1984; 1–35.
This decade will provide many opportunities to 9. Bonnem EM, Spregel RJ. Interferon-alpha: Current status and
pursue new approaches in cancer treatment. These future promise. J Biol Response Modif 1984; 3:580.
approaches will employ new techniques in the laboratory 10. Boss BD, Langman R, Trowbridge I, Dulbecco R, eds. Monoclonal
and clinic, requiring special training and expertise. The antibodies and cancer. New York: Academic Press, 1983; 1–200.
11. Boyd MR. Status of the NCI preclinical antitumor drug discovery
medical oncologist trained in the administration of che-
screen. Prin Pract Oncol Suppl 1989; 3(10):1–12.
motherapy drugs, is not necessarily well prepared to 12. Bunn PA, Foon KA, Ihde DC, et al. Recombinant leukocyte A
give biologicals for cancer treatment. Biotherapy uses interferon: An active agent in advanced cutaneous T-cell lympho-
biological substances that are often active on, or work in mas. Ann Intern Med 1984; 101:484–487.
14 Cancer biotherapy: general principles

13. Carrasquillo JA, Bunn PA, Jr., Kennan AM, et al. Radioimmunodetection 33. Fidler IJ, Berendt M, Oldham RK. The rationale for and design of
of cutaneous T-cell lymphoma with 111In-T101 monoclonal antibody. screening assays for the assessment of biological response modi-
New Engl J Med 1986; 315:673–680. fiers for cancer treatment. J Biol Response Modif 1982; 1:15–26.
14. Carrasquillo JA, Abrams PG, Schroff R, et al. Effect of antibody 34. Foon KA, Bernhard MI, Oldham RK. Monoclonal antibody ther-
dose on the imaging and biodistribution of indium-111 9.2.27 anti- apy: Assessment by animal tumor models. J Biol Response Modif
melanoma monoclonal antibody. J Nucl Med 1988; January, 1982; 1:277–304.
29(1):39–47. 35. Foon KA, Schroff R, Bunn PA, et al. Effects of monoclonal anti-
15. Cheever MA, Greenberg PD, Fefer A, Gillis S. Augmentation of body therapy in patients with chronic lymphocytic leukemia.
the antitumor therapeutic efficacy of long-term cultured T lympho- Blood 1984; 64:1085–1093.
cytes by in vivo administration of purified interleukin-2. J Exp 36. Foon KA, Sherwin SA, Abrams PG, et al. Treatment of advanced
Med 1982; 155:968–980. non-Hodgkin’s lymphoma with recombinant leukocyte A inter-
16. Cheever MA, Greenberg PD, Fefer A. Potential for specific cancer feron. N Engl J Med 1984; 311:1148–1152.
therapy with immune T lymphocytes. J Biol Response Modif 37. Gamm H, Lindemann A, Mertelsmann R, Herrmann F. Phase I
1984; 3:113–127. trial of recombinant human tumour necrosis factor α in patients
17. Cumberlin R, DeMoss E, Lassus M, Friedman M. Isolation perfu- with advanced malignancy. Eur J Cancer 1991; 27:856–863.
sion for malignant melanoma of the extremity: A review. J Clin 38. Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in
Oncol 1985; 3:1022–1031. combination with oxaliplatin, fluorouracil and leucovorin
18. DeVita VT. Progress in cancer management: Keynote address. (FLOFOX4) for previously treated metastatic colorectal cancer:
Cancer 1983; 51:2401–2409. Results from the Eastern Cooperative Oncology Group Study
19. DeVita VT. The relationship between tumor mass and resistance to E3200. J Clin Oncol 2007; 25:1539–1544.
chemotherapy: Implication for surgical adjuvant treatment of can- 39. Goeddel DV, Yelverton E, Ullrich A, et al. Human leucocyte inter-
cer. Cancer 1983; 51:1209–1220. feron produce by E. coli is biologically active. Nature 1980;
20. DeVita VT, Hellman S, Rosenberg SA, eds. Important advances in 287:411–416.
oncology 1989. New York: J.B. Lippincott, 1995; 1–118. 40. Goldstein AL, Chirigos MA, eds. Lymphokines and thymic hor-
21. DeVita VT, Jr., Hellman S, Rosenberg SA, eds. In: Biologic ther- mones; their potential utilization in cancer therapeutics. Progress in
apy of cancer, second edition. Philadelphia, Pennsylvania: J.B. cancer research and therapy, vol. 20. New York: Raven Press, 1981.
Lippincott, 1995; 295–327. 41. Goldstein AL, Chirigos MA. In: Progress in cancer research and
22. DeVita VT, Jr., Hellman S, Rosenberg SA, eds. In: Biologic ther- therapy, vol. 20. New York: Raven Press, 1982; 1–324.
apy of cancer, second edition. Philadelphia, Pennsylvania: J.B. 42. Gray PSW, Leung DW, Pennica D, et al. Expression of human
Lippincott, 1995; 329–345. immune interferon cDNA in E. coli and monkey cells. Nature
23. DeVita VT, Jr., Hellman S, Rosenberg SA, eds. In: Biologic ther- 1982; 295:503–508.
apy of cancer, second edition. Philadelphia, Pennsylvania: J.B. 43. Grem JL, Hoth D, Hamilton MJ, et al. An overview of the current
Lippincott, 1995; 427–433. status and future direction of clinical trials of 5-fluorouracil and
24. Devos R, Cheroutre H, Taya Y, et al. Molecular cloning of human folinic acid. Cancer Treat Rep 1987; 71:1249–1264.
immune interferon cDNA and its expression in eukaryotic cells. 44. Grem JL, McAtee N, Murphy RF, et al. A pilot study of interferon
Nucl Acids Res 1982; 10:2487–2501. alfa-2a in combination with fluorouracil plus high-dose leucovorin
25. Dillman RO, Beauregard JC, Mendelsohn J, et al. Phase I trials of in metastatic gastrointestinal carcinoma. J Clin Oncol 1991;
thymosin fraction 5 and thymosin alpha-1. J Biol Response Modif 9:1811–1820.
1982; 1:35–41. 45. Hanna MG, Key ME, Oldham RK. Biology of cancer therapy:
26. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer Some new insights into adjuvant treatment of metastatic solid
therapy following non-ablative but lymphodepleting chemother- tumors. J Biol Response Modif 1983; 4:295–309.
apy for the treatment of patients with refractory metastatic mela- 46. Hanna MG, Jr., Brandhorst JS, Peters LC. Active specific immuno-
noma. J Clin Onc 2005; 23:2346–2357. therapy of residual micrometastasis: An evaluation of sources,
27. Eberlein TJ, Rosenstein M, Spiess P, et al. Adoptive chemoimmu- doses and ratios of BCG with tumor cells. Cancer Immunol
notherapy of a synergeneic murine lymphoma with long-term lym- Immunother 1979; 7:165–174.
phoid cell lines expanded in T-cell growth factor. Cancer Immunol 47. Hanna MG, Jr., Key ME. Immunotherapy of metastases enhances
Immunother 1982; 13:5–13. subsequent chemotherapy. Science 1982; 217:367–370.
28. Eggermont AM, Schraffordt Koops H, Klausner JM, et al. 48. Haranaka K. Macrophage Symposium 1987, Tumor Necrosis
Isolated limb perfusion with tumor necrosis factor and for limb Factor. Japan, January 5–8, 1987.
salvage in 186 patients with locally advanced extremity sarco- 49. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus
mas. The cumulative multicenter Euro experience. Ann Surg irinotecan, fluorouracil and leucovorin for metastatic colorectal
Dec, 1996; v.224(6). cancer. N Engl J Med 2004; 350:2335–2342.
29. Emery AEH. Recombinant DNA technology. Lancet 1981; 50. Hwang KM, Foon KA, Cheung PH, et al. Selective antitumor
2:1406–1409. effect on L-10 hepatocarcinoma cella of a potent immunoconjugate
30. Evans CH, Lymphotoxin: An immunologic hormone with anticar- composed of the A chain of abrin and a monoclonal antibody to a
cinogenic and antitumor activity. Cancer Immunol Immunother hepatoma-associated antigen. Cancer res 1984; 44:4578–4586.
1982; 12:181–190. 51. Hwang KM, Keenan AM, Frincke J, et al. Dynamic interaction of
31. Feinberg B, Kurzrock R, Talpaz M, et al. A phase I trial of intrave- 111 indium-labeled monoclonal antibodies with surface of solid
nously-administered recombinant tumor necrosis factor-alpha in tumors visualized in vivo by external scintigraphy. J Natl Cancer
cancer patients. J Clin Oncol 1988; 6:1328–1334. Inst 1986; 76:849–855.
32. Fidler IJ, Gersten DM, Hart IR. The biology of cancer invasion and 52. Kavvinaran FF, Hambleton J, Mass RD, et al. Combined analysis
metastasis. Adv Cancer Res 1978; 28:149–250. of efficacy: The addition of bevacizumab to fluorouracil/leucovorin
Robert K. Oldham 15

improves survival for patients with metastatic colorectal cancer. 73. Oldham RK, Smalley RV. Biological and biological response
J Clin Oncol 2005; 23:3706–3712. modifiers. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer:
53. Kahn JO, Kaplan LD, Volberding PA, et al. Intralesional recombi- Principles and practice of oncology. Philadelphia, Pennsylvania:
nant tumor necrosis factor-α for AIDS-associated Kaposi’s sar- J.B. Lippincott, 1985; 2223–2245.
coma. A randomized, double-blind trial. J Acquir Immune Defic 74. Oldham RK, Thurman GB, Talmadge JE, et al. Lymphokines,
monoclonal antibodies and other biological response modifiers in
Syndr 1989; 2:217–223.
the treatment of cancer. Cancer 1984; 54:2795–2810.
54. Kallman RF, ed. Rodent tumor models in experimental cancer 75. Oldham RK, Foon KA, Morgan AC, et al. Monoclonal antibody
therapy. New York: Pergamon Press, 1987; 1–310. therapy of malignant melanoma: In vivo localization in cutaneous
55. Key ME, Bernhard MI, Hoyer LC, et al. Guinea pig 10 hepatocar- metastasis after intravenous administration. J Clin Oncol 1984;
cinoma model for monoclonal antibody serotherapy: In vivo local- 2:1235–1242.
ization of a monoclonal antibody in normal and malignant tissues. 76. Oldham RK. Perspectives on the use of immunotoxins in clinical
J Immunol 1983; 139:1451–1457. medicine. In: Vogel C-W, ed. Immunoconjugates: Antibody con-
56. Key ME, Brandhorst JS, Hanna MC, Jr. Synergistic effects of jugates in radioimaging and therapy of cancer. New York, Oxford
active specific immunotherapy and chemotherapy in guinea pigs University Press. 1987; 281–289.
with disseminated cancer. J Immunol 1983; 130:2987–2992. 77. Oldham RK. Therapeutic monoclonal antibodies: Effects of
tumor cell heterogeneity. In: Therapeutic monoclonal antibodies:
57. Kirkwood JM, Ernstoff MS. Interferon in the treatment of human
Effects of tumor cell heterogeneity. S. Karger Ag. Cancer
cancer. J Clin Oncol 1984; 2:336–352. Treatment Symposium (Germany), 1988.
58. Liao SK, Meranda C, Avner BP, et al. Immunohistochemical phe- 78. Oldham RK, Lewis M, Orr DW, et al. Adriamycin custom-tailored
notyping of human solid tumors with monoclonal antibodies in immunoconjugates in the treatment of human malignancies. Mol
devising biotherapeutic strategies. Cancer Immunol Immunother Biother 1988; 1(2):103–113.
1989; 28:77–86. 79. Oldham RK, Lewis M, Orr DW, et al. Individually specified drug
59. Lienard D, Ewalenko P, Delmitti JJ, et al. High-dose recombinant immunoconjugates in cancer treatment. In: Ceriani RL, ed. Breast
tumor necrosis factor alpha in combination with interferon gamma cancer immunodiagnosis and immunotherapy. New York: Plenum
and melphalan in isolation perfusion of the limbs for melanoma and Publishing 1990; 219–230.
sarcoma. J Clin Oncol 1992; 10:52–60. 80. Oldham RK. Gene therapy and cancer: Is it for everyone? Cancer
Investigation 1992; 10(6):607–609.
60. Mavligit GM, Zukiwski AA, Charnsargavej C. Regional biologic
81. Oldham RK. Cancer biotherapy: 1993 to the millennium and
therapy: Hepatic arterial infusion of recombinant human tumor necro-
more! Cancer Biother 1993; 8(1):1–2.
sis factor in patients with liver metastases. Cancer 1992; 69:557–561.
82. Oldham RK. Cancer biotherapy: The first year. Cancer Biother
61. McCormick DL, Adamowski CB, Fiks A, Moon RC. Lifetime 1994; 9(3):179–181.
dose-response relationships for mammary tumor induction by a 83. Oldham RK, Lewko W, Good R, et al. Growth of tumor derived
single administration of N-methyl-N-nitrosourea. Cancer Res activated T-cells for the treatment of cancer. Cancer Biother
1981; 41:1690–1694. 1994; 9(3):211–224.
62. Ogden JR, Leung K, Kundra SA, et al. Immunoconjugates of dox- 84. Oldham RK, Dillman RO. JCO 2008; 26(11):1774–1777.
orubicin and murine antihuman breast carcinoma monoclonal anti- 85. Lewko WM, Hall PB, Oldham RK. Growth of tumor-dervice
bodies prepared via an n-hydroxysuccinimide active ester activated T cells for the treatment of advanced cancer. Can Bio
intermediate of cis-aconityl-doxorubicin: Preparation and in vitro and Radio 2000; 15(4):357–366.
cytotoxicity. Mol Biother 1989; 1(3):170–174. 86. Oppenheim JJ, Stadler BM, Siraganian RP, et al. Lymphokines:
63. Oldham, RK. Monoclonal antibodies in cancer therapy. J Clin Their role in lymphocyte responses properties of interleukin 1.
Fed Proc 1982; 41:257–262.
Oncol 1983; 1:582–590.
87. Orr DW, Oldham RK, Lewis M, et al. Phase I trial of mitomycin-c
64. Oldham RK. Biologicals: New horizons in pharmaceutical devel- immunoconjugate cocktails in human malignancies. Mol Biother
opment. J Biol Response Modif 1983; 2:199–206. 1989; 1(4):229–240.
65. Oldham RK. Biologicals and biological response modifiers: New 88. Palladino MA, Shalaby MR, Kramer SM, et al. Characterization
strategies for clinical trials. In: Finter NB, Oldham RK, eds. of the antitumor activities of human tumor necrosis factor-alpha,
Interferons IV. Amsterdam: Elsevier Science, 1985; 235–249. and the comparison with other cytokines: Induction of tumor
66. Oldham RK. Interferon: A model for future biological. In: Burke specific immunity. J Immunol 1987; 138:4023–4032.
D, Cantell K, Gresser I, DeMaeyer E, Landy M, Revel M, Vilcek 89. Paranasivam G, Pearson JW, Bohn W, et al. Immunotoxins of a
J, eds. Interferon VI. New York: Academic Press, 1985; 127–143. human melanoma-associated antigen: Comparison of gelonin
67. Oldham RK. In vivo effects of interleukin 2. J Biol Response with ricin and other a-chain conjugates. Cancer Res 1987;
47:3169–3173.
Modif 1984; 3:455–532.
90. Pfreundschuh MG, Steinmetz HT, Tuschen R, et al. Phase I study of
68. Oldham RK. Biologicals and biological response modifiers: The intratumoral application of recombinant human tumor necrosis
fourth modality of cancer treatment. Cancer Treatment Rep 1984; factor. Eur J Cancer Clin Oncol 1989; 25:379–388.
68:221–232. 91. Regenass U, Muller M, Curschellas E, Matter A. Anti-tumor
69. Oldham RK. Biologicals for cancer treatment: Interferons. effects of tumor necrosis factor in combination with chemothera-
Hospital Pract 1985; 20:72–91. peutic agents. Int J Cancer 1987; 39:266–273.
70. Oldham RK. Biotherapy: The fourth modality of cancer treatment. 92. Rosenau W. Lymphotoxin: Properties, role and mode of action.
J Cell Physiol Suppl 1986; 4:91–99. Int J Immunopharm 1981; 3:1–8.
71. Oldham RK, Smalley RV. Immunotherapy: The old and the new. J 93. Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the
Biol Response Modif 1983; 2:1–37. systemic administration of autologous lymphokine-activated killer
72. Oldham RK, Smalley RV. The role of interferon in the treatment of cells and recombinant interleukin-2 to patients with metastatic
cancer. In: Zoon KC, Noguchi PC, Lui T-Y, eds. Interferon: cancer. N Engl J Med 1985; 313:1485–1492.
Research, clinical application and regulatory consideration. 94. Rosenberg SA. Principles and practices of the biologic therapy of
Amsterdam: Elsevier Science, 1984; 191–205. cancer. Philadelphia, Pennsylvania: J.B. Lippincott, 2001.
16 Cancer biotherapy: general principles

95. Schiller JH, Witt PL, Storer B, et al. Clinical and biologic effects 103. Stevenson HC, eds. Adoptive cellular immunotherapy of cancer.
of combination therapy with gamma-interferon and tumor necrosis New York: Marcel Dekker, 1989; 1–236.
factor. Cancer 1992; 69:562–571. 104. Stringfellow DA, Smalley RV. Interferon inducers for clinical
96. Sherwin SA, Knost JA, Fein S, et al. A multiple dose Phase I trial use. In: Finter N, Oldham RK, eds. In vivo application of interferons.
of recombinant leukocyte A interferon in cancer patients. J Am Amsterdam: Elsevier Science, 1984.
Med Assoc 1982; 248:2461–2466. 105. Talmadge JE, Maluish AE, Collins M, et al. Immunomodulation
97. Smalley RV, Long CW, Sherwin SA, Oldham RK. Biological and antitumor effects of MVE-2 in mice. J Biol Response Modif
response modifiers: Current status and prospects as anticancer 1984; 3:634–652.
agents. In: Herberman RB, ed. Basic and clinical tumor immu- 106. Taniguchi T, Mantei N, Schwarzstein M, et al. Human leukocyte
nology. The Hague: Martinus Hijhoff, 1983; 257–300. and fibroblast interferons are structurally related. Nature 1980;
98. Smalley RV, Oldham RK. Biological response modifiers: 285:2848–2852.
Preclinical evaluation and clinical activity. CRC Crit Rev Oncol/ 107. Tanguichi T, Matsui H, Fujita T, et al. Structure and expression of
Hematol 1984; 1:259–280. cloned cDNA for human interleukin-2. Nature 1983; 302: 305–310.
99. Smalley RV, Talmadge JA, Oldham RK, Thurman GB. The thy- 108. Taguchi T. Phase I study of recombinant human tumor necrosis
mosins: Preclinical and clinical studies with fraction V and factor (rHu-TNF:PT-050). Cancer Detect Prev 1988; 12:561–572.
alpha-1. Cancer Treatment Rev 1984; 11:69–84. 109. Terry MD, Rosenberg SA, eds. Immunotherapy of cancer. New
100. Smith JW, Urba WJ, Clark JW, et al. Phase I evaluation of York: Excerpta Medica, 1982; 1–398.
recombinant tumor necrosis factor given in combination with 110. Wadler S, Lembersky B, Atkins M, et al. Phase II trial of fluo-
recombinant interferon-gamma. J Immunother 1991; rouracil and recombinant interferon alfa-2a in patients with
10:355–362. colorectal carcinoma: An Eastern Cooperative Oncology Group
101. Spriggs DR, Sherman ML, Michie H, et al. Recombinant human study. J Clin Oncol 1991; 9:1806–1810.
tumor necrosis factor administered as a 24-hour intravenous infu- 111. Weissman C, Nagata S, Boll W, et al. Structure and expression of
sion. A Phase I and Pharmacologic Study. J Natl Cancer Inst human alpha interferon genes. In: Miwa M, et al., eds. Primary
1988; 80:1039–1044. and tertiary structure of nucleic acids and cancer research. Tokyo:
102. Stevenson HC, Ochs JJ, Halverson L, et al. Recombinant alpha Japan Science Society Press, 1982; 1–22.
interferon in retreatment of two patients with pulmonary lym- 112. West WH, Tauer KW, Yannelli JR, et al. Constant infusion
phoma, dramatic responses with resolution of pulmonary compli- recombinant interleukin-2 in adoptive immunotherapy of
cations. Am J Med 1984; 77:355–358. advanced cancer. N Engl J Med 1987; 316(15):898–905.
2 The pathogenesis of cancer metastasis:
relevance to therapy
SUN-JIN KIM, CHERYL HUNT BAKER, YASUHIKO KITADAI, TORU NAKAMURA,
TOSHIO KUWAI, TAKAMITSU SASAKI, ROBERT LANGLEY, AND ISAIAH J. FIDLER

Introduction design of more effective therapeutic modalities. In this


chapter, we discuss the biology of cancer metastasis
Metastasis, the spread of malignant tumor cells from a with special emphasis on recent data demonstrating that
primary neoplasm to distant parts of the body where the microenvironment of different organs can influence
they multiply to form new growths, is a major cause of the biological behavior of tumor cells at different steps
death from cancer. The treatment of cancer poses a major of the metastatic process and the development of bio-
problem to clinical oncologists, because by the time logic diversity in malignant neoplasms. These findings
many cancers are diagnosed, metastasis may already have have obvious implications for the therapy of neoplasms
occurred, and the presence of multiple metastases makes in general and metastases in particular.
complete eradication by surgery, radiation, drugs, or
biotherapy nearly impossible. Metastases can be located
in different organs and in different locations within
the same organ. These aspects significantly influence the The Pathogenesis of Cancer
response of tumor cells to therapy and the efficiency of
anticancer drugs, which must be delivered to tumor lesions
Metastasis
to destroy cells without leading to undesirable side effects. The process of cancer metastasis is dynamic, complex
Similarly, immune effector cells of current biotherapeu- and consists of multiple, sequential, and interrelated
tic regimens may not reach or localize in metastases with steps shown schematically in Fig. 1. To produce a clini-
different organs. cally relevant lesion, metastatic cells must survive all
Exacerbating the problems of treating metastatic dis- the steps of the process. If the disseminating tumor
ease is the fact that tumor cells in different metastases cell fails to complete any one of these steps, it will fail
and in some instances even in different regions within a to produce a metastasis. Thus, the successful metastatic
single metastasis may respond differently to treatment. cell has been likened to a decathlon champion who must
Tumor cell resistance to current therapeutic modalities be proficient in all ten events to be successful, not just a
is the single most important reason for the lack success few [58]. The outcome of this process depends on both
in treating many types of solid neoplasms. In part, the the intrinsic properties of the tumor cells and their inter-
emergence of treatment-resistant tumor cells is due to actions with host factors [48, 56, 57, 99].
the heterogeneous nature of malignant neoplasms. This The essential steps in the formation of a metastasis
diversity, which permits selected variants to develop are: (1) After the initial transforming event, either uni-
from the parent tumor, implies not only that the primary cellular or multicellular, growth of neoplastic cells must
tumor and metastases can differ in their response to be progressive, with nutrients for the expanding tumor
treatment, but also that individual metastases can differ mass initially supplied by simple diffusion. (2) Extensive
markedly from one another. vascularization must occur if a tumor mass is to exceed
Insight into the molecular mechanisms that regulate 1–2 mm in diameter. The synthesis and secretion of
the pathogenesis of cancer metastasis as well as a better proangiogenic angiogenesis factor probably plays a key
understanding of the interaction between metastatic role in establishing a neocapillary network from the
cells and the host microenvironment should provide a surrounding host tissue. (3) Local invasion of the host
foundation for the design of new therapeutic approaches. stroma by some tumor cells could occur by several
Moreover, the development of adequate models that nonmutually exclusive mechanisms. (4) Thin-walled
allow for the isolation and characterization of cells venules, like lymphatic channels, offer very little resis-
possessing metastatic potential will be invaluable in the tance to penetration by tumor cells and provide the most

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 17


© Springer Science + Business Media B.V. 2009
18 The pathogenesis of cancer metastasis: relevance to therapy

THE PATHOGENESIS OF A METASTASIS


TRANSFORMATION ANGIOGENESIS MOTILITY & INVASION

Capillaries,
ARREST IN Venules, lymphatic vessels
CAPILLARY
BEDS EMBOLISM &
CIRCULATION

TRANSPORT

ADHERENCE
MULTICELL AGGREGATES
(lymphocytes,platelets)

EXTRAVASATION
INTO ORGAN RESPONSE TO
PARENCHYMA MICROENVIRONMENT

METASTASIS OF
METASTASES
TUMOR CELL
PROLIFERATION
& ANGIOGENESIS
METASTASES

Figure 1. The pathogenesis of cancer metastasis

common pathways for tumor cell entry into the circula- growing, the micrometastases must develop a vascular
tion. Although clinical observations have suggested that network and continue to evade the host immune system.
carcinomas frequently metastasize and grow via the Moreover, the cells can invade, penetrate blood vessels,
lymphatic system, whereas malignant tumors of mesen- and enter the circulation to produce additional metasta-
chymal origin more often spread by the hematogenous ses (Fig. 1).
route, the presence of numerous venolymphatic anasto-
moses invalidates this concept. (5) Detachment and embo-
lization of small tumor cell aggregates occurs next, the Neovascularization-
vast majority of circulating tumor cells being rapidly
destroyed. (6) Once the tumor cells have survived the Angiogenesis
circulation, they must (7) arrest in the capillary beds of Oxygen can diffuse from capillaries for only 150–200 μm.
organs, either by adhering to capillary endothelial cells When distances of cells from a blood supply exceed
or by adhering to subendothelial basement membrane, this, cell death follows [80]. Thus, the expansion of tumor
which may be exposed. (8) Extravasation occurs next, masses beyond 1 mm in diameter depends on neovascu-
probably by the same mechanisms that influence initial larization, i.e., angiogenesis [78, 79]. The formation of
invasion. (9) Proliferation within the organ parenchyma new vasculature consists of multiple, interdependent
completes the metastatic process (10). To continue steps. It begins with local degradation of the basement
Sun-Jin Kim et al. 19

membrane surrounding capillaries, followed by invasion and intravenously (to produce lung metastasis) of athy-
of the surrounding stroma and migration of endothelial mic nude mice. Subcutaneous tumors, lung lesions, and
cells in the direction of the angiogenic stimulus. Proli- liver lesions expressed high, intermediate, and no IL-8,
feration of endothelial cells occurs at the leading edge respectively, at both the mRNA and protein levels.
of the migrating column, and the endothelial cells begin Melanoma cells established from the tumors growing
to organize into three-dimensional structures to form in vivo exhibited similar levels of IL-8 mRNA transcripts
new capillary tubes. Differences in cellular composi- as continuously cultured cells, thus demonstrating that
tion, vascular permeability, blood vessel stability, and the differential expression of IL-8 was not due to the
growth regulation distinguish vessels in neoplasms from selection of a subpopulation of cells [97].
those in normal tissue [1, 3, 79, 156]. IL-8 expression can be upregulated by coculturing
The onset of angiogenesis involves a change in the local melanoma cells with keratinocytes (skin) and inhibited
equilibrium between proangiogenic and antiangiogenic by coculturing melanoma cells with hepatocytes (liver).
molecules [55]. Some of the common proangiogenic We also investigated the effects of two cytokines pro-
factors include bFGF, which induces the proliferation of duced by keratinocytes (IL-1, IFN-β) and two cytokines
a variety of cells and has also been shown to stimulate produced by hepatocytes (TGF-α, TGF-β) on the regu-
endothelial cells to migrate, to increase production of lation of IL-8 in human melanoma cells. IL-1 upregu-
proteases, and to undergo morphogenesis [80]. Likewise, lated the expression of IL-8 in human melanoma cells at
VEGF/VPF has been shown to induce the proliferation both the mRNA and protein levels in a dose- and time-
of endothelial cells, to increase vascular permeability, dependent manner in the presence of de novo protein
and to induce production of urokinase plasminogen acti- synthesis. IFN-β did not affect constitutive IL-8 mRNA
vator by endothelial cells [46]. Additional proangiogenic and protein production in human melanoma cells, but it
factors include IL-8 [268], platelet-derived endothelial did block the induction of IL-8 by IL-1. TGF-β inhib-
cell growth factor, which has been shown to stimulate ited the expression of IL-8, while TGF-α had no effect
endothelial cell DNA synthesis and to induce production on IL-8 expression [226].
of FGF, hepatocyte growth factor (HGF), or scatter fac- Patients with renal cell carcinoma exhibit high levels of
tor, that increases endothelial cell migration, invasion, bFGF in the serum or the urine that inversely correlates
and the production of proteases, and platelet-derived with survival [181, 184]. Human renal cancer cells
growth factor (PDGF). The production of bFGF, VEGF, implanted into the kidney of nude mice were highly meta-
and IL-8 by tumor or host cells or the release of proan- static to the lung, whereas renal cancer cells implanted
giogenic molecules from the extracellular matrix is subcutaneously remain local [227]. The subcutaneous or
known to induce the growth of endothelial cells and intramuscular tumors expressed a lower level of mRNA
formation of blood vessels. Further, the organ microen- transcripts for bFGF than did tumor cells growing in cul-
vironment can directly contribute to the induction and ture, whereas the renal tumors had 20 times higher levels
maintenance of the proangiogenic factors bFGF [226, of bFGF mRNA and protein levels as compared with the
227] and IL-8 [228]. cultured cells. A histopathologic examination of the tumors
The production of angiogenic molecules, e.g., VEGF, demonstrated that the subcutaneous tumors had few blood
bFGF, and IL-8 by melanoma cells is regulated by com- vessels, whereas the renal tumors had many [227, 228].
plex interactions with keratinocytes in the skin [106]. Direct correlation between the level of bFGF and
Reports from our laboratory showed that IL-8 is an advanced disease was also reported for patients diagnosed
important molecule in melanoma growth and progres- with colon carcinoma. Patients with Duke’s C or D tumors
sion. Constitutive expression of IL-8 directly correlated had markedly higher levels of bFGF in the blood than
with the metastatic potential of human melanoma cells. patients with Duke’s B. In situ hybridization analysis
Further, IL-8 induced proliferation, migration, and inva- revealed that bFGF was overexpressed at the tumor
sion of endothelial cells and, hence, neovascularization periphery associated with cell division. Northern blot
[97]. Several organ-derived cytokines (produced by infla- analysis detected no mRNA transcripts for bFGF [129]. In
mmatory cells) are known to induce expression of IL-8 a follow-up study of patients with colon cancer, bFGF
in normal and transformed cells [106]. Since IL-8 expres- expression was found to be highest in the primary tumors
sion in melanocytes and melanoma cells can be induced of patients who presented with metastatic disease and
by inflammatory signals, the question of whether specific therefore identify a cohort of patients who appeared to be
organ microenvironments could influence the expression free of metastatic disease at the time of surgery (low bFGF
of IL-8 was analyzed. Melanoma cells were implanted expression) as compared to another cohort of patients who
into the subcutis, the spleen (to produce liver metastasis), did develop metastatic disease (high bFGF) [130].
20 The pathogenesis of cancer metastasis: relevance to therapy

Tumor Cell Invasion metalloproteinases with apparent molecular masses of 98,


92, 80, 68, and 64-kDa have been detected in highly meta-
To reach blood vessels or lymphatics, tumor cells must static cells. Poorly metastatic cells, on the other hand,
penetrate host stroma that includes basement membrane. appear to secrete very low amounts of only the 92-kDa
The interaction with the basement membrane consists of metalloproteinase [171, 172].
attachment, matrix dissolution, motility and penetration
[154]. At least three nonmutually excluding mechanisms
can be involved in tumor cell invasion of tissues. First,
mechanical pressure produced by rapidly proliferating
Lymphatic Metastasis
neoplasms may force cords of tumor cells along tissue Early clinical observations led to the impression that
planes of least resistance [154, 155]. Second, increased carcinomas spread mainly by the lymphatic route and
cell motility can contribute to tumor cell invasion. Most mesenchymal tumors spread mainly by means of the
tumor cells possess the necessary cytoplasmic machinery bloodstream. We now know, however, that the lymphatic
for active locomotion and increased tumor cell motility and vascular systems have numerous connections and
is preceded by a loss of cell-to-cell cohesive forces. that disseminating tumor cells may pass from one
In epithelial cells, the loss of cell-to-cell contact is associated system to the other [27]. For these reasons, the division
with downregulation of the expression of E-cadherin, a of metastasis into lymphatic spread and hematogenous
cell surface glycoprotein involved in calcium-dependent spread is arbitrary. During invasion, tumor cells can
homotypic cell-to-cell cohesion. Reduced levels of easily penetrate small lymphatic vessels and be passively
E-cadherin are associated with a decrease in cellular/ transported in the lymph. Tumor emboli may be trapped
tissue differentiation and increased grade in carcinomas in the first lymph node encountered on their route, or
[120]. Many differentiated carcinomas express higher they may bypass regional draining lymph nodes to form
levels of E-cadherin mRNA, as do adjacent normal epi- distant nodal metastases (“skip metastasis”). Although
thelial cells, whereas poorly differentiated carcinomas this phenomenon was recognized in the late 1800s
do not. Mutations in the E-cadherin gene and abnormal- [193], its implications for treatment were frequently
ities of α-catenin, which is an E-cadherin-associated ignored in the development of surgical approaches to
protein, have been associated with the transition of cells treat cancers [263, 264].
from the noninvasive to the invasive phenotype [261]. Regional lymph nodes (RLN) in the area of a primary
Third, invasive tumor cells secrete enzymes capable neoplasm may become enlarged as a result of hyperplasia
of degrading basement membranes, which constitute a or growth of tumor cells in the node. Although the use
barrier between epithelial cells and the stroma. Epithelial of morphologic criteria for assessing prognoses based
cells and stromal cells produce a complex mixture of col- on lymph node appearance is debatable, lymphocyte-
lagens, proteoglycans, and other molecules, which contains depleted lymph nodes are believed to indicate a less
ligands for adhesion receptors and is permeable to mole- favorable prognosis than those demonstrating reactive
cules but not to cells [230]. morphologic characteristics [16]. Hyperplastic responses
To invade the basement membrane, a tumor cell must could indicate reactivity to autochthonous tumors, and
first attach to extracellular matrix (ECM) components this could benefit the host.
by a receptor-ligand interaction. One group of such cell Whether the RLN can retain tumor cells and serve as
surface receptors are the integrins which specifically a temporary barrier for cell dissemination is controver-
bind cells to laminin, collagen, or fibronectin [218]. sial. In most experimental animal systems used to inves-
Many integrins that bind to different components of the tigate this question, normal lymph nodes were subjected
ECM are expressed on the surface of human carcinoma to a sudden challenge with a large number of tumor cells,
cells. Tumor progression has been associated with a a situation that may not be analogous to RLN at the early
gradual decrease of integrin expression suggesting that stages of cancer spread in humans, when small numbers
the loss of integrins, coupled with the loss of E-cadherin, of cancer cells continuously enter the lymphatics. This
may facilitate detachment from a primary neoplasm. issue is important because of practical considerations
Subsequent to binding, tumor cells can degrade for surgical management of such neoplasms as cutaneous
connective-tissue ECM and basement membrane compo- melanoma. It raises the question, is elective prophylactic
nents [177]. The production of enzymes such as type lymph node dissection appropriate for the treatment of
IV collagenase (gelatinase, matrix metalloproteinase) and micrometastases? The biologic justification for elective
heparinase in metastatic tumor cells correlates with invasive lymph node dissection in patients with melanoma
capacity of human carcinoma cells. Type IV collagenolytic presumes that metastasis of some cutaneous melanomas
Sun-Jin Kim et al. 21

occurs first in the RLN, and that only at a later time do To a large degree, the rapid death of most circulating
tumor cells gain access to the circulation to reach distant tumor cells is probably due to such simple mechanical
organs. If this is the case, and RLN can act as a tempo- factors as blood turbulence. Tumor cell survival can be
rary barrier to the spread of cancer, removing the RLN increased by aggregation. Tumor cells can aggregate
with micrometastases could clearly increase the cure rate with each other or with host cells, such as platelets and
in subgroups of patients with melanoma. Some evidence lymphocytes [67].
exists that patients with melanomas of intermediate Once metastatic cells reach the microcirculation, they
thickness (1–4 mm) do in fact have an improved survival interact with cells of the vascular endothelium. These
rate subsequent to elective lymph node dissection. interactions include nonspecific mechanical lodgment
Similarly, some data suggest that an improved survival of tumor cell emboli as well as formation of stable adhe-
rate can be achieved for selected patients with head and sions between tumor cells and small-vessel endothelial
neck cancers by elective lymph node dissection or local cells. The organ distribution of metastatic foci is believed
treatment with x-irradiation [25]. to depend, in part, on the ability of blood-borne malig-
Recent advances in mapping of the lymphatics draining nant cells to adhere to specific endothelium and produce
cutaneous melanoma (by the use of dyes or radioactive endothelial cell retraction [185, 186].
tracers) have allowed surgeons to identify the lymph The formation of fibrin clots at sites of tumor cell
node draining the tumor site (i.e., the sentinel lymph arrest in the microcirculation can result in blood vessel
node) [36]. The presence of melanoma micrometastases damage [45, 46]. In some tumor systems, fibrin forma-
in sentinel lymph nodes is correlated with poor progno- tion is not essential for tumor cell implantation or
sis and hence indicates wide field dissection. A series of metastasis formation. The increased coagulability often
more than 500 melanoma cases with longer than 4 years’ observed in patients with cancer may be related to the
median clinical follow-up concluded that absence of high levels of thromboplastin found in certain tumors or
disease in sentinel lymph node correlates with increased to production of high levels of procoagulant-A activity,
disease-free status (in other nodes) and few or no skip which can directly activate factor X in the clotting pro-
metastases [119, 175]. These data suggest that elective cess. Since reduced blood flow could lead to increased
lymph node dissection when metastatic cells are present trapping of circulating tumor cells and perhaps to their
in sentinel lymph nodes produces beneficial results in increased survival, the use of anticoagulants in the treat-
patients with melanoma. ment or control of metastasis has been tried, albeit to a
limited success.
The adhesion of tumor cells to the vascular endothe-
lium is regulated by mechanisms similar to those used by
Hematogenous Metastasis leukocytes. The initial attachment of leukocytes to vas-
During blood-borne metastasis, tumor cells must survive cular endothelial cells is regulated by the selectin family
transport in the circulation, adhere to small blood of adhesion molecules which consists of three closely
vessels or capillaries, and invade the vessel wall. The related cell surface molecules. E-selectin, which is
mere presence of tumor cells in the circulation does not expressed by endothelial cells, mediates initial attach-
in itself constitute metastasis, since most cells released ment of lymphocytes (and tumor cells) by interaction
into the bloodstream are eliminated rapidly [60, 263, with specific carbohydrate ligands that contain sialylated
264]. Using radiolabeled tumor cells, we found that by fucosylated lactosamines. The expression of mucin-type
24 h after entry into the circulation, less than 1% of the carbohydrates on the surface of human colon carcinoma
cells are still viable, and less than 0.1% of tumor cells has been correlated with their metastatic potential [161],
placed into the circulation eventually survive to produce perhaps through differential interaction with E-selectins
metastases [60]. expressed on specific endothelial cells. The development of
Although most tumor cells are destroyed in the blood- firm adhesion requires the interaction of other adhesion
stream, it seems that the greater the number of cells molecules, another selective process in metastasis. Several
released by a primary tumor, the greater the probability classes of cell-to-cell adhesion molecules regulate this
that some cells will survive to form metastases. The adhesion. These include the hyaluronate receptor CD44
number of tumor emboli in the circulation appears to and its splice-variants [183], the integrins α5β1, α6β1,
correlate well with the size and clinical duration of the and α6β4, and the galactoside-binding galectin-3 [218].
primary tumor, and the development of necrotic and hemo- The arrest of tumor cells in capillary beds leads to the
rrhagic areas in large tumors facilitates this process by retraction of endothelial cells and the exposure of the
providing tumor cells easy access to the circulation [263]. tumor cells to the ECM. The adhesion of metastatic cells
22 The pathogenesis of cancer metastasis: relevance to therapy

to components of the ECM, such as fibronectin, laminin, several experiments [73]: Two weeks after normal,
and thrombospondin, facilitates metastasis to specific tumor-free mice were joined parabiotically to metastasis-
tissues, and peptides containing sequences of these com- bearing animals, there was no evidence of any tumor
ponents of the ECM can reduce formation of hematogenous growth in the “guest” animals. However, when the para-
metastases [246]. biont animals were allowed to survive for 4 weeks after
Extravasation of arrested tumor cells is believed to separation from the metastasis-bearing animals, 40%
operate by mechanisms similar to those responsible for developed lung metastases. Since the host mice did not
local invasion. Tumor cells can grow and destroy the have primary tumors at the time of parabiosis, the
surrounding vessel, invade by penetrating the endothe- metastases in the guest mice could have only arisen as
lial basement membrane, or they can follow migrating metastasis from metastases [101].
white blood cells [50]. The ability of malignant cells to
extravasate into surrounding tissues of particular organs
seems to arise, in part, from their selective adherence to
and invasion of certain tissues [187]. Malignant cells The Biologic and Metastatic
frequently penetrate thin-walled capillaries but rarely
invade arteries or arteriole walls, which are rich in elas-
Heterogeneity of Neoplasms
tin fibers. This resistance to invasion is not necessarily Only a few tumor cells that enter the circulation can
mediated by mechanical strength alone. Connective produce metastases. In fact, since less than 0.01% of
tissues have been shown to produce protease inhibitors, circulating cells are likely to produce a secondary
and these may block enzyme-dependent processes of growth, the development of metastases could represent
invasion. the fortuitous survival of a few tumor cells or the selec-
The invasion, survival, and growth of malignant cells tion from the parent tumor of a subpopulation of meta-
at particular secondary sites also involve their responses static cells endowed with properties that enhance their
to tissue or organ factors. Tumor cells can recognize survival [65, 66, 71]. Data generated by our research
tissue-specific motility factors that direct their move- group and many others prove that neoplasms are bio-
ment and invasion [196]. After tumor cells invade organ logically heterogeneous and that metastasis is indeed a
parenchyma, they must also respond to organ-specific selective process.
factors that influence their growth [187]. The first experimental proof of metastatic heteroge-
neity of neoplasms was provided by Fidler and Kripke
in 1977 working with the murine B16 melanoma [71].
Metastasis of Metastases Using the modified fluctuation assay of Luria and Delbruck
[157], they showed that different tumor cell clones, each
The tumor cells proliferating within metastases can derived from an individual cell isolated from the parent
invade host stroma, penetrate blood vessels, and enter tumor, varied dramatically in their ability to produce
the circulation to produce secondary metastases, the pulmonary nodules after intravenous inoculation into
so-called “metastasis of metastases” [73, 235, 236]. syngeneic recipient mice. Control subcloning proce-
Hart and Fidler used the preferential growth of B16 dures demonstrated that the observed diversity was not
melanoma metastases in specific organs. Following the a consequence of the cloning procedure [71]. The find-
intravenous injection of B16 melanoma cells into syn- ing that preexisting tumor cell subpopulations prolifer-
geneic mice, tumor growths developed in the lungs and ating in the same tumor exhibit heterogeneous metastatic
in fragments of lung or ovarian tissue implanted intra- potential has since been confirmed in many laboratories
muscularly into the quadriceps femoris but not in renal with a wide range of experimental animal tumors of dif-
tissue implanted as a control [101]. Tumor growth in the ferent histories and histologic origins [reviewed in 58,
specific transplanted organ could have been caused by 63, 64, 208, 209]. In addition, studies using young nude
the arrest and growth of tumor cells immediately follo- mice as models for metastasis of human neoplasms have
wing intravenous injection, i.e., “initial metastases.” shown that several human tumor lines and freshly iso-
Alternatively, tumor cells injected intravenously could lated tumors such as colon carcinoma, renal carcinoma,
have been arrested in the lungs, where they developed; and prostate cancer also contain subpopulations of cells
once metastases were established, tumor cells could enter with widely differing metastatic properties [64].
the circulation to be arrested at other organs and produce We studied the biologic and metastatic heterogeneity
“secondary metastases” [235]. To distinguish between in a mouse melanoma induced in C3H mice by chronic
these possibilities, Nicolson and Fidler performed exposure to ultraviolet B radiation and painting with
Sun-Jin Kim et al. 23

croton oil [134, 135]. One mouse thus treated developed We addressed the question of whether the cells that
a melanoma designated by Kripke as K-1735 [134]. The survive to form metastases possess a greater metastatic
original K-1735 melanoma was established in culture capacity than most cells in the parent neoplasm [243].
and immediately cloned [69]. In an experiment similar Some support for this possibility comes from the initial
in design to the one described for the B16 melanoma, in vivo selection experiments of the highly metastatic
the clones differed greatly from each other and from the B16-F10 cell line derived from the parent B16 mela-
parent tumor in their ability to produce lung metastases. noma [66]. Comparable results have been obtained with
In addition to differences in number of metastases, we the K-1735 tumor. When cells derived from the parent
also found significant variability in the size and pigmen- tumor were injected intramuscularly into the hind foot
tation of the metastases. Metastases to the brain, heart, pads of syngeneic mice, the resulting skin tumors
liver and skin were found as well; those growing in the produced spontaneous pulmonary metastases. Four cell
brain were uniformly pigmented, whereas those grow- lines were established from four individual lung nod-
ing in the lymph nodes, heart, liver, or skin generally ules harvested from four different mice. The finding that
had no pigment. all the lines derived from metastatic deposits produced
To determine whether the absence of metastasis pro- significantly more metastases than cells of the parent
duction by some (but not all) clones of the K-1735 was line was good evidence for the hypothesis that metastasis
a consequence of their immunologic rejection by the is a selective process, that is, cells populating metasta-
normal host [134–136], we examined their metastatic ses have an increased metastatic capacity [243].
behavior in young nude mice. In addition to the lack of Our studies demonstrate that the K-1735 melanoma is
a functional T-cell system, unstressed 3-week-old nude heterogeneous and contains both nonmetastatic and met-
mice are also deficient in natural killer cell activity. In astatic cells. In contrast, individual metastases (sponta-
such recipients, the immunologic barrier to metastatic neous pulmonary metastases) are more uniform. This
cells that also may be highly immunogenic is removed, suggests that metastatic foci could develop by a clonal
and they may thus successfully complete the process. expansion of a few surviving metastatic cells. Moreover,
This was true for cells of two clones that did not pro- it explains the observations showing that tumor cells in
duce metastases in normal syngeneic mice but produced primary and metastatic lesions differ in their antigenic
tumor foci in the young nude recipients. Most of the properties, biochemical characteristics, and response to
nonmetastatic clones were nonmetastatic in both normal cytotoxic drugs [reviewed in 61–63].
syngeneic and in the nude recipients. Therefore, the
clones’ failure to metastasize in syngeneic mice proba-
bly was not caused by their immunologic rejection by
the host but by their inability to complete one or another Clonal Origin of Cancer
step in the complex metastatic process.
Metastases
Multiple metastases proliferating in a host, even in the
same organ, often exhibit diverse biological characteris-
Enhanced Metastatic Potential tics of, for example, hormone receptors, antigenicity or
of Tumor Cells Harvested from immunogenicity, and response to various chemothera-
peutic agents. This diversity may result from the nature
Metastases of the pathogenesis of metastasis, the process of tumor
Our studies and most data reported by others have led us evolution and progression, or both.
to conclude that metastasis is a highly selective process Pathologists have long been aware that neoplasms
regulated by a number of as yet imperfectly understood frequently exhibit different morphological appearances
mechanisms. This belief is contrary to the once widely in different areas. For this reason, the malignant or benign
accepted notion that neoplastic dissemination is the ulti- nature of a tumor cannot be determined with confidence
mate expression of cellular anarchy. In fact, suggesting unless multiple sections obtained from all parts of the
that cancer metastasis is a selective process is a more opti- tumor are examined. The zonal differences in tumors
mistic view in terms of cancer therapy than one that are not restricted to morphology alone but include bio-
postulates that tumor dissemination is an entirely random logical characteristics such as growth rates, sensitivity
event. Belief that certain rules govern the spread of neo- to cytotoxic drugs, antigenicity, and pigmentation [74].
plastic disease implies that elucidation and understanding Since primary tumors are not uniform, it is possible that
of these rules will lead to better therapeutic interventions. tumor cell aggregates entering the circulation from one
24 The pathogenesis of cancer metastasis: relevance to therapy

zone of the tumor may be different from those entering can be protected from destruction in the circulation, and
from another zone. If an embolic aggregate originates a large aggregate of cells can more readily arrest in the
from a primary tumor’s homogeneous zone, regardless capillary bed of an organ. Since the aggregates we injected
of whether only one cell or several cells survived to pro- were large, each containing more than 20 cells, the results
liferate in distant organs, the resulting metastasis would suggest that the melanoma lung metastases resulted from
be like a primary tumor of unicellular origin. If a mixed the proliferation of a single viable cell within the embolus.
embolus derived from an area of zonal junctions enters Thus, regardless of whether an embolus is initially homo-
the circulation, the unicellular or multicellular origin geneous or heterogeneous, metastases can be unicellular
of the metastasis would depend on whether a single cell in origin.
or multiple cells survived to proliferate. To determine Collectively, these observations indicate that different
whether individual metastases are clonal and whether metastases arise from different progenitor cells and
different metastases can be produced by different pro- account for the well-documented differences in behavior
genitor cells, Talmadge et al. [244] performed a series of different metastases. Among individual metastases of
of experiments using the fact that x-irradiation of tumor proved clonal origins, however, heterogeneity can develop
cells induces random chromosome breaks and rear- rapidly to create significant intralesional heterogeneity.
rangements. Analyzing the karyotype composition of 21
individual melanoma lung metastases after cultivating
cells from individual lesions, this research group found
unique karyotypic patterns of abnormal, marker chromo-
Development of Biological
somes in most of the lines established from metastases, Diversity within and Among
which suggested that each metastasis originated from a
single progenitor cell. Similar results have been obtained
Metastases
in other rodent tumor systems. These studies revealed Clinical and histologic observations of neoplasms have
that the majority of metastases are of clonal origin. suggested that tumors undergo a series of changes during
Moreover, variant clones with diverse phenotypes are the course of the disease. A tumor initially diagnosed as
formed, rapidly resulting in the generation of significant benign, for example, can evolve over a period of many
cellular diversity within individual metastases [244]. months into a malignant tumor. This can best be demon-
Cancer metastases of a clonal origin can be produced strated in the case of human cutaneous melanoma where
by two different mechanisms, proliferation of a single the transformation of normal melanocytes and their
cell or of many cells. In the case of the second possibility, conversion into metastatic cells has been studied in detail
the cell aggregate at the metastatic site must have a by Clark and coworkers [32, 33]. This progression is
homogeneous composition. To determine which of these gradual and consists of a series of discrete irreversible
possibilities is responsible for the generation of clonal steps [107]. To explain the process of tumor evolution
K-1735 melanoma metastases, we injected C3H mice and progression as originally defined by Foulds in 1954
intravenously with aggregates of K-1735 cells consisting [81], Nowell [190] suggested that acquired genetic
of two distinct subpopulations [75]. Cells of line K-1735-M2 variability within developing clones of tumors, coupled
are highly metastatic and exhibit a stable normal karyo- with host selection pressures, can result in the emer-
type. Cells of the X-met-21 line are also highly meta- gence of new tumor cell variants that exhibit increasing
static but exhibit a stable, submetacentric chromosomal growth autonomy or malignancy. Nowell’s hypothesis
marker. After mixed aggregates (>20 cells) of these two [190] predicted that accelerating tumor progression
cell types were injected, individual lung metastases toward malignancy can be accompanied by increasing
were recovered and cultured, and each metastatic line genetic instability of the evolving cells. To test this
was subjected to chromosome analysis. We reasoned hypothesis, we have examined the metastatic stability
that if an experimental metastasis originated from a and rates of mutation of paired metastatic and nonmeta-
single proliferating cell, all tumor cells within the meta- static cloned lines isolated from four different mouse
static focus should express either the K-1735-M2 or the neoplasms. We found that highly metastatic cells were
X-met-21 chromosomal profile. This indeed was the phenotypically less stable than their nonmetastatic
case. Analysis of the distribution and fate of circulating counterparts. Moreover, in highly metastatic clones, the
tumor emboli has demonstrated that multicellular aggre- rate of spontaneous mutation was found to be several-
gates are more likely to give rise to a metastasis than a fold higher than in low-metastatic clones. These results
single-tumor-cell embolus. This is probably so because are in accord with the hypothesis that tumor progres-
tumor cells not on the periphery of circulating emboli sion occurs as a result of acquired genetic alterations.
Sun-Jin Kim et al. 25

Similar data have been reported for other neoplasms carcinoma cell lines. Inter- and intratumoral heterogeneity
[reviewed in 62, 65]. Evidence that genetic mechanisms in expression of these growth factors and their receptors
can be responsible for tumor progression comes from was reported. Different proteins produced by tumor cells
mutagenesis experiments using nitrosoguanidine. can regulate the interaction between tumor cells and
The finding that metastatic cells exhibit higher muta- the organ microenvironment [57]. Many protein tyrosine
tion rates than nonmetastatic cells [31], and that hetero- kinase receptors on tumor cells and tumor-associated endo-
geneity develops more rapidly in tumors containing few thelial cells can be induced or upregulated by ligands
subpopulations of cells [186, 208, 210] suggest that produced by tumor cells via autocrine and paracrine path-
accelerated tumor evolution and progression will result ways [55]. In many clinical trials, the presence of targets
in the rapid development of biologic diversity in metas- in patients was not confirmed, and the response rate was
tases, especially when such lesions are of clonal origin. unpredictable [247]. For inhibition of phosphorylated
EGFR, the presence of mutated receptors was reported
to be a predictable variable [158], but significant thera-
Intratumoral Heterogeneity for peutic responses of cancer cells with wild-type receptors
has also been reported [23, 269, 270]. These confusing
Expression of Tyrosine Kinase criteria for selection of patients for treatment strongly
indicate the need for better methodologies.
Growth Factor Receptors The data demonstrating inter- and intratumoral het-
It is well recognized that tumor progression, angiogene- erogeneity for expression of EGFR, VEGFR2, and
sis, and metastasis are regulated by the interaction of PDGFR-β in different human colon cancer specimens
tumor cells with the host organ microenvironment [70]. of different stages suggest that targeting a single tyrosine
The expression of growth factors and their receptors kinase receptor is not likely to provide significant thera-
varies within different zones of any given organ and may peutic results. In other words, targeted therapy is effec-
also differ among different cells within a tumor [62, 63]. tive only against its target and eliminating tumor cells
Overexpression of transforming growth factor-alpha that are dependent on one pathway, e.g., EGFR, is not
(TGF-α), epidermal growth factor (EGF), and the EGF likely to prevent the proliferation of tumor cells that are
receptor (EGFR) has been reported to be associated with independent of this pathway. These results agree with a
poor prognosis in different neoplasms [2, 111, 170, 253, recent report demonstrating biological heterogeneity of
271]. Expression of vascular endothelial growth factor tyrosine kinase receptors in other cancers [30, 173].
(VEGF) is associated with increased vascular permeabil- Indeed, simultaneous blocking of two protein tyrosine
ity, cell proliferation, and survival of endothelial cells kinase pathways produce more efficient therapeutic
[85, 86, 148, 272]. The expression of VEGF has also effects in prostate [128] and pancreatic [269] carcinoma
been correlated with microvessel density of neoplasms than did blocking of a single such pathway. Therapeutic
[52, 242] metastasis, and, hence, poor prognosis [53, 116, efficacy was further increased when three protein
140, 234, 262]. The expression of PDGF and its receptor tyrosine kinase pathways were inhibited [270].
(PDGFR) by tumor cells, tumor-associated endothelial Cancers are biologically heterogeneous for multiple
cells [126, 251], and pericytes and myofibroblasts [127, properties, including antigenicity, sensitivity to chemo-
131, 151, 269, 270] is common to many neoplasms. therapeutic agents, invasion, and metastasis [76, 77].
In clinical specimens of human colon carcinomas, while The progressive growth, metastasis, and survival of
PDGF is produced by tumor cells, the expression of tumor cells depend on their interaction with the organ
PDGFR is restricted to stromal cells, including tumor- microenvironment. The expression of multiple tyrosine
associated endothelial cells [131], i.e., tumor-associated kinase receptors by different tumor cells within a single
stromal cells generate a microenvironment favorable to neoplasm indicates that targeting a single tyrosine
the survival and progressive growth of tumor cells [213]. kinase may not produce eradication of the disease.
Specifically, PDGFR signaling is related to recruitment
of pericytes and control of interstitial fluid pressure [14,
192, 202, 213], which are favorable to the survival and Zonal Heterogeneity for Gene
progressive growth of neoplasms [155].
The expression of TGF-α, EGF, VEGF, and PDGF-β
Expression
and their respective receptors was examined in 12 human A better understanding of the cross-talk between cancer
colon cancer surgical specimens and in orthotopic tumors cells and the organ microenvironment involving multiple
in nude mice produced by two distinct human colon genes first requires the identification of gene expression
26 The pathogenesis of cancer metastasis: relevance to therapy

profiles within the tumor. Among current methods for receptor protein tyrosine kinase signaling pathways,
establishing these profiles, microarray analysis is the most response to stress, small GTPase-mediated signal trans-
powerful, and several such studies have been under- duction, hexose metabolism, cell death, response to
taken to predict disease outcome for individual patients external stimulus, and carbohydrate metabolism. These
[9, 10, 29, 115, 207, 216, 258, 259] and to identify data clearly demonstrate zonal heterogeneity for gene
cancer patients who should receive chemotherapy [17]. expression profiles within single tumors and suggest
For example, pretherapeutic gene expression profiling that characterization of zonal gene expression profiles is
has been undertaken to identify breast cancer patients essential if microarray analyses of genetic profiles are to
who should receive specific chemotherapy [17], to produce reproducible data, predict disease prognosis,
predict response of rectal cancer patients to preopera- and allow design of specific therapeutics [179, 180].
tive chemoradiotherapy [89], and to predict response of
breast cancer patients to docetaxel [28], and a specific
gene expression pattern was correlated with recurrence
in Dukes’ B colon carcinomas [265]. None of these Tumor-Organ Interaction: The
studies, however, accounted for the biologic heteroge-
neity of gene expression within a single tumor. Tumor
“Seed and Soil” Hypothesis
cells depend on multiple and redundant pathways for Clinical observations of cancer patients and studies with
growth, survival, and adaptation to the host microenvi- experimental rodent tumors have led cancer biologists
ronment [70, 76, 77, 155]. In malignant neoplasms, to conclude that the metastatic pattern of certain tumors
genetic instability of cancer cells frequently yields is organ-specific and independent of vascular anatomy,
extensive intratumoral heterogeneity in the pattern of rate of blood flow, and number of tumor cells delivered
structural chromosome aberrations [90, 92, 93, 95]. to each organ [reviewed in 62, 65, 263, 264]. Indeed, the
Three dimensional tumor growth and uneven fractional distribution and fate of hematogenously disseminated,
division of tumor cells within a single tumor mass can radiolabeled melanoma cells in experimental animals
give rise to biologically different central and peripheral conclusively demonstrated that tumor cells can reach
zones. The microenvironment of the central zone differs the microvasculature of many organs, but growth in the
significantly from that of the peripheral zone, and organ parenchyma occurs in only specific organs [60].
zonal heterogeneity for several molecules in different These findings, however, were not new.
tumor systems has been reported [102, 105, 129, 130, More than 100 years earlier, Paget reached a similar
138, 139, 191, 225]. Among these, bFGF, matrix metal- conclusion. In 1889, he asked, “What is it that decides
loproteinase-2 (MMP-2), and MMP-9 have been shown what organs shall suffer in a case of disseminated
to be highly expressed at the invasive edge of tumors cancer?” Paget’s study was motivated by the discrepancy
[105, 130, 131, 138, 139], whereas the cell-to-cell cohe- between considerations of blood flow and the frequency
sion molecule, E-cadherin, was downregulated at the of metastases in different organs. He examined the
periphery of the tumors [105, 130, 131, 138, 139, 191]. autopsy records of 735 women who died of breast cancer
In fact, the ratio of expression of MMP-2 and MMP-9 to and many other patients with different neoplasms and
E-cadherin (MMP/E-cadherin ratio) at the periphery of noticed the high frequency of breast cancer metastasis to
the tumors correlated with metastatic potential and recur- the ovaries and the variations in incidence of skeletal
rent disease [138, 139]. metastases produced by different primary tumors. These
Affymetrix HG-U133-Plus 2.0 array and laser capture findings were not compatible with the view that meta-
microdissection techniques, demonstrated that different static spread was due to “a matter of chance” or that tissues
zones of the same pancreatic tumor exhibit differential “played a passive role” in the process. Paget [193] con-
expression of genes [179, 180]. The study of 1,222 genes cluded that metastasis occurred only when certain favored
demonstrated statistically significant differences. Bioinfor- tumor cells (the “seed”) had a special affinity for the growth
matic functional analysis revealed that 346 upregulated milieu provided by certain specific organs (the “soil”). The
genes in the peripheral zone were related to cytoskeleton formation of metastasis required the interaction of the right
organization and biogenesis, cell cycle, cell adhesion, cells with the compatible organ environment.
cell motility, DNA replication, localization, integrin- In 1928, Ewing [50] challenged Paget’s seed and soil
mediated signaling pathway, development, morpho- theory and hypothesized that metastatic dissemination
genesis, and IκB kinase/NF-κB cascade. In the central occurs purely by mechanical factors that are a result of
zone, 876 upregulated genes were related to the regula- the anatomical structure of the vascular system. Both
tion of cell proliferation, transcription, transmembrane of these explanations have been evoked separately or
Sun-Jin Kim et al. 27

together in order to explain the metastatic site preference A clear demonstration of organ-site specific metastasis
of certain types of neoplasms. In a review of clinical comes from studies of experimental brain metastasis.
studies on site preferences of metastases produced by Two murine melanomas were injected into the carotid
different human neoplasms, Sugarbaker [235] concluded artery to simulate the hematogenous spread of tumor
that common regional metastatic involvements could be emboli to the brain. The K-1735 melanoma generated
attributed to anatomical or mechanical considerations, lesions only in the brain parenchyma, whereas the B16
such as efferent venous circulation or lymphatic drain- melanoma produced only meningeal growths [219].
age to regional lymph nodes, but that metastasis in distant Similarly, different human melanomas [220] injected
organs from numerous types of cancers were indeed site- into the internal carotid artery of nude mice also pro-
specific. duced unique patterns of brain metastasis. Distribution
Experimental data supporting Paget’s 1889 seed and analysis of radiolabeled melanoma cells injected into
soil hypothesis were provided a century later by Hart and the internal carotid artery ruled out the possibility that
Fidler [101], who studied the preferential growth of B16 the patterns of initial cell arrest in the microvasculature
melanoma metastases in specific organs. Following the of the brain predicted the eventual sites of growth.
intravenous (i.v.) injection of B16 melanoma cells into Rather, the different sites of tumor growth in the brain
syngeneic C57BL/6 mice, tumor growths developed in involved interactions between the metastatic cells and
the natural lungs and in grafts of pulmonary or ovarian brain endothelial cells, and the response of tumor cells
tissue implanted either subcutaneously (s.c.) or intramus- to local growth factors. In other words, site-specific
cularly (i.m.). In contrast, neoplastic lesions failed to metastases were produced by tumor cells that are recep-
develop in control grafts of similarly implanted renal tive to their new environment.
tissue or at the site of surgical trauma. Parabiosis experi-
ments suggested that the growth of the B16 melanoma in
ectopic lung or ovarian tissue was due to the immediate
arrest of circulating neoplastic cells and not to shedding of
Regulation of Tumor Cell Gene
malignant cells from foci growing in the natural lungs. Expression by the Organ
Quantitative analysis of tumor cell arrest and distribution
using cells labeled with [125I]-5-iodo-2´-deoxyuridine
Microenvironment
indicated that the growth of tumors in the implanted organs Tumor cells with different metastatic capabilities have
was not due to an enhanced initial arrest of B16 cells. No been shown to differ in expression of proteins, such as
significant differences in immediate tumor cell arrest were collagenases, E-cadherin, IL-8, bFGF, and many others
detected between implanted fragments of lungs (tumor- [65]. In most cases, however, these differential expres-
positive) and kidney (tumor-negative) or between organ- sions were most evident in tumor cells growing at
bearing and contralateral control limbs. These data anatomically correct sites. Many biologic investigations
demonstrated that the outcome of metastasis is dependent of solid tumor cells often use cell lines growing in vitro
on both tumor cell properties and host factors and sup- as monolayer cultures. While easy to accomplish, mono-
ported the seed and soil hypothesis as an explanation of layer cultures are not subjected to any cross talk, e.g.,
the nonrandom pattern of cancer metastasis. paracrine signaling pathways associated with growth
The introduction of peritoneovenous shunts for palli- in vivo [37].
ation of malignant ascites provided an opportunity to Clinical observations of cancer patients and studies in
study some of the factors affecting metastatic spread in rodent models of cancers have concluded that certain
humans. Tarin et al. [245] described the outcome in tumors tend to metastasize to certain organs [264]. The
patients with malignant ascites draining into the venous concept that metastasis results only when certain tumor
circulation, with the resulting entry of viable tumor cells cells interact with a specific organ microenvironment
into the jugular veins. Good palliation with minimal was originally proposed in Paget’s venerable “seed and
complications was reported for 29 patients with various soil” hypothesis [193]. Indeed, spontaneous metastasis
neoplasms. The autopsy findings in 15 patients substan- is produced by tumors growing at orthotopic sites,
tiated the clinical observations that the shunts did not whereas the same tumor cells implanted into ectopic
significantly increase the risk of metastasis. In fact, sites fail to produce metastasis [124].
despite continuous entry of millions of tumor cells into To determine the influence of the microenvironment
the circulation, metastases in the lung (the first capillary on changes in gene expression, microarray analyses
bed encountered) were rare. These results provide com- were done on three variant lines of a human pancreatic
pelling verification of the seed and soil hypothesis. cancer with different metastatic potentials [141, 179].
28 The pathogenesis of cancer metastasis: relevance to therapy

The variant lines were grown in tissue culture in the from the kidney or colon do not. The incubation of
subcutis (ectopic) or pancreas (orthotopic) of nude mice. human colon cancer cells [51] and human renal cancer
Compared with tissue culture, the number of genes cells [91] with IFN-β significantly reduced the expres-
whose expression was affected by the microenviron- sion and activity of collagenase type independently of
ment was upregulated in tumors growing in the subcutis antiproliferative activity.
and pancreas. In addition, highly metastatic pancreatic
carcinoma cells growing in the pancreas expressed
significantly higher levels of 226 genes than did the low
metastatic variant cells. Growth of the tumor cells in the
Regulation of Angiogenesis by
subcutis did not yield similar results, indicating that the Organ Microenvironment
the orthotopic microenvironment significantly influences
The intensity of the angiogenic response varies consid-
gene expression in pancreatic cancer cells [141, 179].
erably among different types of tumors and within
different zones of a single tumor [48]. The rate of tumor
cell division is still several orders of magnitude greater
Regulation of the Invasive than the rate of neovascularization. As tumors expand,
their microenvironment is often hypoxic [260]. Hypoxia
Phenotype by the Microenvironment is often associated with the activation of the transcrip-
The metastatic capacity of human colon cancer cells grow- tion factor hypoxia-inducible factor-1 alpha (HIF-1α) to
ing in orthotopic tissues of nude mice directly correlates initiate the transcription of genes, e.g., VEGF/VPF [54].
with the level of collagenase type IV activity [233]. VEGF/VPF increases the permeability of blood vessels
Histological examination of the human colon carcinomas by stimulating the functional activity of vesicular-vacuolar
growing in the subcutis, wall of the colon, or kidney of organelles, clusters of cytoplasmic vesicles and vacuoles
nude mice revealed a thick pseudocapsule around the located in microvascular endothelial cells [45–47].
subcutaneous but not cecal or kidney tumors [61, 70, 76]. VEGF also induces migration, protease production, and
These differences suggested that the organ environment endothelial cell proliferation [54]. In addition, VEGF/
could influence the ability of metastatic cells to invade VPF regulates endothelial cell survival by activating the
host stroma. Significant differences were found in the phosphatidylinositol-3 kinase/Akt signal transduction
levels of secreted type IV collagenases between human pathway and stimulating expression of the antiapoptotic
colon cancer cells growing subcutaneously or in the proteins Bcl-2 and A1 [110].
cecum of nude mice. In the medium conditioned by HIF-1α signaling also encodes the polypeptide chains
human colon cancer cells derived from subcutaneous of PDGF [100]. The functional activity of PDGF is, to a
implants, we detected only a latent form of the 92-kDa large extent, determined by the anatomical location of a
type IV collagenase. In contrast, both latent and active specific tumor. For example, in pancreatic tumors,
forms of the 92-kDa type IV collagenase were found in PDGF has been shown to stabilize developing vascular
culture medium conditioned by tumor cells harvested networks by recruiting pericytes to support the imma-
from cecal tumors. Moreover, cancer cells grown in the ture blood vessel walls [13, 14]. In tumors of the central
cecum secreted more than twice as much enzymes as nervous system, PDGF stimulates the release of VEGF
the subcutaneous tumors [178]. from the tumor-associated endothelium [96]. In contrast,
The invasive ability of human colon cancer cells is tumors in the skin rely on PDGF signaling to regulate
directly influenced by organ-specific fibroblasts. Primary the level of interstitial fluid pressure in the tumor [199].
cultures of nude mouse fibroblasts from skin, lung, and In prostate cancer bone metastasis (see next section),
colon were established, and invasive and metastatic PDGF functions as a survival factor for tumor endothe-
human colon cancer cells were cultured alone or with lial cells by activating the intracellular effectors MAPK
the fibroblasts. The cancer cells grew on monolayers of and Akt [142, 143].
all three fibroblast cultures but did not invade through The vascular endothelium is regarded as structurally
skin fibroblasts [51]. Cancer cells growing on plastic and functionally heterogeneous [88]. To examine this
and on colon or lung fibroblasts produced significant diversity, we generated a broad panel of microvascular
levels of latent and active forms of type IV collagenase, endothelial cells from various organs of H-2Kb-tsA58
whereas colon cancer cells cocultivated with nude mouse transgenic mice [142]. cDNA expression profiles generated
skin fibroblasts did not. One possible explanation is that on the endothelial cells predicted significant organ-
fibroblasts from the skin produce IFN-β, whereas those specific differences in expression levels of tyrosine kinase
Sun-Jin Kim et al. 29

receptors, chemokine receptors, and proteins that regu- Macrophage-derived metalloelastase has been shown to
late the efflux of toxic substrates; these were confirmed be responsible for the generation of angiostatin in Lewis
at the protein level. Endothelial cells derived from the lung carcinoma [42], and the addition of plasminogen to
mouse brain expressed measurable levels of PDGF-Rβ, 3LL Lewis lung carcinoma cells cultured in vitro did not
the chemokine receptor CXCR-2, and P-glycoprotein, result in the generation of angiostatin. However, its addi-
whereas endothelial cells from the pulmonary circula- tion to co-cultured macrophages and carcinoma cells did
tion did not express detectable levels of these proteins. [42], suggesting that elastase activity in macrophages
The organ-derived endothelial cells also exhibited vast was significantly enhanced by the cytokine GM-CSF
differences in response to stimulation with endothelial which was secreted by the tumor cells [137], leading to
cell mitogens. Endothelial cells originating from the the generation of plasminogen.
brain and liver showed the greatest increase in cell divi-
sion in response to basic fibroblast growth factor, while
EGF was the most potent mitogen for endothelial cells Regulation of Response to
derived from the lung and uterus.
Chemotherapy by the Organ
Microenvironment
Influence of Lymphoid Cells Clinical observations suggest that the organ environment
can influence the response of tumors to chemotherapy.
on Angiogenesis For example, in women with breast cancer, lymph node
The regulation of angiogenesis by T lymphocytes, and skin metastases are more sensitive to chemothera-
macrophages, and mast cells, is well established [55, 59, peutic agents than are metastases residing in either the
68, 83, 160, 164, 169, 204, 223, 240]. For example, lung or bone [41]. Several intrinsic properties of tumor
invasive cutaneous melanoma is often associated with cells can render them resistant to chemotherapeutic
a local inflammatory reaction involving T lympho- drugs, including increased expression of the mdr genes,
cytes and macrophages, a condition often associated leading to overproduction of the transmembrane trans-
with an increased risk of metastasis [19, 217]. The port protein P-glycoprotein (P-gp) [18, 249]. Expression
relatively slow growth of tumors in aging mice has been of P-gp often parallels increases in dose or duration of
closely linked with decreased vascularization [133] chemotherapy. Indeed, increased levels of P-gp can be
associated with a diminished immunological response induced by selecting tumor cells for resistance to natural
[38, 92]. The role of tumor vascularization and its effect product amphiphilic anticancer drugs. Nevertheless,
on tumor growth in immunosuppressed mice was inves- elevated expression of P-gp accompanied by develop-
tigated and it was concluded that the growth of the ment of the multidrug resistance (MDR) phenotype has
immunogenic B16 melanoma was delayed in myelosup- also been found in many solid tumors of the colon,
pressed mice as compared to control mice [98]. Similarly, kidney, and liver that had not been previously exposed
tumor growth in mice pretreated with doxorubicin to chemotherapy [249].
(DXR) and then injected with normal splenocytes 1 day One of the most striking examples of site-specific
before tumor challenge was comparable to that in the variations in therapeutic response was observed following
control mice, implicating myelosuppression as a cause of implantation of colon carcinoma cells into different
retardation of tumor growth and vascularization. Similar anatomic locations of nude mice (using the highly meta-
results were obtained with athymic nude mice [98]. static KM12L4 human colon carcinoma cell line) or
Many other studies have recognized the importance of syngeneic BALB/c mice (using the CT-26 murine colon
macrophages in tumor angiogenesis [5, 204–206, 239]. carcinoma). Mice received injections of the KM12L4a
Macrophages can produce more than 20 molecules that cells into either the subcutis (ectopic site), spleen (lead-
influence endothelial cell proliferation, migration, and ing to experimental liver metastasis), or cecum (growth
differentiation [204, 205]. Macrophages may modify the at the orthotopic site). Tumor-bearing mice were given
extracellular matrix, thereby modulating angiogenesis doxorubicin and subsequently evaluated for response to
either through direct production of extracellular matrix treatment. Up to 80% growth of subcutaneous tumors
components or proteases that alter the structure and was inhibited by two i.v. injections of doxorubicin as
composition of the extracellular matrix [240]. Macro- compared to about 40% inhibition of the intracecal
phages have also been shown to produce antiangiogenic tumors and less than 10% inhibition of lesions in the
molecules, such as thrombospondin-1 [40, 152, 153, 205]. liver [267]. Similar results were obtained with murine
30 The pathogenesis of cancer metastasis: relevance to therapy

colon cancer cells. Subcutaneous tumors were sensitive to up-regulate the growth factor receptors and the acti-
to treatment with doxorubicin; lung metastases were vation of these receptors on tumor cells (autocrine effect)
insensitive [42]. However, the tumor cells at both of and on tumor-associated endothelial cells (paracrine
these sites were equally sensitive to 5-FU, a drug whose effect). These effects can lead to downstream signa-
activity is not influenced by expression of the MDR ling that activate mitogen-activated protein kinases
phenotype. Northern blot analysis showed that the rela- and phosphatidylinositol-3 kinase pathways and lead to
tive expression of the mdr1 and mdr3 genes was great- downregulation of the apoptotic proteins caspases 3 and
est in the cecum, liver, and lungs of mice, and this 8 [8, 142, 143, 229]. Activation of growth factor recep-
expression correlated with the relative resistance of tors on tumor cells and on the tumor-associated endothe-
tumor cells. Indeed, this expression of mdr was transient lial cells is likely to increase the expression of
and the subsequent culture of cells from a liver metasta- antiapoptotic proteins, causing endothelial cell resis-
sis for 7–10 days resulted in a decrease of mdr expres- tance to chemotherapy, even though these cells divide
sion to the level in tumor cells maintained in culture every 30–40 days. Inhibition of this growth factor recep-
[43]. These events resulted from organ-specific modula- tor activation by receptor antagonists in tumor-associ-
tion of tumor cell properties. These findings are not ated endothelial cells (and tumor cells) can lead to BAX
restricted to experimental systems. In patients with induction, activation of caspase-8, and down-regulation
colon carcinoma, elevated P-gp expression is found on of BCL-2 and NF-κB [1, 24], thereby increasing the
the invasive edge of the primary tumor (growing in the susceptibility of tumors to chemotherapy. If the tumor-
colon) and in metastases located in lymph node, lung, associated endothelial cells continue to cycle, the use of
and liver [129]. Whether this finding is due to selection receptor antagonists [6, 7, 125–128] combined with
or adaptation is unclear. anticycling drugs can cause the destruction of the vascu-
lature within neoplasms, leading to the apoptosis of
adjacent tumor cells [20, 21, 193, 232].
Targeting the Vasculature
Because systemic antitumor therapy fails primarily because
of the genetic instability and biologic heterogeneity
Targeting the Expression of
of neoplasms, therapeutic agents that target a tumor’s Platelet-Derived Growth Factor
vasculature, a genetically stable and essential compo-
nent of tumors, have been explored as an alternative to
Receptor by Reactive Stroma
conventional therapy. The structure and architecture of Colon carcinoma cells produce various growth factors
tumor vasculature can differ dramatically from those of and cytokines that contribute to progressive growth and
normal organs [48, 182, 189]. Modern techniques, such metastasis [117]. One example is the family of PDGFs,
as phage display targeting, have defined “vascular members of a family of dimeric disulfide-bonded growth
addresses” that may be distinct for different organs and factors exerting their biological effects through activa-
for tumors in those organs [194]. In normal human tion of two structurally related tyrosine kinase recep-
tissues, for example, endothelial cells are long lived tors, the PDGF-α- and -β receptors [104]. PDGF consists
and recycle every 3–5 years, whereas tumor-associated of dimeric forms, including PDGF-AA, PDGF-BB,
endothelial cells can recycle every 30–40 days. The rate PDGF-AB, PDGF-CC, and PDGF-DD [15, 144, 149].
of endothelial cell turnover has been reported to be The α-receptor binds all possible forms of PDGF except
<0.1% in normal human tissues, whereas in malignant PDGF-DD, whereas the PDGF-β receptor preferentially
human tumors, it is 2–9.9% [48, 49]. binds PDGF-BB. PDGF-Rβ is expressed on many tumor
Endothelial cells that line blood vessels in different types, and PDGF-BB is an important autocrine growth
organs are also phenotypically distinct. The expression factor for many cell types, including gliomas, sarcomas,
of various cytokines and growth factors and their rele- pancreatic carcinoma, and prostate cancer [103]. PDGF-R
vant receptors on both tumor cells and organ-specific signaling has also been reported to stimulate angiogenesis
endothelial cells differs in different organ microenvi- [213], recruit pericytes [14, 192], and control the intersti-
ronments, and the interaction of these growth factor tial fluid pressure in stroma, thereby affecting transvas-
ligands (i.e., EGF, TGF-α, VEGF, and PDGF) with their cular transport of chemotherapeutic agents [200–203].
relevant receptors is essential for endothelial cell A recent study of human colon cancer clinical specimens
survival and growth [6, 7, 128, 142, 143]. For example, revealed that PDGF-Rβ and phosphorylated PDGF-Rβ are
tumor cells that produce growth factor ligands are likely not expressed on tumor cells but, rather, are predominantly
Sun-Jin Kim et al. 31

expressed in stromal cells and in pericytes surrounding the parenchyma. These cells are activated by various
the tumor microvessels [131, 132, 238, 241]. stimuli from tumor cells and undergo transformation
Imatinib, a derivative of 2-phenylaminopyrimidine, into myofibroblasts, which are characterized by expres-
was originally developed as a competitor for an ATP- sion of αSMA [211]. In general, tumor-associated stroma
binding site of the Abl protein tyrosine kinase [44]; is an abundant source of tumor-promoting growth
however, it is also a potent tyrosine kinase inhibitor of factors and cytokines, and stromal cells activated by
c-Kit and PDGF-R [22]. We have reported that imatinib cancer cells can in turn regulate the growth and progres-
can slow both the progressive growth of human pancre- sion of carcinoma cells [112, 165, 222, 250]. The pro-
atic carcinoma in nude mice [114, 270] and the growth gressive growth of colorectal tumors in experimental
of experimental bone metastasis of human prostate animals has been correlated with proliferation of myofi-
cancer [127, 251]. broblasts, whereas regression of colon tumors has been
To examine the role of PDGFR on colon cancer stromal linked to a fibrous capsule, suggesting that the presence
cells, we examined the therapeutic effect of imatinib of reactive myofibroblasts may inhibit growth of tumor
administered as a single agent or in combination with the cells [150]. Wounding has been associated with tumor-
chemotherapeutic irinotecan against human colon carci- promoting effects [224] and, functionally, tumor-associ-
noma cells growing in orthotopic (cecum and liver) and ated stromal cells are similar to stromal cells in healing
ectopic (subcutis) organs of nude mice. Blockade of wounds insofar as expression of myodifferentiation
PDGF-Rβ signaling by oral administration of imatinib or markers, production of ECM, expression of growth fac-
imatinib combined with irinotecan significantly inhibited tors and cytokines, and neovascularization [121–123].
the growth of orthotopic tumors and the incidence of The combination of imatinib and irinotecan com-
lymph node metastasis in nude mice. Histopathological pletely inhibited tumor cell growth at the abdominal
analysis of human colon carcinoma growing in the cecum wound healing site induced at the time of tumor cell
and liver of nude mice treated with imatinib alone or injection into the cecum [131]. Imatinib may inhibit
with imatinib combined with irinotecan demonstrated stromal reaction at the orthotopic primary site and the
decreased stromal reaction and inhibition of phosphory- surgical wound. PDGF-R signaling pathway also plays
lated PDGF-Rβ in the tumor-associated stromal cells. an important role in increasing tumor interstitial hyper-
These effects were associated with the inhibition of tumor tension, which may block the accumulation of antitu-
cell proliferation, an increase of apoptosis in stromal mor drugs [201–203].
cells, and a decrease in the number of pericytes surrounding The microvasculature in both tumor tissue and normal
the tumor-associated microvessels. In contrast, treatment colon mucosa consists of endothelial cells, pericytes (mural
of mice with imatinib alone or imatinib combined with cells or smooth muscle cells), and basement membranes.
irinotecan did not affect the growth of colon carcinoma All of these components are thought to be abnormal in
cells in the subcutaneous space, demonstrating once again tumor vessels. Pericytes are key cells in vascular develop-
that the biology of tumors differs with the organ microen- ment, stabilization, maturation, and remodeling [12, 87].
vironment [58, 76]. Functional-blocking antibodies that target PDGF-Rβ
In general, tumor cells in a neoplasm are biologically block pericyte recruitment during vascular development
heterogeneous and their phenotype can be modified by [252]. In our study, desmin-positive pericytes were found
the organ microenvironment [63, 74]. Histologically, on microvessels in both normal organs and colon cancers,
human carcinoma tissues are composed of both paren- albeit with different morphological characteristics. Speci-
chyma and stroma. Tumor stroma consists of fibroblasts, fically, pericytes in tumor-associated vessels, but not those
smooth muscle cells, inflammatory cells, microvessels, in vessels of normal colon mucosa, were enlarged and
and abundant extracellular matrix (ECM) [99]. Tumor- overexpressed PDGF-Rβ and p-PDGF-Rβ [131]. In agree-
associated stroma at both primary sites and metastatic ment with earlier reports [14, 269, 270], we found that
sites generate a favorable microenvironment for the treatment with imatinib decreased pericyte coverage on
survival of cancer cells [155]. Fibroblasts are activated tumor-associated endothelial cells. The inhibition of
by various growth factors and cytokines that are released PDGF-R signaling by a protein tyrosine kinase inhibitor
by cancer cells, for example TGF-β, PDGF, and bFGF. decreased pericyte recruitment and attachment to endothe-
Activated fibroblasts express αSMA, leading to the term lial cells and destabilized tumor vasculature by killing
‘myofibroblasts’ [45]. Stromal reaction (desmoplasia), pericytes. In the absence of decreased vessel density in
such as myodifferentiation of fibroblasts [109, 145, 215] tumors treated with imatinib combined with irinotecan,
clearly alters the stromal phenotype. In the liver, hepatic functional rather than quantitative changes of the tumor
stellate cells are the only mesenchymal cells present in vasculature by imatinib may diminish tumor growth [84].
32 The pathogenesis of cancer metastasis: relevance to therapy

Recently, targeting the VEGF pathway to inhibit tumor To determine whether the PDGF-R expressed on
angiogenesis is attracting attention as a novel cancer tumor-associated endothelial cells could be the primary
therapy [113]; however, it has been suggested that VEGF- target of imatinib, we selected a multidrug-resistant
targeting therapies are mostly active against immature variant of the human PC-3MM2 prostate cancer. These
vessels [13], i.e., normalization of the tumor vasculature cells were implanted into the tibia of nude mice, and
[118]. Because imatinib inhibits pericyte coverage on when the lesions were growing progressively, the mice
tumor vasculature, it will be very interesting to deter- were randomized to different treatment regimens. The
mine if the combined inhibition of PDGF-R and VEGF-R bone tumors were resistant to taxol, whereas treatment
signaling may produce synergistic antivascular effects with imatinib and taxol produced a significant decrease
[45, 118, 200]. Multi-target tyrosine kinase inhibitors are in tumor incidence, bone lysis, and incidence of lymph
under current investigation in clinical trials. Sorafenib node metastasis. Initially, this treatment produced apop-
and sunitinib target not only multiple VEGF-Rs but also tosis of tumor-associated endothelial cells (but not
PDGF-Rβ, and phase II studies show promising activity tumor cells), followed 1 week later by apoptosis of the
of these molecules in renal cell carcinoma [180], likely tumor cells. Collectively, these studies demonstrated
due to inhibition of angiogenesis [266]. that the primary targets for imatinib treatment are the
tumor-associated endothelial cells, i.e., an anti-vascular
therapy [126].

Anti-Vascular Therapy of Prostate


Cancer Bone Metastasis Targeting Platelet-Derived
In clinical specimens of human prostate cancer bone
metastasis and in experimental models of orthotopic
Growth Factor Receptor on
human prostate cancers in the long bones of nude mice, Endothelial Cells of Multidrug
the expression of PDGF and PDGF-R correlates with
the growth of metastatic tumor cells in the bone paren-
Resistant Prostate Cancer
chyma. Specifically, prostate cancer cells growing adja- The major cause of death from prostate cancer is due to
cent to bone tissue express high levels of the PDGF metastases that are resistant to conventional therapies.
protein and the PDGF-R, which is phosphorylated, and Genetic instability of tumor cells leading to biological
tumor-associated endothelial cells express high levels heterogeneity is largely responsible for the development
of phosphorylated PDGF-R [127, 128]. Oral adminis- of hormone-refractory prostate cancer cells [10, 11, 56,
tration of the PDGF-R tyrosine kinase inhibitor imatinib 197, 198, 254–257], and selection pressures by chemo-
(STI571) to nude mice blocks the PDGF signaling path- therapeutic agents results in the emergence of cells that
way by inhibiting phosphorylation of its receptors [251]. are resistant to chemotherapy [122, 147, 159, 198, 221,
Oral administration of STI571 or STI571 plus injectable 231]. These resistant cells often express the MDR1 gene
taxol to male nude mice reduced the incidence and size and its product, P-glycoprotein (P-gp) [94, 248, 257].
of primary tumors and bone lesions and prevented bone Methods to overcome multidrug resistant activity by
lysis as measured by digital radiography. Immuno- blocking transmembrane drug efflux pump action [82,
histochemical analysis of control, untreated bone 166–168, 176], liposomal encapsulation of cytotoxic
lesions, demonstrated that human prostate cancer cells drugs [18], immunotherapeutic monoclonal antibody
growing adjacent to the bone expressed high levels of targeting P-glycoprotein [214] or protein toxins selec-
PDGF and activated (phosphorylated) PDGF-R, whereas tively killing cells expressing P-glycoprotein on their
tumor cells growing in the adjacent musculature follo- surfaces [168] are under clinical investigation.
wing lysis of the bone did not. Treatment with STI571 Multidrug resistance-associated protein (MRP) was
and more so with STI571 plus taxol significantly inhib- found in multidrug resistant prostate cancer cell lines
ited phosphorylation of PDGF-R on tumor cells and that do not express P-glycoprotein [34]. MRP1 [237,
endothelial cells, decreased tumor cell proliferation, 256] and MRP2 [255] have been reported to be expressed
and induced significant apoptosis in tumor cells and in prostate cancer cell lines, but there are very few
tumor-associated endothelial cells. These data indicate compounds with proven MRP1-associated multidrug
that targeting PDGF-R phosphorylation can produce resistant-modulating capacity at present. Glutathione and
significant therapeutic effects against prostate cancer glutathione-S-transferase (GST) detoxification systems
bone metastasis [125–128]. are another drug-resistance mechanism reported to
Sun-Jin Kim et al. 33

protect cancer cells against the lethal effects of chemo- tumor-associated endothelial cells as well as PDGF-R-
therapy [174]. GST-π inactivation is related to early steps expressing tumor cells. The expression of phosphorylated
of prostate carcinogenesis [146, 237], whereas the hor- PDGF-R is known to activate anti-apoptotic pathways
mone-independent disseminated prostate cancers clearly involving Akt, PI3K, MAPK, and Bcl-2 [142, 143], and
expressed GST-π [256]. However, reversal of multidrug treatment with imatinib can inhibit this effect. These data
resistance in prostate cancer by challenging the glutathi- suggest that a major target for imatinib and paclitaxel
one pathway has been observed in vitro [212]. Many therapy could well be the tumor-associated endothelial
changes in the regulatory processes of apoptosis appar- cell. We tested this possibility by using MDR human
ently contributed to the malignant phenotype of prostate prostate cancer cells.
cancer cells [35, 39, 188, 256], and methods to stimulate The acquisition of multidrug resistance by leukemia
apoptosis are currently under development. However, cells [162], hepatocellular carcinoma [273], and human
despite progress in our understanding of the biology of lung carcinoma [163] has been shown to decrease
MDR, therapeutic approaches that directly target tumor tumorigenicity and prolong tumor cell doubling time.
cells have not been successful. In agreement with these reports, the in vivo growth of the
To overcome multidrug resistance of prostate cancer, human prostate cancer PC-3MM2 selected for multidrug
one may wish to affect the host factors in the organ resistance (PC-3MM2-MDR) was slower than the paren-
microenvironment, such as the vasculature [26, 63, 72, tal tumor cells. The ‘seed and soil’ theory [63, 193] held
78–80, 122]. Because all cells in the body depend on an true regardless of the MDR phenotype. Similar to parental
adequate supply of oxygen and nutrients and the ability cells, MDR tumor cells growing in the bone (but not in
to remove toxic molecules, therapy directed against the muscle) expressed IL-8, bFGF, EGF, EGF-R, PDGF,
tumor-associated endothelial cells can destroy tumor and PDGF-R. Endothelial cells of tumor-associated vessels
cells, regardless of their biological heterogeneity [77]. in bone lesions also expressed PDGF-R on their surface,
Recent reports have demonstrated statistically signifi- and treatment with imatinib and paclitaxel inhibited the
cant therapeutic results in experimental human prostate phosphorylation of the PDGF-R on both tumor cells and
cancer bone metastasis in nude mice treated with a com- endothelial cells. The PC-3MM2-MDR bone lesions
bination of paclitaxel and various tyrosine kinase inhibi- responded to systemic administration of imatinib and
tors directed toward PDGFR or EGFR [125–128, 251]. paclitaxel (but not to paclitaxel administered alone),
These experimental data have been successfully translated raising the possibility that imatinib could have sensi-
to clinical trials [45, 46]. Since both tumor cells and tumor- tized the tumor cells to paclitaxel. Immunohistochemical
associated endothelial cells can express these activated analyses revealed that early apoptosis (after 2–3 weeks
growth factor receptors, it has not been clear whether of chemotherapy) was mostly limited to tumor-associ-
the combination treatment targeted the tumor cells or the ated endothelial cells. The multidrug resistant tumor
endothelial cells or both. We reported that multidrug resis- cells were still treatment-resistant. In contrast, parental
tant PC-3MM2-MDR human prostate cancer cells growing cells underwent extensive apoptosis in mice treated with
in the prostate of nude mice are resistant to systemic imatinib and paclitaxel.
administration of paclitaxel. Targeting the phosphorylated The PDGF-R has been shown to regulate the intersti-
PDGF-R on tumor-associated endothelial cells leads to tial hypertension and transcapillary transport of mole-
regression of the multidrug resistant prostate cancer and cules [108], and the administration of imatinib to mice
inhibition of lymph node metastasis. bearing tumors can increase the concentration of a
As stated previously, regardless of sensitivity or resis- systemically administered chemotherapeutic drug within
tance to hormones or chemotherapeutic drugs, all tumor a tumor by twofold to threefold [201–203]. Since the
cells are dependent on a viable vasculature for growth PC-3MM2-MDR cells were >50-fold more resistant to
and survival [56, 62, 123]. Tumor cells interact with paclitaxel even in the presence of imatinib, the in vivo
host factors in the microenvironment to induce growth therapeutic response of the PC-3MM2-MDR bone
and expansion of vasculature [70, 72, 121]. We have tumors to imatinib and more so to imatinib plus pacli-
reported [126–128] that PDGF produced by prostate taxel could not have been due to changes in interstitial
cancer cells growing adjacent to bone can increases and fluid pressure. Endothelial cells in normal tissues rarely
activate the expression of PDGF-R on tumor-associated divide, whereas 2–3% of endothelial cells in prostate
endothelial cells by a paracrine mechanism. The systemic cancer divide daily [4, 48]. These dividing endothelial
administration of imatinib inhibited the phosphoryla- cells should be sensitive to anticycling drugs such as
tion of PDGF-R (but not EGF-R) [128], and the combi- paclitaxel. As stated above, the first wave of apoptosis
nation of imatinib and paclitaxel induced apoptosis of in bone tumors from mice treated with imatinib and
34 The pathogenesis of cancer metastasis: relevance to therapy

paclitaxel for only 2 weeks occurred in tumor-associated using homogeneous therapy. In other words, it is unlikely
endothelial cells, followed by apoptosis of tumor cells that therapy of cancer in general and metastasis in
and ultimately necrosis. By the fourth week of treatment particular can be accomplished using a single modality.
with imatinib and paclitaxel or imatinib alone, concurrent Second, cancer is a chronic disease. A chronic disease
apoptosis of tumor cells and tumor-associated endo- must be treated chronically, i.e., by management. Third,
thelial cells was observed. Without paclitaxel, imatinib cancer is the disease of the “seed” and the “soil”. Since
may produce therapeutic effects by the blockade of the outcome of metastasis depends on multiple interac-
PDGF-R, which serves as a survival factor [142, 143]. tions of metastatic cells with homeostatic mechanisms
Therefore, targeting the PDGF-R on tumor-associated in the organ microenvironment, therapy for metastasis
endothelial cells by imatinib and paclitaxel can produce should be targeted not only against tumor cells, but also
therapeutic results in multidrug resistant human prostate against the homeostatic factors that helps metastatic
cancer experimental bone metastasis. cells grow and survive.

Conclusions References
1. Arteaga CL. The epidermal growth factor receptor: from mutant
A current definition of the seed and soil hypothesis oncogene in nonhuman cancers to therapeutic target in human
consists of three principles. First, neoplasms are biologi- neoplasia. J Clin Oncol 2001; 19: 32s–40s.
cally heterogeneous and contain subpopulations of cells 2. Atlas I, Mendelsohn J, Baselga J, Fair WR, Masui H, Kumar R.
with different properties. Second, the process of metas- Growth regulation of human renal carcinoma cells: role of trans-
tasis is selective for cells that must succeed in all the forming growth factor α. Cancer Res 1992; 52: 3335–9.
3. Auerbach W, Auerbach R. Angiogenesis inhibition: a review.
steps of this complex process. Although some of the Pharmacol Ther 1994; 63: 265–311.
steps in this process contain stochastic elements, as a 4. Augustin HG, Kmeta J, Alves F, et al. Quantitating angiogenesis
whole, metastasis favors the survival and growth of a and assessing the causal relationship between angiogenesis and
few subpopulations of cells that preexist within the tumorigenesis: problems and progress. Ann Hematol 2002;
81(Suppl 2): 68–72.
parent neoplasm. Thus, metastases can have a clonal
5. Baker CH, Fidler IJ. Tumor microenvironment and angiogenesis.
origin, and different metastases can originate from the In: Davis DW, Herbst RS, Abbruzzese JL, eds. Antiangiogenic
proliferation of different single cells. Third, the out- cancer therapy. Boca Raton, FL: CRC Press, 2008: 131–47.
come of metastasis depends on multiple interactions 6. Baker CH, Kedar D, McCarty MF, Tsan R, Weber KL, Bucana CD,
(“cross-talk”) of metastatic cells with homeostatic Fidler IJ. Blockade of epidermal growth factor receptor signaling
on tumor cells and tumor-associated endothelial cells for therapy
mechanisms, which the tumor cells can exploit. A con-
of human carcinomas. Am J Pathol 2002; 161: 929–38.
vincing example of this principle is the establishment of 7. Baker CH, Solorzano CC, Fidler IJ. Blockade of the vascular
tumor-associated vasculature. endothelial growth factor receptor and epidermal growth factor
Tumor growth and metastasis depend on an adequate receptor signaling for therapy of metastatic human pancreatic can-
blood supply. The extent of angiogenesis within and cer. Cancer Res 2002; 62: 1996–2003.
8. Bancroft CC, Chen Z, Yeh J, Sunwoo JB, Yeh NT, Jackson S,
around tumors is regulated by the balance between Jackson C, Van Waes C. Effects of pharmacologic antagonists of
proangiogenic and antiangiogenic molecules. The cross- epidermal growth factor receptor, PI3K and MEK signal kinases
talk between tumor cells, normal cells, leukocytes, and on NF-kappaB and AP-1 activation and IL-8 and VEGF expres-
stromal cells occurs through growth factors, growth sion in human head and neck squamous cell carcinoma lines. Int J
factor receptors, and cytokine signaling, all of which are Cancer 2002; 99: 538–48.
9. Barrier A, Boelle PY, Roser F, et al. Stage II colon cancer prognosis
released and act on cells within the tumor microenviron- prediction by tumor gene expression profiling. J Clin Oncol 2006;
ment. These interactions enhance the establishment of 24: 4685–91.
angiogenesis that in turn regulates the proliferation and 10. Beer DG, Kardia SL, Huang CC, et al. Gene expression profiles
survival of metastatic cells. Interruption of one or more predict survival of patients with lung adenocarcinoma. Nat Med
2002; 8: 816–24.
of these interactions can lead to the inhibition or regres-
11. Beer T, Raghavan D. Chemotherapy for hormone refractory prostate
sion of metastasis. Because the growth of primary cancer: beauty is in the eye of the beholder. Prostate 2000; 45:
tumors is often controlled with surgery and/or irradia- 183–93.
tion, antiangiogenic agents may be most beneficial to 12. Benjamin LE, Hemo I, Keshet E. A plasticity window for blood
prevent widespread and metastatic disease. To succeed, vessel remodeling is defined by pericyte coverage of the preformed
endothelial network and is regulated by PDGF-B and VEGF.
several principles must be considered. First, it is essen-
Development 1998; 125: 1591–8.
tial to remember that all neoplasms are biologically 13. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic
heterogeneous. One cannot treat a heterogeneous disease switch. Nat Rev Cancer 2003; 3: 401–10.
Sun-Jin Kim et al. 35

14. Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D. 34. Cole SP, Bhardwaj G, Gerlach JH, et al. Overexpression of a trans-
Benefits of targeting both pericytes and endothelial cells in the tumor porter gene in a multidrug-resistant human lung cancer cell line.
vasculature with kinase inhibitors. J Clin Invest 2003; 111: 1287–95. Science 1992; 258: 1650–4.
15. Bergsten E, Uutela M, Li X, Pietras K, Ostman A, Heldikn CH, 35. Colombel M, Symmans F, Gil S, et al. Detection of the apoptosis-
Alitalo K, Eriksson U. PDGF-D is a specific, protease-activated suppressing oncoprotein Bcl-2 in hormone-refractory human pros-
ligand for the PDGF beta-receptor. Nat Cell Biol 2001; 3: 512–6. tate cancers. Am J Pathol 1993; 143: 390–400.
16. Black MM, Freeman C, Mork T, Harvei S, Cutler SJ. Prognostic 36. Cox CE, Pendas S, Cox JM, et al. Guidelines for sentinel node
significance of microscopic structure of gastric carcinomas and biopsy and lymphatic mapping of patients with breast cancer. Ann
their regional lymph nodes. Cancer 1971; 27: 703–10. Surg 1998; 227: 645–53.
17. Bogaerts J, Cardoso F, Buyse M, et al. Gene signature evaluation 37. Debnath J, Brugge JS. Modeling glandular epithelial cancers in
as a prognostic tool: challenges in the design of the MINDACT three-dimensional cultures. Nat Rev Cancer 2005; 5(9): 675–88.
trial (Review). Nat Clin Pract Oncol 2006; 3: 540–51. 38. Ding A, Hwang S, Schwab R. Effect of aging on murine mac-
18. Bradley G, Juranka PE, Ling V. Mechanism of multidrug resis- rophages: diminished response to IFN-γ for enhanced oxidative
tance. Biochim Biophys Acta 1988; 984: 87–128. metabolism. J Immunol 1994; 153: 2146–52.
19. Bröcker EB, Reckenfeld C, Hamm H, Ruiter DJ, Sorg C. Macrophages 39. Dinjens WN, van der Weiden MM, Schroeder FH, Bosman FT,
in melanocytic naevi. Arch Dermatol Res 1992; 284: 127–31. Trapman J. Frequency and characterization of p53 mutations in
20. Bruns CJ, Harbison MT, David DW, et al. Epidermal growth factor primary and metastatic human prostate cancer. Int J Cancer 1994;
receptor blockade with C225 plus gemcitabine results in regres- 56: 630–3.
sion of human pancreatic carcinoma growing orthotopically in 40. DiPietro L, Polverini PJ. Angiogenic macrophages produce the
nude mice by antiangiogenic mechanisms. Clin Cancer Res 2000; angiogenic factor thrombospondin 1. Am J Pathol 1993; 143:
6: 1936–48. 678–84.
21. Bruns CJ, Solorzano CC, Harbison MT, et al. Blockade of epider- 41. Donelli MG, Russo R, Garattini S. Selective chemotherapy in rela-
mal growth factor receptor signaling by a novel tyrosine kinase tion to the site of tumor transplantation. Int J Cancer 1967; 2:
inhibitor leads to apoptosis of endothelial cells and therapy of 421–4.
human pancreatic carcinoma. Cancer Res 2000; 60: 2926–35. 42. Dong Z, Kumar R, Yang X, Fidler IJ. Macrophage-derived metal-
22. Buchdunger E, Cioffi CL, Law N, et al. Abl protein-tyrosine kinase loelastase is responsible for the generation of angiostatin in Lewis
inhibitor STI571 inhibits in vitro signal transduction mediated by lung carcinoma. Cell 1997; 88: 801–10.
c-kit and platelet-derived growth factor receptors. J Pharmacol 43. Dong Z, Radinsky R, Fan D, et al. Organ-specific modulation of
Exp Ther 2000; 295: 139–45. steady-state mdr-1 gene expression and drug resistance in murine
23. Busby JE, Kim SJ, Yazici S, et al. Therapy of multidrug resistant colon cancer cells. J Natl Cancer Inst 1994; 86: 913–20.
human prostate tumors in the prostate of nude mice by simultane- 44. Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective
ous targeting of the epidermal growth factor receptor and vascular inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl posi-
endothelial growth factor receptor on tumor-associated endothelial tive cells. Nat Med 1996; 2: 561–6.
cells. Prostate 2006; 66: 1788–98. 45. Dvorak HF. Tumors: wounds that do not heal: similarities between
24. Busse D, Yakes FM, Lenferink AE, Arteaga CL. Tyrosine kinase tumor stroma generation and wound healing. N Engl J Med 1986;
inhibitors: rationale, mechanisms of action, and implications for 315: 1650–9.
drug resistance. Semin Oncol 2001; 28: 47–55. 46. Dvorak HF, Brown LF, Detmar M, Dvorak AM. Vascular permea-
25. Byers RM. Modified neck dissection: a study of 967 cases from bility factor/vascular endothelial growth factor, microvascular
1970 to 1980. Am J Surg 1985; 150: 414–21. hyperpermeability, and angiogenesis. Am J Pathol 1995; 146:
26. Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. 1029–39.
Nature 2000; 407: 249–57. 47. Dvorak HF, Seneger DR, Dvorak AM. Fibrin as a component of
27. Carr I. Lymphatic metastasis. Cancer Metastasis Rev 1983; 22: the tumor stroma: origins and biological significance. Cancer
307–19. Metastasis Rev 1983; 2: 41–75.
28. Chang JC, Wooten EC, Tsimelzon A, et al. Gene expression profiling 48. Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH,
for the prediction of therapeutic response to docetaxel in patients Augustin HG. Heterogeneity of angiogenesis and blood vessel
with breast cancer. Lancet 2003; 362: 362–9. maturation in human tumors: implications for antiangiogenic
29. Chen CN, Lin JJ, Chen JJ, et al. Gene expression profile predicts tumor therapies. Cancer Res 2000; 60: 1388–93.
patient survival of gastric cancer after surgical resection. J Clin 49. Erber R, Thurnher A, Katsen AD, et al. Combined inhibition of
Oncol 2005; 23: 7286–95. VEGF and PDGF signaling enforces tumor vessel regression by
30. Ciardiello F, Bianco R, Caputo R, et al. Antitumor activity of interfering with pericyte-mediated endothelial cell survival mech-
ZD6474, a vascular endothelial growth factor receptor tyrosine anisms. FASEB J 2004; 18: 338–40.
kinase inhibitor, in human cancer cells with acquired resistance to 50. Ewing J. Neoplastic diseases. 6th edt. Philadelphia, PA: WB
anti-epidermal growth factor receptor therapy. Clin Cancer Res Saunders, 1928.
2004; 10: 784–93. 51. Fabra A, Nakajima M, Bucana CD, Fidler IJ. Modulation of the
31. Cifone MA, Fidler IJ. Increasing metastatic potential is associated invasive phenotype of human colon carcinoma cells by fibroblasts
with increasing genetic instability of clones isolated from murine from orthotopic or ectopic organs of nude mice. Differentiation
neoplasms. Proc Natl Acad Sci USA 1982; 78: 6949–52. 1992; 52: 101–10.
32. Clark WH, Elder DE, Guerry D, Epstein ME, Green MH, 52. Ferrara N, Alitalo K. Clinical applications of angiogenic growth
VanHorn M. A study of tumor progression: the precursor lesions factors and their inhibitors. Nat Med 1999; 5: 1359–64.
of superficial spreading and nodular melanoma. Hum Pathol 53. Ferrara N, Davis-Smyth T. The biology of vascular endothelial
1984; 15: 1147–62. growth factor. Endocrinol Rev 1997; 18: 4–25.
33. Clark WH, Elder DE, VanHorn M. The biologic forms of malig- 54. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its
nant melanoma. Hum Pathol 1986; 17: 443–69. receptors. Nat Med 2003; 9: 669–76.
36 The pathogenesis of cancer metastasis: relevance to therapy

55. Fidler IJ. Angiogenic heterogeneity: regulation of neoplastic 77. Fidler IJ, Yano S, Zhang R-D, Fujimaki T, Bucana CD. The seed
angiogenesis by the organ microenvironment (Editorial). J Natl and soil hypothesis: vascularization and brain metastases (Personal
Cancer Inst 2001; 93: 1040–1. View). Lancet Oncol 2002; 3: 53–7.
56. Fidler IJ. Biology of cancer metastasis. In: Abeloff MD, Armitage 78. Folkman J. How is blood vessel growth regulated in normal and
JO, Niederhuber JE, Kastan MB, McKenna WG, eds. Clinical neoplastic tissue? G H A Clowes Memorial Award Lecture. Cancer
oncology, 3rd edt. Philadelphia, PA: Elsevier Sciencem 2004; Res 1986; 46: 467–73.
59–79. 79. Folkman J. The role of angiogenesis in tumor growth. Semin
57. Fidler IJ. Critical determinants of metastasis. Semin Cancer Biol Cancer Biol 1992; 3: 65–71.
2002; 12: 89–96. 80. Folkman J, Klagsburn M. Angiogenic factors. Science 1987; 235:
58. Fidler IJ. Critical factors in the biology of human cancer metasta- 444–7.
sis: twenty-eighth G H A Clowes Memorial Award Lecture. Cancer 81. Foulds L. The experimental study of tumor progression: a review.
Res 1990; 50: 6130–8. Cancer Res 1954; 14: 327–39.
59. Fidler IJ. Lymphocytes are not only immunocytes (Guest 82. Fracasso PM, Westervelt P, Fears CL, et al. Phase I study of pacli-
Editorial). Biomedicine 1980; 32: 1–3. taxel in combination with a multidrug resistance modulator, PSC
60. Fidler IJ. Metastasis: quantitative analysis of distribution and fate 833 (Valspodar), in refractory malignancies. J Clin Oncol 2000;
of tumor emboli labeled with 125I-5-iodo-2´-deoxyuridine. J Natl 18: 1124–34.
Cancer Inst 1970; 45: 773–82. 83. Freeman MR, Schneck FX, Gagnon ML, et al. Peripheral blood
61. Fidler IJ. Orthotopic implantation of human colon carcinomas into T-lymphocytes and lymphocytes infiltrating human cancers
nude mice provides a valuable model for the biology and therapy express vascular endothelial growth factor: a potential role for T
of cancer metastasis. Cancer Metastasis Rev 1991; 10: 229–43. cells in angiogenesis. Cancer Res 1995; 55: 4140–5.
62. Fidler IJ. The organ microenvironment and cancer metastasis. 84. Furuhashi M, Sjoblom T, Abramsson A, et al. Platelet-derived
Differentiation 2002; 70: 498–505. growth factor production by B16 melanoma cells leads to increased
63. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ pericyte abundance in tumors and an associated increase in tumor
hypothesis revisited (Timeline). Nat Rev Cancer 2003; 3: 453–8. growth rate. Cancer Res 2004; 64: 2725–33.
64. Fidler IJ. Rationale and methods for the use of nude mice to study 85. Gerber HP, Dixit V, Ferrara N. Vascular endothelial growth factor
the biology and therapy of human cancer metastasis. Cancer induces expression of the antiapoptotic proteins Bcl-2 and A1 in
Metastasis Rev 1986; 5: 29–49. vascular endothelial cells. J Biol Chem 1998; 273: 13313–6.
65. Fidler IJ. “Seed and soil” revisited: contribution of the organ 86. Gerber HP, McMurtrey A, Kowalski J, et al. Vascular endothelial
microenvironment to cancer metastasis. In: Brodt P, ed. Surgical growth factor regulates endothelial cell survival through the phos-
oncology clinics of NA: cancer metastasis: biological and clinical phatidylinositol 3´-kinase/Akt signal transduction pathway: require-
aspects. Philadelphia, PA: WB Saunders, 2001: 257–69. ment for Flk-1/KDR activation. J Biol Chem 1998; 273: 30336–43.
66. Fidler IJ. Selection of successive tumor lines for metastasis. Nature 87. Gerhardt H, Betsholtz C. Endothelial-pericyte interactions in
(New Biol) 1973; 242: 148–9. angiogenesis. Cell Tissue Res 2003; 314: 15–23.
67. Fidler IJ, Bucana C. Mechanism of tumor cell resistance to lysis by 88. Gerritsen ME. Functional heterogeneity of vascular endothelial
syngeneic lymphocytes. Cancer Res 1977; 37: 3945–56. cells. Biochem Pharmacol 1987; 36: 2701–11.
68. Fidler IJ, Gersten DM, Kripke ML. Influence of the immune status 89. Ghadimi BM, Grade M, Difilippantonio MJ, et al. Effectiveness of
on the metastasis of three murine fibrosarcomas of different immu- gene expression profiling for response prediction of rectal adeno-
nogenicities. Cancer Res 1979; 39: 3816–21. carcinomas to preoperative chemoradiotherapy. J Clin Oncol 2005;
69. Fidler IJ, Gruys E, Cifone MA, Barnes Z, Bucana C. Demonstration 23: 1826–38.
of multiple phenotypic diversity in a murine melanoma of recent 90. Gisselsson D, Pettersson L, Hoglund M, et al. Chromosomal
origin. J Natl Cancer Inst 1981; 67: 947–56. breakage-fusion-bridge events cause genetic intratumor heteroge-
70. Fidler IJ, Kim SJ, Langley RR. The role of the organ microenvi- neity. Proc Natl Acad Sci USA 2000; 97: 5357–62.
ronment in the biology and therapy of cancer metastasis. J Cell 91. Gohji K, Fidler IJ, Tsan R, Radinsky R, von Eschenbach AC,
Biochem 2007; 101: 927–36. Tsuruo T, Nakajima M. Human recombinant interferons-beta and
71. Fidler IJ, Kripke ML. Metastasis results from pre-existing variant -gamma decrease gelatinase production and invasion by human
cells within a malignant tumor. Science 1977; 197: 893–5. KG-2 renal carcinoma cells. Int J Cancer 1994; 58: 380–4.
72. Fidler IJ, Langley RR, Kerbel RS, Ellis LM. Biology of cancer: 92 Goidl E, ed. Aging and the immune response: cellular and humoral
angiogenesis. In: DeVita VT Jr, Hellman S, Rosenberg SA, eds. aspects. New York: Marcel Dekker, 1987.
Cancer: principles and practice of oncology, 7th edt. Philadelphia, 93. Gorunova L, Hoglund M, Andren-Sandberg A, et al. Cytogenetic
PA: Lippincott & Wilkins, 2005; 129–37. analysis of pancreatic carcinomas: intratumor heterogeneity and
73. Fidler IJ, Nicolson GL. Organ selectivity for implantation survival nonrandom pattern of chromosome aberrations. Genes Chromosomes
and growth of B16 melanoma variant tumor lines. J Natl Cancer Cancer 1998; 23: 81–99.
Inst 1976; 57: 1199–202. 94. Gottesman MM, Pastan I. Biochemistry of multidrug resistance
74. Fidler IJ, Poste G. The cellular heterogeneity of malignant neoplasms: mediated by the multidrug transporter. Annu Rev Biochem 1993;
implications for adjuvant chemotherapy. Semin Oncol 1985; 12: 62: 385–427.
207–21. 95. Griffin CA, Hruban RH, Morsberger LA, et al. Consistent chromo-
75. Fidler IJ, Talmadge JE. Evidence that intravenously derived some abnormalities in adenocarcinoma of the pancreas. Cancer
murine pulmonary metastases can originate from the expansion of Res 1995; 55: 2394–9.
a single tumor cell. Cancer Res 1986; 46: 5167–71. 96. Guo P, Hu B, Gu W, et al. Platelet-derived growth factor-β
76. Fidler IJ, Wilmanns C, Staroselsky A, Radinsky R, Dong Z, Fan D. enhances glioma angiogenesis by stimulating vascular endothelial
Modulation of tumor cell response to chemotherapy by the organ growth factor expression in tumor endothelia and by promoting
environment. Cancer Metastasis Rev 1994; 13: 209–22. pericyte recruitment. Am J Pathol 2003; 162: 1083–93.
Sun-Jin Kim et al. 37

97. Gutman M, Singh RK, Xie K, Bucana CD, Fidler IJ. Regulation 120. Kadowaki T, Shiozaki H, Inoue M, et al. E-cadherin and α-catenin
of IL-8 expression in human melanoma cells by the organ envi- expression in human esophageal cancer. Cancer Res 1994; 54:
ronment. Cancer Res 1995; 55: 2470–5. 291–6.
98. Gutman, M, Singh RK, Yoon S, Xie K, Bucana CD, Fidler IJ. 121. Kerbel RS. Antiangiogenic therapy: a universal chemosensitiza-
Leukocyte-induced angiogenesis and subcutaneous growth of tion strategy for cancer? Science 2006; 312: 1171–5.
B16 melanoma. Cancer Biother 1994; 9: 163–70. 122. Kerbel RS. Inhibition of tumor angiogenesis as a strategy to cir-
99. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; cumvent acquired resistance to anticancer therapeutic agents.
100: 57–70. BioEssays 1991; 13: 31–6.
100. Harris AL. Hypoxia – a key regulatory factor in tumour growth. 123. Kerbel RS, Folkman J. Clinical translation of angiogenesis inhib-
Nat Rev Cancer 2002; 2: 38–47. itors. Nat Rev Cancer 2002; 2: 727–39.
101. Hart IR, Fidler IJ. Role of organ selectivity in the determination 124. Killion JJ, Radinsky R, Fidler IJ. Orthotopic models are neces-
of metastatic patterns of B16 melanoma. Cancer Res 1980; 40: sary to predict therapy of transplantable tumors in mice. Cancer
2281–7. Metastasis Rev 1999; 17: 279–84.
102. Hedley D, Pintilie M, Woo J, et al. Carbonic anhydrase IX 125. Kim SJ, Uehara H, Karashima T, Shepherd DL, Killion JJ, Fidler
expression, hypoxia, and prognosis in patients with uterine cervi- IJ. Blockade of epidermal growth factor receptor signaling in
cal carcinomas. Clin Cancer Res 2003; 9: 5666–74. tumor cells and tumor-associated endothelial cells for therapy of
103. Heldin CH, Ostman A, Ronnstrand L. Signal transduction via androgen-independent human prostate cancer growing in the
platelet-derived growth factor receptors. Biochim Biophys Acta bone of nude mice. Clin Cancer Res 2003; 9: 1200–10.
1998; 1378: F79–F113. 126. Kim SJ, Uehara H, Yazici S, et al. Targeting platelet-derived
104. Heldin CH, Westermark B. Mechanism of action and in vivo growth factor receptor on endothelial cells of multidrug resistant
role of platelet-derived growth factor. Physiol Rev 1999; 79: prostate cancer. J Natl Cancer Inst 2006; 98: 783–93.
1283–316. 127. Kim SJ, Uehara H, Yazici S, et al. Modulation of bone microen-
105. Herbst RS, Yano S, Kuniyasu H, et al. Differential expression of vironment with zoledronate enhances the therapeutic effects of
E-cadherin and type IV collagenase genes predicts outcome in STI571 and paclitaxel against experimental bone metastasis of
patients with stage I non-small cell lung carcinoma. Clin Cancer human prostate cancer. Cancer Res 2005; 65: 3707–15.
Res 2000; 6: 790–7. 128. Kim SJ, Uehara H, Yazici S, et al. Simultaneous blockade of plate-
106. Herlyn M. Human melanoma: development and progression. let-derived growth factor-receptor and epidermal growth factor-
Cancer Metastasis Rev 1990; 9: 101–29. receptor signaling and systemic administration of Paclitaxel as
107. Herlyn M, Clark WH, Rodeck U, Mancianti ML, Jambrosic J, therapy for human prostate cancer metastasis in bone of nude
Koprowski H. Biology of tumor progression in human melano- mice. Cancer Res 2004; 64: 4201–8.
cytes. Lab Investigation 1987; 56: 461–74. 129. Kitadai Y, Ellis LM, Takahashi Y, et al. Multiparametric in situ
108. Heuchel R, Berg A, Tallqauist M, et al. Platelet-derived growth mRNA hybridization analysis to detect metastasis-related genes
factor β receptor regulates interstitial fluid homeostasis through in surgical specimens of human colon carcinoma. Clin Cancer
phosphatidylinositol-3′ kinase signaling. Proc Natl Acad Sci Res 1995; 1: 1095–102.
USA 1999; 86: 11410–5. 130. Kitadai Y, Ellis LM, Tucker SL, et al. Multiparametric in situ
109. Hewitt RE, Powe DG, Carter GI, Turner DR. Desmoplasia and its mRNA hybridization analysis to predict disease recurrence in
relevance to colorectal tumour invasion. Int J Cancer 1993; 53: patients with colon carcinoma. Am J Pathol 1996; 149: 1541–51.
62–9. 131. Kitadai Y, Sasaki T, Kuwai T, et al. Expression of activated plate-
110. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth let-derived growth factor receptor in stromal cells of human
factor pathway in tumor-growth and angiogenesis. J Clin Oncol colon carcinomas is associated with metastatic potential. Int J
2005; 23: 1011–27. Cancer 2006; 119: 2567–74.
111. Hofmockel G, Riess S, Bassukas ID, Dammrich J. Epidermal 132. Kitadai Y, Sasaki T, Kuwai T, et al. Targeting the expression of
growth factor family and renal cell carcinoma: expression and platelet-derived growth factor receptor by reactive stroma inhib-
prognostic impact. Eur Urol 1997; 4: 478–84. its growth and metastasis of human colon carcinoma. Am J Pathol
112. Huang S, Van Arsdall M, Tedjarati S, et al. Contributions of stromal 2006; 169: 2054–65.
metalloproteinase-9 to angiogenesis and growth of human ovarian 133. Kreisle RA, Stebler BA, Ershler WB. Effect of host age on tumor-
carcinoma in mice. J Natl Cancer Inst 2002; 94: 1134–42. associated angiogenesis in mice. J Natl Cancer Inst 1990; 82: 44–7.
113. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus 134. Kripke ML. Speculation on the role of ultraviolet radiation in the
irinotecan, fluorouracil, and leucovorin for metastatic colorectal development of malignant melanoma. J Natl Cancer Inst 1979;
cancer. N Engl J Med 2004; 350: 2335–42. 63: 541–5.
114. Hwang RF, Yokoi K, Bucana CD, et al. Inhibition of platelet- 135. Kripke ML. Immunologic mechanisms in UV radiation carcino-
derived growth factor receptor phosphorylation by STI571 genesis. Adv Cancer Res 1981; 34: 69–105.
(Gleevec) reduces growth and metastasis of human pancreatic 136. Kripke ML, Fidler IJ. Enhanced experimental metastasis of
carcinoma in an orthotopic nude mouse model. Clin Cancer Res UV-induced fibrosarcomas in UV-irradiated syngeneic mice.
2003; 9: 6534–44. Cancer Res 1980; 40: 625–9.
115. Iizuka N, Oka M, Yamada-Okabe H, et al. Oligonucleotide 137. Kumar R, Dong Z, Fidler IJ. Differential regulation of metal-
microarray for prediction of early intrahepatic recurrence of loelastase activity in murine peritoneal macrophages by GM-CSF
hepatocellular carcinoma after curative resection. Lancet 2003; and M-CSF. J Immunol 1996; 157: 5104–11.
361: 923–9. 138. Kuniyasu H, Ellis LM, Evans DB, et al. Relative expression of
116. Ikeda N, Adachi M, Taki T, et al. Prognostic significance of E-cadherin and type IV collagenase genes predicts disease out-
angiogenesis in human pancreatic cancer. Br J Cancer 1999; 79: come in patients with resectable pancreatic carcinoma. Clin
1553–63. Cancer Res 1999; 5: 25–33.
117. Ito M, Yoshida K, Kyo E, et al. Expression of several growth fac- 139. Kuniyasu H, Troncoso P, Johnston D, et al. Relative expression
tors and their receptor genes in human colon carcinomas. of type IV collagenase, E-cadherin, and vascular endothelial
Virchows Arch B Cell Pathol Incl Mol Pathol 1990; 59: 173–8. growth factor/vascular permeability factor in prostatectomy
118. Jain R. Normalization of tumor vasculature: an emerging concept specimens distinguishes organ-confined from pathologically
in antiangiogenic therapy. Science 2005; 307: 58–62. advanced prostate cancers. Clin Cancer Res 2000; 6: 2295–308.
119. Joseph E, Brobeil A, Glass F, et al. Results of complete lymph 140. Kuwai T, Kitadai Y, Tanaka S, et al. Expression of hypoxia-
node dissection in 83 melanoma patients with positive sentinel inducible factor-1alpha is associated with tumor vascularization
nodes. Ann Surg Oncol 1998; 5: 119–25. in human colorectal carcinoma. Int J Cancer 2003; 105: 176–81.
38 The pathogenesis of cancer metastasis: relevance to therapy

141. Kuwai T, Nakamura T, Kim SJ, et al. Intratumoral heterogeneity susceptibility to the host immune response. Cancer Immunol
for expression of tyrosine kinase growth factor receptors in Immunother 2005; 54: 328–36.
human colon cancer surgical specimens and orthotopic tumors. 163. Mattern J, Bak M, Volm M. Intrinsic and acquired multidrug
Am J Pathol 2008; 172: 358–66. resistance in human lung carcinomas grown in nude mice.
142. Langley RR, Fan D, Tsan RZ, et al. Activation of the platelet- Anticancer Res 1990; 10: 177–80.
derived growth factor receptor enhances survival of murine bone 164. Menininger IJ, Zetter BR. Mast cells and angiogenesis. Semin
endothelial cells. Cancer Res (Adv in Brief) 2004; 64: 3727–30. Cancer Biol 1992; 3: 73–9.
143. Langley RR, Ramirez KM, Tsan RZ, Van Arsdall M, Nilsson 165. Micke P, Ostman A. Tumour-stroma interaction: cancer-associ-
MB, Fidler IJ. Tissue-specific microvascular endothelial cell ated fibroblasts as novel targets in anti-cancer therapy? Lung
lines from H-2kb-tsA58 mice for studies of angiogenesis and Cancer 2004; 45(Suppl 2): S163–75.
metastasis. Cancer Res 2003; 63: 2971–6. 166. Mickisch GH, Noordzij MA, vd Gaast A, et al. Dexverapamil to
144. LaRochelle WJ, Jeffers M, McDonald WF, et al. PDGF-D, a new modulate vinblastine resistance in metastatic renal cell carcinoma.
protease-activated growth factor. Nat Cell Biol 2001; 3: 517–21. J Cancer Res Clin Oncol 1995; 121(Suppl 3): R11–6.
145. Lazard D, Sastre X, Frid MG, Glukhova MA, Thiery JP, 167. Mickisch GH, Pai LH, Gottesman MM, Pastan I. Monoclonal
Koteliansky VE. Expression of smooth muscle-specific pro- antibody MRK16 reverses the multidrug resistance of multidrug-
teins in myoepithelium and stromal myofibroblasts of normal resistant transgenic mice. Cancer Res 1992; 52: 4427–32.
and malignant human breast tissue. Proc Natl Acad Sci USA 168. Mickisch GH, Rahman A, Pastan I, Gottesman MM. Increased
1993; 90: 999–1003. effectiveness of liposome-encapsulated doxorubicin in multi-
146. Lee WH, Morton RA, Epstein JI, et al. Cytidine methylation of drug-resistant-transgenic mice compared with free doxorubicin.
regulatory sequences near the pi-class glutathione S-transferase J Natl Cancer Inst 1992; 84: 804–5.
gene accompanies human prostatic carcinogenesis. Proc Natl 169. Miguez M, Davel L, deLustig ES. Lymphocyte-induced angio-
Acad Sci USA 1994; 91: 11733–7. genesis: correlation with the metastatic incidence of two murine
147. Lehnert M. Clinical multidrug resistance: a multifactorial prob- mammary adenocarcinomas. Inv Metastasis 1986; 6: 313–20.
lem. Eur J Cancer 1996; 32A: 912–20. 170. Moch H, Sauter G, Buchholz N, et al. Epidermal growth factor
148. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. receptor expression is associated with rapid tumor cell proli-
Vascular endothelial growth factor is a secreted angiogenic mito- feration and renal cell carcinoma. Hum Pathol 1997; 11:
gen. Science 1989; 246: 1306–9. 1255–9.
149. Li X, Ponten A, Aase K, et al. PDGF-C is a new protease-acti- 171. Morikawa K, Walker SM, Jessup JM, Fidler IJ. In vivo selection
vated ligand for the PDGF alpha-receptor. Nat Cell Biol 2000; 2: of highly metastatic cells from surgical specimens of different
302–9. primary human colon carcinoma implanted into nude mice.
150. Lieubeau B, Garrigue L, Barbieux I, Meflah K, Gregoire M. The Cancer Res 1988; 48: 1943–8.
role of transforming growth factor beta 1 in the fibroblastic reac- 172. Morikawa K, Walker SM, Nakajima M, Pathak S, Jessup JM,
tion associated with rat colorectal tumor development. Cancer Fidler IJ. The influence of organ environment on the growth, selec-
Res 1994; 54: 6526–32. tion, and metastasis of human colon cancer cells in nude mice.
151. Lindmark G, Sundberg C, Glimelius B, Pahlman L, Rubin K, Cancer Res 1988; 48: 6863–71.
Gerdin B. Stromal expression of platelet-derived growth factor 173. Moroni M, Veronese S, Benvenuti S, et al. Gene copy number for
beta-receptor and platelet-derived growth factor B-chain in col- epidermal growth factor receptor (EGFR) and clinical response
orectal cancer. Lab Invest 1993; 69: 682–9. to anti-EGFR treatment in colorectal cancer: a cohort study.
152. Lingen MW, Polverini PJ, Bouck N. Inhibition of squamous cell Lancet Oncol 2005; 6: 279–86.
carcinoma angiogenesis by direct interaction of retinoic acid with 174. Morrow CS, Cowan KH. Glutathione S-transferase and drug
endothelial cells. Lab Invest 1996; 74: 476–83. resistance. Cancer Cells 1990; 2: 15–22.
153. Lingen MW, Polverini PJ, Bouck N. Retinoic acid induces cells 175. Morton DL, Wen D-R, Wong JH, Cochran AJ. Technical details
cultured from oral squamous cell carcinomas to become antian- of intraoperative lymphatic mapping for early stage melanoma.
giogenic. Am J Pathol 1996; 149: 247–58. Arch Surg 1992; 127: 392–9.
154. Liotta LA. Tumor invasion and metastases – Role of the extracel- 176. Motzer RJ, Lyn P, Fischer P, et al. Phase I/II trial of dexverapamil
lular matrix: Rhoads memorial award lecture. Cancer Res 1986; plus vinblastine for patients with advanced renal cell carcinoma.
46: 1–7. J Clin Oncol 1995; 13: 1958–65.
155. Liotta LA, Kohn EC. The microenvironment of the tumor-host 177. Nakajima M, Irimura T, Nicolson GL. Heparanases and tumor
invasion field. Nature 2001; 375–9. metastasis. J Cell Biochem 1988; 36: 157–64.
156. Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer metastasis 178. Nakajima M, Morikawa K, Fabra A, Bucana CD, Fidler IJ.
and angiogenesis: an imbalance of positive and negative regula- Influence of organ environment on extracellular matrix degrada-
tion. Cell 1991; 64: 327–36. tive activity and metastasis of human colon carcinoma cells.
157. Luria SE, Delbruck M. Mutations of bacteria from virus sensitivity J Natl Cancer Inst 1990; 82: 1890–8.
to virus resistant. Genetics 1943; 28: 491–511. 179. Nakamura T, Fidler IJ, Coombes KR. Gene expression profile of
158. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the metastatic human pancreatic cancer cells depends on the organ
epidermal growth factor receptor underlying responsiveness of microenvironment. Cancer Res 2007; 67: 139–48.
non-small-cell lung cancer to gefitinib. N Engl J Med 2004; 350: 180. Nakamura T, Kuwai T, Kitadai Y, et al. Zonal heterogeneity for
2129–39. gene expression in human pancreatic carcinoma. Cancer Res
159. Man S, Bocci G, Francia G, Green S, et al. Anti-tumor and anti- 2007; 67: 7597–604.
angiogenic effects in mice of low-dose (metronomic) cyclophos- 181. Nanus DM, Schmitz-Dräger BJ, Motzer RJ, et al. Expression of
phamide administered continuously through the drinking water. basic fibroblast growth factor in primary human renal tumors:
Cancer Res 2002; 62: 2731–5. correlation with poor survival. J Natl Cancer Inst 1994; 85:
160. Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin 1598–9.
and function of tumor-associated macrophages. Immunol Today 182. Nels V, Denzer K, Drenchahn D. Pericyte involvement in capil-
1992; 13: 265–70. lary sprouting during angiogenesis in situ. Cell Tissue Res 1992;
161. Mareel M, Vleminck K, Vermeulen S, et al. Homotypic cell-cell 270: 469–74.
adhesion molecules and tumor invasion. In: Graumann W, Drukker J, 183. Nesbit M, Herlyn M. Adhesion receptors in human melanoma
eds. Progress in histo- and cytochemistry: Histochemistry of recep- progression. Inv Metastasis 1994–1995; 14: 131–8.
tors, vol 26. Sutttgart: Fischer Verlag, 1992; 95–106. 184. Nguyen M, Watanabe H, Budson AE, Richie JP, Hayes DF,
162. Martin-Orozco E, Ferragut JA, Garcia-Penarrubia P, Ferrer- Folkman J. Elevated levels of an angiogenic peptide, basic fibro-
Montiel A. Acquisition of multidrug resistance by L1210 blast growth factor, in the urine of patients with a wide spectrum
leukemia cells decreases their tumorigenicity and enhances their of cancers. J Natl Cancer Inst 1994; 86: 356–61.
Sun-Jin Kim et al. 39

185. Nicolson GL. Metastatic tumor cell attachment and invasion populations of B16 melanoma cells. Proc Natl Acad Sci USA
assay utilizing vascular endothelial cell monolayer. J Histochem 1981; 78: 6226–30.
Cytochem 1982; 30: 214–20. 211. Ramadori G, Veit T, Schwogler S, et al. Expression of the gene of
186. Nicolson GL. Tumor cell instability, diversification, and progres- the alpha-smooth muscle-actin isoform in rat liver and in rat fat-
sion to the metastatic phenotype: from oncogene to oncofetal storing (ITO) cells. Virchows Arch B Cell Pathol Incl Mol Pathol
expression. Cancer Res 1987; 47: 1473–87. 1990; 59: 349–57.
187. Nicolson GL, Dulski KM. Organ specificity of metastatic tumor 212. Ripple M, Mulcahy RT, Wilding G. Characteristics of the gluta-
colonization is related to organ-selective growth properties of thione/glutathione-S-transferase detoxification system in mel-
malignant cells. Int J Cancer 1986; 38: 289–94. phalan resistant human prostate cancer cells. J Urol 1993; 150:
188. Nigro JM, Baker SJ, Preisinger AC, et al. Mutations in the p53 209–14.
gene occur in diverse human tumour types. Nature 1989; 342: 213. Risau W, Drexler H, Mironov V, Smits A, Siegbahn A, Funa K,
705–8. Heldin CH. Platelet-derived growth factor is angiogenic in vivo.
189. Nor JE, Polverni PJ. Role of endothelial cell survival and death Growth Factors 1992; 7: 261–6.
signals in angiogenesis. Angiogenesis 1999; 3: 101–16. 214. Rittmann-Grauer LS, Yong MA, Sanders V, Mackensen DG.
190. Nowell PC. The clonal evolution of tumor cell populations. Reversal of Vinca alkaloid resistance by anti-P-glycoprotein
Science 1976; 194: 23–8. monoclonal antibody HYB-241 in a human tumor xenograft.
191. Ohmori H, Fujii K, Sasahira T, et al. Determinants for predic- Cancer Res 1992; 52: 1810–6.
tion of malignant potential by metalloproteinase: E-cadherin 215. Rønnov-Jessen L, Petersen OW, Bissell MJ. Cellular changes
ratio in prostate core needle biopsy. Pathobiology 2006; 73: involved in conversion of normal to malignant breast: importance
98–104. of the stromal reaction. Physiol Rev 1996; 76: 69–125.
192. Ostman A. PDGF receptors-mediators of autocrine tumor growth 216. Rosenwald A, Wright G, Chan WC, et al. The use of molecular
and regulators of tumor vasculature and stroma. Cytokine Growth profiling to predict survival after chemotherapy for diffuse large-
Factor Rev 2004; 15: 275–86. B-cell lymphoma. N Engl J Med. 2002; 346: 1937–47.
193. Paget S. The distribution of secondary growths in cancer of the 217. Ruiter DJ, Bhan AK, Harrist TJ, Sober AJ, Mihm MC Jr. Major
breast. Lancet 1889; 1: 571–3. histocompatibility antigens and mononuclear inflammatory infil-
194. Pasqualini R, Arap W, McDonald DM. Probing the structural and trate in benign nevomelanocytic proli and malignant melanoma.
molecular diversity of tumor vasculature. Trends Mol Med 2002; J Immunol 1982; 129: 2808–15.
8: 563–71. 218. Ruoslahti E. Fibronectin and its α5β1 integrin receptor in malig-
195. Patel PH, Chaganti RS, Motzer RJ. Targeted therapy for meta- nancy. Inv Metastasis 1994–1995; 14: 87–94.
static renal cell carcinoma. Br J Cancer 2006; 94: 614–9. 219. Schackert G, Fidler IJ. Site-specific metastasis of mouse melano-
196. Pauli BU, Schwartz DE, Thonar EJM, Kuttner KE. Tumor inva- mas and a fibrosarcoma in the brain or the meninges of syngeneic
sion and host extracellular matrix. Cancer Metastasis Rev 1983; animals. Cancer Res 1988; 48: 3478–84.
2: 129–53. 220. Schackert G, Price JE, Zhang RD, Bucana CD, Itoh K, Fidler IJ.
197. Petrylak DP. Chemotherapy for advanced hormone refractory Regional growth of different human melanoma as meta-stases
prostate cancer. Urology 1999; 54: 30–5. in the brain of nude mice. Am J Pathol 1990; 136: 95–102.
198. Petrylak DP, MacArthur RB, O’Connor J, et al. Phase I trial of 221. Sharifi N, Gulley JL, Dahut WL. Androgen deprivation therapy
Docetaxel with estramustine in androgen-independent prostate for prostate cancer. J Am Med Assoc 2005; 294: 238–44.
cancer. J Clin Oncol 1999; 17: 958–67. 222. Shimizu S, Yamada N, Sawada T, et al. In vivo and in vitro inter-
199. Pietras K. Increasing tumor uptake of anticancer drugs with ima- actions between human colon carcinoma cells and hepatic stellate
tinib. Semin Oncol 2004; 31: 18–23. cells. Jpn J Cancer Res 2000; 91: 1285–95.
200. Pietras K, Hanahan D. A multi-targeted, metronomic, and maxi- 223. Sidky YA, Auerbach R. Lymphocyte-induced angiogenesis in
mum-tolerated dose “chemo-switch” regimen is antiangiogenic, tumor-bearing mice. Science 1976; 192: 1237–8.
producing objective responses and survival benefit in a mouse 224. Sieweke MH, Bissell MJ. The tumor-promoting effect of wound-
model of cancer. J Clin Oncol 2005; 23: 939–52. ing: a possible role for TGF-beta-induced stromal alterations.
201. Pietras K, Ostman A, Sjoquist M, et al. Inhibition of platelet- Crit Rev Oncol 1994; 5: 297–311.
derived growth factor receptors reduces interstitial hypertension 225. Sigalotti L, Fratta E, Coral S, et al. Intratumor heterogeneity of
and increases transcapillary transport in tumors. Cancer Res cancer/testis antigens expression in human cutaneous melanoma
2001; 61: 2929–34. is methylation-regulated and functionally reverted by 5-aza-2′-
202. Pietras K, Rubin K, Sjoblom T, et al. Inhibition of PDGF receptor deoxycytidine. Cancer Res 2004; 64: 9167–71.
signaling in tumor stroma enhances antitumor effect of chemo- 226. Singh RK, Bucana CD, Gutman M, Fan D, Wilson MR, Fidler IJ.
therapy. Cancer Res 2002; 62: 5476–84. Organ-site dependent expression of bFGF in human renal cell
203. Pietras K, Stumm M, Hubert M, et al. STI571 enhances the thera- carcinoma cells. Am J Pathol 1994; 145: 365–74.
peutic index of epothilone B by a tumor selective increase of drug 227. Singh RK, Gutman M, Bucana CD, Sanchez R, Llansa N, Fidler IJ.
uptake. Clin Cancer Res 2003; 9: 3779–87. Interferons alpha and beta downregulate the expression of basic
204. Polverini PJ. How the extracellular matrix and macrophages con- fibroblast growth factor in human carcinomas. Proc Natl Acad
tribute to angiogenesis-dependent diseases. Eur J Cancer 1996; Sci USA 1995; 92: 4562–6.
32A: 2430–7. 228. Singh RK, Gutman M, Radinsky R, Bucana CD, Fidler IJ.
205. Polverini PJ, Cotran PS, Gimbrone MA Jr. Activated macrophages Expression of interleukin 8 correlates with the metastatic poten-
induce vascular proliferation. Nature 1977; 269: 804–6. tial of human melanoma cells in nude mice. Cancer Res 1994; 54:
206. Polverini JJ, Leibovich SJ. Induction of neovascularization in 3242–7.
vivo and endothelial cell proliferation in vitro by tumor-associated 229. Slaton JW, Perrotte P, Inoue K, Dinney CPN, Fidler IJ. Interferon-
macrophages. Lab Invest 1984; 51: 635–42. alpha-mediated downregulation of angiogenesis-related genes
207. Pomeroy SL, Tamayo P, Gaasenbeek M, et al. Prediction of and therapy of bladder cancer are dependent on optimization of
central nervous system embryonal tumour outcome based on dose and schedule. Clin Cancer Res 1995; 2726–34.
gene expression. Nature 2002; 415: 436–42. 230. Sloane BF, Honn KV. Cysteine proteinase and metastasis. Cancer
208. Poste G. Pathogenesis of metastatic disease: implications for cur- Metastasis Rev 1984; 3: 249–65.
rent therapy and for the development of new therapeutic strate- 231. Smith DC, Esper P, Strawderman M, Redman B, Poenta KJ.
gies. Cancer Treat Rep 1986; 70: 183–99. Phase II trial of oral estramustine, oral etoposide, and intravenous
209. Poste G, Fidler IJ. The pathogenesis of cancer metastasis. Nature paclitaxel in hormone-refractory prostate cancer. J Clin Oncol
1979; 283: 139–46. 1999; 17: 1664–71.
210. Poste G, Doll J, Fidler IJ. Interactions among clonal subpopula- 232. Solorzano CC, Baker CH, Tsan R, et al. Optimization for the
tions affect stability of the metastatic phenotype in polyclonal blockade of epidermal growth factor receptor signaling for
40 The pathogenesis of cancer metastasis: relevance to therapy

therapy of human pancreatic carcinoma. Clin Cancer Res 2001; 254. van Brussel JP, Busstra MB, Lang MS, Catsburg T, Schroder FH,
7: 2563–72. Mickisch GH. A phase II study of temozolomide in hormone-
233. Stetler-Stevenson WG. Invasion and metastasis. In: DeVita WT refractory prostate cancer. Cancer Chemother Pharmacol 2000;
Jr, Hellman S, Rosenberg SA, eds. Cancer: principles and prac- 45: 509–12.
tice of oncology, 7th edt. Philadelphia, PA: Lippincott, Williams 255. van Brussel JP, Mickisch GHJ. Multidrug resistance in prostate
& Wilkins, 2005; 113–126. cancer. Onkologie 2003; 26: 175–81.
234. Strohmeyer D, Rossing C, Bauerfeind A, et al. Vascular endothe- 256. van Brussel JP, van Steenbrugge GJ, Romijn JC, Schroder FH,
lial growth factor and its correlation with angiogenesis and p53 Mickisch GH. Chemosensitivity of prostate cancer cell lines and
expression in prostate cancer. Prostate 2000; 45: 216–24. expression of multidrug resistance-related proteins. Eur J Cancer
235. Sugarbaker EV. Cancer metastasis: a product of tumor-host inter- 1999; 35: 664–71.
actions. Curr Probl Cancer 1979; 3: 1–59. 257. van Brussel JP, van Steenbrugge GJ, Van Krimpen C, et al.
236. Sugarbaker EV, Cohen AM, Ketcham AS. Do metastases metas- Expression of multidrug resistance related proteins and prolifera-
tasize? Ann Surg 1971; 174: 161–70. tive activity is increased in advanced clinical prostate cancer. J
237. Sullivan GF, Amenta PS, Villanueva JD, Alvarez CJ, Yang JM, Urol 2001; 165: 130–5.
Hait WN. The expression of drug resistance gene products during 258. van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene expression
the progression of human prostate cancer. Clin Cancer Res 1998; signature as a predictor of survival in breast cancer. N Engl J Med
4: 1393–403. 2002; 347: 1999–2009.
238. Sundberg C, Ljungstrom M, Lindmark G, Gerdin B, Rubin K. 259. van ‘t Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression
Microvascular pericytes express platelet-derived growth factor- profiling predicts clinical outcome of breast cancer. Nature 2002;
beta receptors in human healing wounds and colorectal adenocar- 415: 530–6.
cinoma. Am J Pathol 1993; 143: 1377–88. 260. Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant
239. Sunderkötter C, Goebeler M, Schulze-Osthoff K, Bhardwaj R, progression. Meth Enzymol 2004; 381: 335–54.
Sorg C. Macrophage-derived angiogenesis factors. Pharmacol 261. Vermeulen SJ, Bruyneel EA, Bracke ME, et al. Transition from
Ther 1991; 51: 195–216. the noninvasive to the invasive phenotype and loss of α-catenin
240. Sunderkötter C, Steinbrink K, Goebeler M, Bhardwaj R, Sorg C. in human colon cancer cells. Cancer Res 1995; 55: 4722–8.
Macrophages and angiogenesis. J Leukoc Biol 1994; 55: 410–22. 262. Volm M, Koomagi R, Mattern J. Prognostic value of vascular
241. Takahashi Y, Bucana CD, Liu W, et al. Platelet derived endothe- endothelial growth factor and its receptor Flt-1 in squamous cell
lial cell growth factor in human colon cancer angiogenesis: role lung cancer. Int J Cancer 1997; 74: 64–8.
of infiltrating cells. J Natl Cancer Inst 1996; 88: 1091–2. 263. Wang Y, Jatkoe T, Zhang Y, et al. Gene expression profiles and
242. Takahashi Y, Kitadai Y, Bucana CD, Cleary KR, Ellis LM. molecular markers to predict recurrence of Dukes’ B colon can-
Expression of vascular endothelial growth factor and its receptor, cer. J Clin Oncol 2004; 22: 1564–71.
KDR, correlates with vascularity, metastasis, and proliferation of 264. Weiss L. Metastasis of cancer: a conceptual history from antiq-
human colon cancer. Cancer Res 1995; 55: 3964–8. uity to the 1990s. Cancer Metastasis Rev 2000; 19: 193–400.
243. Talmadge JE, Fidler IJ. Enhanced metastatic potential of tumor 265. Weiss L. Principles of metastasis. Orlando, FL: Academic Press,
cells harvested from spontaneous metastases of heterogeneous 1985.
murine tumors. J Natl Cancer Inst 1982; 69: 975–80. 266. Wilhelm SM, Carter C, Tang L, et al. BAY 43–9006 exhibits
244. Talmadge JE, Wolman SR, Fidler IJ. Evidence for the clonal broad spectrum oral antitumor activity and targets the RAF/
origin of spontaneous metastasis. Science 1982; 217: 361–3. MEK/ERK pathway and receptor tyrosine kinases involved in
245. Tarin D, Price JE, Kettlewell MGW, Souter RG, Vass ACR, Crossley B. tumor progression and angiogenesis. Cancer Res 2004; 64:
Mechanisms of human tumor metastasis studied in patients with 7099–109.
peritoneovenous shunts. Cancer Res 1984; 44: 3584–92. 267. Wilmanns C, Fan D, O’Brian CA, Radinsky R, Bucana CD, Tsan R,
246. Terranova VP, Hujanen ES, Martin GR. Basement membrane and Fidler IJ. Modulation of doxorubicin sensitivity and level of
the invasive activity of metastatic tumor cells. J Natl Cancer Inst P-glycoprotein expression in human colon carcinoma cells by
1986; 77: 311–6. ectopic and orthotopic environments in nude mice. Int J Oncol
247. Thatcher N, Chang A, Parikh P, et al. Gefitinib plus best support- 1993; 3: 413–22.
ive care in previously treated patients with refractory advanced 268. Xu L, Xie K, Mukaida N, Matsushima K, Fidler IJ. Hypoxia-
non-small-cell lung cancer: results from a randomised, placebo- induced elevation in interleukin-8 expression by human ovarian
controlled, multicentre study (Iressa Survival Evaluation in Lung carcinoma cells. Cancer Res 1999; 59: 5822–9.
Cancer). Lancet 2005; 366: 1527–37. 269. Yokoi K, Kim SJ, Thaker P, et al. Induction of apoptosis in tumor-
248. Theyer G, Schirmbock M, Thalhammer T, et al. Role of the associated endothelial cells and therapy of orthotopic human
MDR-1-encoded multiple drug resistance phenotype in prostate pancreatic carcinoma in nude mice. Neoplasia 2005; 7:
cancer cell lines. J Urol 1993; 150: 1544–7. 696–704.
249. Tsuruo T. Mechanisms of multidrug resistance and implications 270. Yokoi K, Sasaki T, Bucana CD, et al. Simultaneous inhibition of
for therapy. Jpn J Cancer Res 1988; 79: 285–96. EGFR, VEGFR, and platelet-derived growth factor receptor sig-
250. Tuxhorn JA, Ayala GE, Rowley DR. Reactive stroma in prostate naling combined with gemcitabine produces therapy of human
cancer progression. J Urol 2001; 166: 2472–83. pancreatic carcinoma and prolongs survival in an orthotopic nude
251. Uehara H, Kim SJ, Karashima T, et al. Effects of blocking plate- mouse model. Cancer Res 2005; 65: 10371–80.
let-derived growth factor-receptor signaling in a mouse model of 271. Yoshida K, Tosaka A, Takeuchi S, Kobayashi N. Epidermal
prostate cancer bone metastasis. J Natl Cancer Inst 2003; 95: growth factor receptor content in human renal cell carcinomas.
458–70. Cancer 1994; 73: 1913–8.
252. Uemura A, Ogawa M, Hirashima M, et al. Recombinant angio- 272. Yuan F, Chen Y, Dellian M, Safabakhsh N, Ferrara N, Jain RK.
poietin-1 restores higher-order architecture of growing blood Time-dependent vascular regression and permeability changes in
vessels in mice in the absence of mural cells. J Clin Invest 2002; established human tumor xenografts induced by an anti-vascular
110: 1619–28. endothelial growth factor/vascular permeability factor antibody.
253. Uhlman DL, Nguyen P, Manicel JC, et al. Epidermal growth factor Proc Natl Acad Sci USA 1996; 93: 14765–70.
receptor and transforming growth factor α expression in papil- 273. Zhai BJ, Wu F, Shao ZY, Hu K, Wang ZB. Establishment of
lary and non papillary renal cell carcinoma: correlation with human multidrug-resistant hepatocellular carcinoma cell line
metastatic behavior and prognosis. Clin Cancer Res 1995; 1: (HepG2/Adm) and biological characteristics evaluation. Ai
913–20. Zheng 2004; 23: 391–5.
3 Developmental therapeutics
and the design of clinical trials
ROBERT K. OLDHAM

New techniques in biotechnology and the use of biolo- speed the translation of new approaches to patients [6, 8,
gicals in cancer treatment have made it apparent that 17, 21, 22, 24, 30, 32, 33, 41, 55, 57, 61, 76–82, 90, 91,
developmental therapeutics for biotherapy are different 94, 98, 99, 103].
from standard drug development. Millions of chemicals Although the concept of biotherapy is not new, the
have been screened as anticancer agents, but less than 75 use of recombinant genetics to produce highly purified
have reached the clinic as commercial pharmaceuticals. biologicals as medicinals dates from about 1980 [31].
Perhaps 20 of these drugs can be classed as moderately A member of the alpha-interferon family was the first
effective; the rest are only marginally useful and all can biological produced by recombinant methods to be used
be highly toxic. With the discovery of monoclonal anti- as an anticancer medicine in humans [103, 105]. In the
bodies and conjugates thereof, the exploitation of bioen- 25 years since the first alpha-interferon molecule was
gineering to produce purified, characterized lymphokines/ prepared by recombinant methods, a large number of
cytokines and other biologicals, and further information recombinant molecules (lymphokines, cytokines, mono-
on the mechanism of action of these natural molecules, clonal antibodies, growth and differentiation factors,
the rate of development for biological therapeutics has angiogenesis factors and cell receptors) have become
risen dramatically. Because of their selectivity and with available or are being prepared for testing in the clinic
the implicit biological diversity of cancer, new approaches (see Chapter 8).
are needed to efficiently bring biotherapy to the clinic. Historical aspects in the development of immunother-
Previously, there have always been fewer promising agents apy have been reviewed [12, 75]. Before the 1980s, the
(drugs) to test than patients who needed new approaches. term immunotherapy was considered to be synonymous
In fact, the drug development paradigm has been a slow with biological therapy by most investigators. However,
and laborious, with each drug taking some 8–12 years to it is now clear that there are many biological approaches
gain commercial approval, at a cost of more than US$500 that may affect cancer growth and metastases, yet are
million per drug. Taxol, a drug approved for general use, not within the immune system. Thus, biotherapy now
entered clinical trials in the late 1980s and only became refers to agents derived from biological sources and/or
generally available in 1998; taking more than a decade to the use of agents that affect biological responses. The term
pass through our current system of drug development. “biologicals” describes agents extracted from or pro-
On average, fewer than four new chemotherapy drugs duced from biological materials. With biotechno-logy,
per year have been approved for general use by oncolo- this involves the use of recombinant genetics to isolate
gists. This expensive and slow paradigm reflects both the the gene, transfect it into an appropriate producer organ-
toxicity and marginal effectiveness of chemotherapeutic ism, and then the isolation and purification of the protein
drugs as well as a bureaucratic regulatory system more product. Genomics and proteonomics are processes to
fearful of criticism over toxicity than a willingness to pursue yield the “code” to eventually prepare biological therapy
opportunities for seriously ill patients [33, 70, 71, 83]. by chemical synthesis. The types of materials that alter
More recently, a large number of biological substances biological responses for the benefit of the patient have
and targeted therapies have been making their way to been called biological response modifiers (BRM). The
the clinic, increasing the difficulty of decisions as to the use of biologicals and BRM in the treatment of cancer
order and amount of preclinical and clinical testing. and other diseases is biotherapy (a term I initially used
Hundreds of biologic agents are in clinical testing with in 1984 to describe this fourth modality of cancer treat-
more agents to test than there are easily available patients ment) [84, 85]. As is always the case, nomenclature
for testing. The current system for drug development is can be confusing and terms may be variously defined
not sufficiently flexible, and needs major changes in by different individuals. In the broadest sense, biothe-
direction and technique to optimize clinical testing and rapy includes blood products, transplanted organs, bone

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 41


© Springer Science + Business Media B.V. 2009
42 Development therapeutics and the design of clinical trials

marrow/stem cell transplants, antibiotics (often derived formulation, documentation of biological activity and
by extraction from biological organisms and later syn- purity, early studies in the laboratory and in experimental
thesized), and a variety of other agents and approaches. animals to determine the mechanism of action and
However, in this chapter, the focus will primarily be on toxicity, and compilation of all the preclinical data into an
BRM, recombinant biologicals and gene engineered or investigational new drug application (INDA). It costs mil-
activated cells being developed as medicinals. lions of dollars for a pharmaceutical company to file a
Immunotherapy has had a checkered past. Kari Cantell single INDA. During preclinical development, companies
said it well when describing some of the early interferon make projections about the potential market size and
research: “Much second class research was carried out profitability as justification for the investment. These
with third class preparations slightly contaminated projections are difficult, and are most accurate when
with interferon” [5]. In addition, there have been ques- related to an existing drug in a known market. It follows
tionable approaches (“alternative medicine”) used by that predicting the market size for a totally new agent or
certain practitioners, and even frank quackery by others new approach is much more difficult.
who purported to deliver “immunotherapy” for cancer Subsequent to the INDA, further preclinical work on
patients. Even for the very dedicated scientists who have the mechanism of action and preclinical toxicology of a
explored immunotherapy over the past several decades, new drug is done. In addition, early-phase clinical studies
there have been many pitfalls relating to the purity are begun to determine biological activity in humans.
of their preparations, the source of the materials, and Although the process is not uniform for all classes of
the assays and techniques by which their measurements pharmaceuticals, studies are termed “phase I” when the
were made. dose of the drug is escalated to determine its biological
Huge expenditures by the National Institute of Health activity and its toxicity. Based on the preclinical toxicology
(NIH) in the area of molecular, biological and viral onco- information in small animals and sometimes in primates,
logy over the last 40 years led, in large part, to the current projections are made for the starting dose in humans. A low
technology of “genetic engineering.” In contrast to previous starting dose is selected to avoid severe toxicity in the
attempts to develop biological therapy, we now have initial patients in a phase I clinical trial. The rate of dose
techniques available that can isolate a single gene and use escalation in these trials and the acceptability of toxic
it in a bio-manufacturing process to produce absolutely side effects vary with the population at risk and are
pure proteins identical to those found in the body. It is related to the seriousness of the disorder and the treat-
with these techniques that thousands of biological com- ment alternatives for the patient. Because the starting
pounds and their synthetic analogs and components are doses are very low and because of phase I trial design,
being developed. Many of these biologicals will be using different patients at each dose level (to avoid
candidates for use as medicinals. A major value of the cumulative toxicity), the chance of a therapeutic effect
research done thus far with interferon is that it may be for the initial patients receiving a new agent is nearly
viewed as a model for the development of other biologi- zero. It is axiomatic that the phase I trials are designed
cal substances as medicinals [61, 76]. Interferon research to accumulate the maximum amount of clinical infor-
in particular and biotherapy in general must be seen as a mation with the least toxicity. Generally, the end point
new challenge in developmental therapeutics, with agents for phase I trials is the achievement of a maximum toler-
to be tested appearing at a still accelerating rate. No ated dose (MTD), the dose at which side effects are
longer can the historical drug development paradigm be unacceptable to the physician and the patient within the
used [55]. The methods used by the Food and Drug design of that clinical trial. These trials may require
Administration [11], National Institutes of Health, and 100–500 patients to reach agreement on the MTD in
the pharmaceutical industry in the development of drugs two to four schedules and routes of administration. In
must be drastically changed to a new paradigm that will addition, pharmacokinetic and metabolism studies are
accommodate the realities relevant to biologicals and done as part of Phase I. Once the MTD is established,
their use in medicine [33, 55, 71, 82, 86, 87, 90]. subsequent investigators can be reasonably assured that
the upper limit for the dose to be administered is well
defined and that therapeutic activity will not be missed
in later phase trials because of sub-therapeutic doses.
Drug Development Phase II studies are then conducted to determine the
The current process of drug development involves a therapeutic activity of the new drug in various types of
very long and costly set of procedures [33, 55, 80]. This cancer. Patients with specific cancers are selected in
includes the initial concept, extraction or synthesis, order that these trials can be conducted in reasonably
Robert K. Oldham 43

uniform patient groups. Based on the preclinical infor- Most drugs fail at the phase I/II level, being either too
mation and the clinical toxicology studies, as well as toxic or inactive. Drugs that complete phase II trials
pharmacokinetic and metabolic considerations, thera- with acceptable toxicity and activity in at least one
peutic doses are selected that represent the investigator’s cancer have a greater than 50% chance of successfully
“best guess” as to the therapeutically active dose range. passing phase III with eventual approval by the FDA.
In addition, schedule and route of administration must be For this reason, it has been suggested that drugs should
considered. In phase II trials, it is often necessary to be approved for general use at the end of phase II to
administer an agent at different doses by different routes speed up the process of bringing new drugs to the clinic
(e.g., intramuscular, intravenous, subcutaneous, oral) [47, 83].
and on different schedules (e.g., once a day, three times a The final step in the commercial development of a
day, 24-h infusion, etc.). Phase II trials are considered new drug requires the filing of a new drug application
complete when a substantial body of data exists concer- (NDA). All the information available under the INDA
ning the therapeutic activity of a new drug with refer- and all of the data from the phase I, II, and III testing are
ence to best dose, route of administration, and schedule made available to the FDA for review and consideration.
for therapeutic efficacy. If one accepts classical criteria Only the FDA is authorized to approve a new drug for
for cancer subtypes, over 100 histologic types of cancer sale in the United States; this usually involves defining
exist. If one assumes a standard statistical criterion of at the drug dose, route of administration, schedule, toxic-
least 14 patients treated to look for one response so as ity, and therapeutic activity in specific diseases (indica-
not to miss a 20% response rate, then a new drug would tions). After FDA approval, the company may begin
need to be tested in a minimum of 1,400 patients by each to advertise the agent for the approved use only. Uses
schedule and route to assure clinicians of its inactivity in outside the specific FDA approval indication(s) are termed
each tumor type; and if one assumes three routes and five “off label” uses. Perhaps 50% of all cancer drug therapy
schedules must be tested, over 21,000 patients would is “off label” use by physicians but the pharmaceutical
need to participate just to prove a specific drug ineffective. industry is specifically prohibited by the FDA from
Such studies demonstrating inactivity sacrifice patients providing clinical trial information to clinicians on “off
“for the good of the system.” Such FDA-mandated test- label” drug activity.
ing stretches ethical standards to the limit. The long time and enormous expenditure in the
Following the completion of adequate phase I and current drug development system have been justified as
phase II trials, phase III trials to compare a new agent being in the public interest inasmuch as the FDA requires
with standard treatment are conducted. The extent of extensive testing so active agents with reasonable toxi-
phase III trials depends on the treatment alternatives cities (safe drugs) can be made available as medicinals.
available. In the case of diseases for which other thera- The definitions of reasonable toxicity and of therapeutic
pies are effective, such trials may need to be extensive, efficacy have been the subject of considerable debate,
controlled, and employ random designs, sometimes with both in the general sense and relative to specific drugs.
blinding of the study both to the physician and to the Often, the long-term effects and/or toxicities of approved
patient. Such phase III trials have produced an enormous drugs have prompted secondary revisions in the regula-
literature on the subject of ethics, trial design, end points, tory process, further lengthening and expanding the steps
and the assessment of efficacy [9, 23, 28, 38, 71, 82, 91, necessary for new drugs to be approved. This process
92, 95, 106, 107, 110]. If, in such phase III trials, a new would seem to be totally in the public interest. However,
agent proves therapeutically superior without unaccept- when viewed in another context, the process clearly has
able side effects, in comparison with or in addition to some exclusionary aspects [47, 66, 71, 83]. The FDA
“standard” therapy, the new agent is very likely to receive defines a single standard for drug development in the
FDA approval. Since “standard therapy” is often only United States. Marked differences exist between coun-
marginally effective, large randomized trials proving a tries as to a regulatory body’s role in drug development,
new treatment is significantly (usually marginally) better and there are marked differences in the number of drugs
means most patients derive little or no real therapeutic available to patients in different countries. The regula-
benefit in the trials. Where efficacious therapy with an tory agency of each country has its own view of what is
approved drug is not available, fewer phase III data may ethical and in the public interest (different standards,
be needed to gain approval, and patients with advanced different ethics). Thus, there can be reasonable debate
cancer are often randomized against best supportive care on which rules are the best for the development of new
or a placebo. This latter practice also stretches ethical drugs. Finally, it is obvious that the long development
practice to the limit [47, 71, 95]. times and the huge expenditures required effectively
44 Development therapeutics and the design of clinical trials

restrict competition in the area of drug development. Perhaps the most cogent example relates to the devel-
Such restriction of competition gives major pharmaceu- opment of monoclonal antibodies. For years there have
tical firms a virtual monopoly on drug development. been sufficient data to indicate that monoclonal antibodies
Although most pharmaceutical firms would undoubtedly are eventually going to be very useful both diagnosti-
say that they would prefer a lower cost for drug deve- cally and therapeutically [10, 26, 36, 40, 51, 52, 54, 62,
lopment, one may view the process as being in their best 73, 94, 102, 103]. Now there are therapeutic antibodies
interests (barrier to entry), since it restricts competition approved for colon and breast cancer, lymphoma and
from smaller firms that cannot marshal the resources to leukemia with hundreds more in testing [8, 19, 21, 41,
carry out these extensive and expensive studies [47, 55, 94, 103]. The problems of developing new monoclonal
71, 79, 80, 82, 83]. antibodies for therapy is quite different from those
Thus, our drug development paradigm is highly restric- previously encountered with drugs. The most striking of
tive. It has worked reasonably well over the past 50 years these problems relate to market size. Generally speaking,
only because of public acceptance of a conservative one looks at the population afflicted with a particular
regulatory structure and because of the small number of disease and makes the presumption that a new drug will
relatively toxic drugs that actually were available for be active in a certain percentage of patients with that
clinical testing. The advent of biologicals has placed great disease. The percentage is often reasonably high and
pressure on this paradigm. Indeed, the effective develop- allows the market forecast to be applicable to a substan-
ment of biologicals will require changes in the regulations tial number of patients. For example, the number of
and policies for the development of new pharmaceuticals patients at risk per year with lung cancer is reasonably
[47, 55, 71, 78, 82, 90, 91]. well defined, and once some evidence of clinical activity
of a new drug in lung cancer is available, the market size
for that drug can easily be calculated. For monoclonal
antibodies, the situation is very different. At the extreme
Biologicals and BRM is anti-idiotypic monoclonal antibody therapy. It has
been demonstrated in preclinical models and, to a more
Development limited extent, in humans that anti-idiotypic antibody
A process of developmental therapeutics similar to that can be made to the specific tumor antigen (idiotype)
described for chemicals (drugs) is now being applied to of the neoplastic cell [11, 29, 39, 51, 65]. Such anti-
biopharmaceuticals. Historically, for new biologicals, idiotypic antibodies or idiotype vaccines derived therefrom
the information on composition, formulation, and purity have proven useful in controlling the growth of the B-cell
was often imprecise. In contrast to drugs, which are lymphomas. However, a critical problem for the devel-
generally small, synthesized molecules with a chemical opment of these reagents is market size; here, it is the
definition that is quite straightforward, biologicals have individual patient with that particular neoplasm. Thus,
had a more variable developmental process. Preparing a the market size might be as small as one. Even with some
biological for testing has involved extraction from a cross-reactivity, many of these antibodies or vaccines
microorganism, from a fraction of a cell culture, or from are expected to apply only to very small populations,
a tissue (complex chemical mixture). Such extractions much smaller than even the “orphan drugs” envisioned
yielded complex mixtures with defined biological activity, by FDA policy. Obviously, most pharmaceutical com-
where the precise chemical composition was incomplete panies will never develop these kinds of “individualized
or unknown [75]. Biotechnology is now making avail- or personalized medicines” under existing FDA guide-
able a range of biologicals (lymphokines/cytokines, lines [37, 71, 93, 94].
monoclonal antibodies, antigens, growth and maturation A less extreme example relates to the development of
factors) that are pure and as well defined as small mole- monoclonal antibodies for tumor-associated antigens
cule drugs [55, 57]. that may be restricted to subpopulations of cancer cells
Market size, the potential for profit, the long develop- [19, 20]. Considerable heterogeneity exists within any
ment period of preclinical and clinical testing, and the one histologic type of cancer (between patients) and
large investment necessary to develop new therapeutic even within a single patient’s cancer [54, 64]. It may be
agents have defined the scope of drug development. The that for any one cancer only a small portion of the patient
development and testing of new biologicals will require population will have a particular antigen or array of
extensive procedural and regulatory changes if we are to antigens on the cancer cell surface. Therefore, a mono-
be successful in bringing these agents to the clinic clonal antibody might be applicable only to 1%, 5%, or
quickly and efficiently [47, 55, 83, 92]. 10% of the patients with a particular type of neoplasm.
Robert K. Oldham 45

Because of clonal heterogeneity, both between patients responses in the context of the clinical trials [37, 48, 50,
and in different clones within individual patients, there 62, 105]. Since one may be administering the biological
may be the need to use multiple antibodies (“cocktails”) to stimulate a particular biological response, for which
in treatment [1, 46, 69, 90]. Given the over 100 histologic the dose-response curve may not be known a priori, one
types of cancer and the heterogeneity within each cancer must perform studies with pharmacokinetics to assay
type, market calculations may define very narrow appli- serum availability, and also measure the desired biologi-
cations [55, 63]. These considerations restrict market size cal effects to determine the optimal dose at which it
to a level unapproachable given drug development costs might alter a particular biological response (optimal bio-
under the current drug development paradigm. logical response modification, OBRM). Finally, sched-
Perhaps less obvious, but equally problematic, will be ule and route of administration have already proven
the development of other biologicals for treatment. For important [42, 76]. The pharmacokinetics after intrave-
lymphokines and cytokines, there is already evidence of nous and intramuscular administration differ for the
both antigen-specific and non-antigen-specific signals alpha interferons [43], and there has been a variable lack
that may have growth regulatory effects (See Chapter 8). of absorption of intramuscularly administered beta- and
One can visualize, with an antigen-specific lymphokine, gamma-interferons [34, 74]. Thus, the proper design of
how the signal may relate to the specific antigen and be phase I biotherapy trials must take into account appro-
applicable in a single patient, or a very few patients (e.g., priate measurements of bioavailability, pharmacokinet-
transfer factor, IgE suppressor factors). Less restricted, ics, biological response modification, and toxicity, all in
but still highly restricted compared to standard drug the context of escalating doses, to determine the dose–
development, is the development of non-antigen-specific response curves for each of these properties. Responses
lymphokines (e.g., interferons, lymphotoxins, interleu- to biologicals vary substantially between patients, and
kins), which are active against certain classes of cells, escalating doses in individual patients can yield valu-
thus making them clinically applicable only in selected able data without subjecting patients to “tests” with no
populations [55, 57]. therapeutic opportunity.
Like most biologicals, the interferons may act through
a diverse set of biological mechanisms. Cell surface recep-
tors for their activity exist, and responses to the adminis-
Interferons: the Early Model tration of biologicals are somewhat predetermined by the
Interferons represented early models for new biological condition and biological receptor repertoire of the patient.
approaches in cancer treatment [57, 61, 76]. “Natural” This situation is in direct contradistinction to that of drugs,
extracted interferons from stimulated white blood cells in which a totally new chemical is often administered to a
were used in initial clinical trials. The low purity, lot- patient in whom no standard biological response mecha-
to-lot variation, and expense of stimulating leukocytes nism existed a priori. Thus, biologicals may be viewed
to produce interferon, along with the difficulties in the in their physiologic role of correcting immunodeficiency
extraction and purification methods, limited the clinical states, as well as in the pharmacologic role of augment-
use of these materials. These preparations were typical of ing host responses and perhaps having direct antitumor
early forms of nonspecific immunotherapy [5, 75]. With effects.
the advent of increased interferon availability through Clearly, it was rational to test chemical drugs to MTD
recombinant genetics, and with the very high purity and to treat just below this dose in a “kill or cure” approach
(greater than 99%) of these preparations, extensive trials to cancer therapy. Current evidence suggests the OBRM
were completed for alpha-interferon preparations which dose and the MTD dose should be determined in cancer
led to the approval of alpha-interferon as the first geneti- biotherapy to properly design effective therapeutic trials.
cally engineered anticancer biopharmaceutical [42, 76]. Once determined, the design of therapeutic trials for bio-
The design of phase I biotherapy trials should differ therapy may differ greatly from classical chemotherapy
markedly from those for drugs. The dose-response curve studies.
for these agents may be very broad (and sometimes mul-
tiphasic), with peak effects at different doses for each
system responding to the biopharmaceutical. For exam-
ple, the immunomodulatory activity of alpha-interferon
Biotherapy Trial Strategies
can be seen at very low doses, whereas the antiprolifera- Many strategies exist or can be envisioned for conduc-
tive activity appears to be more reproducible at higher ting clinical trials with new biologicals. Some of these
doses. In biotherapy, there is a need to measure biological strategies have already been utilized in the early-phase
46 Development therapeutics and the design of clinical trials

testing of the interferons [4, 27, 59, 60, 103]. Two under- information before the higher dose is initiated in new
lying principles are apparent wherein biologicals and patients. By utilizing different groups of patients at each
drugs differ: when biologicals are administered, patients dose level, cumulative toxicity for any one patient is
already have physiologic mechanisms and receptors avoided. Patients receive a predetermined number of
that respond to them; and biologicals are derivatives of doses at each dose level and then are followed. During this
natural products of the mammalian or human genome, type of study, biological response modification and clini-
and may be expected to have less acute and chronic toxi- cal toxicity are assessed [4, 48–50, 53]. Pharmacokinetics
city than drugs at similar biologically effective doses. are also done in selected patients, so that the bioavailability
However, when high doses are used to exploit a certain can be determined [103].
action of a biological, acute and chronic toxicities may The dose-escalation scheme can be a modified
appear. These two considerations will not necessarily Fibonacci series, or some variation of this classical dose-
apply to those BRM that are well-defined chemical enti- escalation method. The considerations involved in dose-
ties and behave more in the manner of drugs with regard escalation methods dictated by drug toxicities do not
to toxicity. necessarily apply to biologicals. Thus, some inves-tigators
have used much faster dose escalations for biologicals.
Often, the dose escalation schedule is rather empirical,
Empirical Clinical Testing
the one selected being based on the best guess of the
In many early interferon trials a set dose of a “natural” investigators involved. This phase I drug development
interferon was given to a variety of patients with neo- strategy is based on historical data using new drugs and
plastic or viral disorders [57, 62, 76]. The chosen dose has several potential disadvantages. Since each patient
was the one expected, based on preclinical information receives only a particular dose for a defined period of
or other clinical data, to be relatively nontoxic and yet to time, it is highly likely that a large percentage of the
have sufficient biological effects to be therapeutically patients will receive sub-therapeutic doses if antitumor
active. In this context, most early clinical trials with effects are observed only at higher dose levels. Early
leukocyte interferon preparations were conducted at doses clinical trials with biologicals have not produced severe
under five million units per day, although it became cumulative toxicity. This clinical trial strategy, which
apparent later that doses up to 60 million units per day was designed to avoid the cumulative toxic effects seen
could be tolerated by the patients for a certain number of with drugs, is inappropriate for biotherapy. Since toler-
days. These trials were conducted as preliminary feasibi- ance (tachyphylaxis) may develop for some effects
lity trials or pilot phase II trials to gain some information (fever), entering different patients on progressively
on the biological effects of the interferon preparations. higher doses may expose patients to avoidable acute
Much of the information derived from them was anec- toxicities. Finally, the patients who are exposed to only
dotal, but they yielded preliminary information about one dose level are not individually assessed for biologi-
the biological effects and toxicities of alpha-interferons. cal response or for antitumor response over a wide
Now that the development of biotherapy is proceeding dosage range. This could give them a greater opportunity
more rapidly, other strategies are to be preferred. for optimization of biological and therapeutic response
[59]. This may be particularly true in the very-early-phase
Escalating dose Trials Using Different trials in which biological effects and antitumor effects
are totally unknown.
Groups of Patients
Phase I trials for biologicals have been modeled after Escalating-dose Trials within Individual
the standard phase I trial design used for drugs. Groups
of patients (usually three to five) are treated with a par-
Patients
ticular dose of a new biological with a single route of An alternative clinical trial strategy is the use of an esca-
administration [103]. These trials are begun with very lating-dose trial within individual patients [58, 59, 74, 94].
low (sub-therapeutic) doses based on preclinical infor- There are several variations of this theme, but each
mation, and the schedule is designed to increase the involves starting patients at a low dose and escalating
dose gradually to levels where toxicity occurs [55, 74]. the dose in each patient to determine the biological
Generally, new patients are entered at each higher dose response modifying effects and toxicities over a broad
level after all the patients have been entered on the dose range. This clinical trial strategy is very conserva-
previous dose and have received at least several treat- tive of patients, in that studies in small numbers of
ments. In this way, each patient group provides toxicity patients can give a large amount of information over a
Robert K. Oldham 47

broad dose range in each biotherapy trial. In the context It increases the patient’s therapeutic opportunity without
of this dose escalation, pharmacokinetics and biological undue risk of toxicity. This strategy is ethically prefe-
response modification can be measured in each patient. rable, most acceptable to patients and quite easy to
It is important in such a clinical trial strategy that describe in the informed consent [71].
bioavailability studies be done and the information be
available concomitantly with the study. Appropriate
“wash-out” periods between doses can be utilized so
Schedule
that the administered biological does not circulate in In the initial clinical trials, the schedule of administration
increasing quantities as the trial continues. For example, is generally empirical or is based on preclinical observa-
with the interferon trials, one can determine the serum tions. Once there is information on bioavailability, diffe-
level after each dose and administer the next dose when rent schedules of administration can be designed rationally.
interferon is no longer detectable. This allows the patient Both the bioavailability of the molecule and its biological
to avoid the possibility of severe acute toxic effects based effects are relevant. There are some biologicals that have
on cumulative serum levels. Obviously, biological and a very short serum half-life (measured in a few seconds to
therapeutic effects can still be cumulative and must be minutes) but may produce much longer lasting biological
monitored by appropriate clinical and biological mea- effects. It may be useful to compare the biological effects
surements throughout this type of trial. and toxicities of biologicals when administered under
This strategy offers the possibility of low-, medium-, conditions of rather constant exposure as against inter-
and high-dose therapy for the individual patient in the mittent exposure in order to gain a preliminary sense of
context of a single clinical trial. It maximizes the oppor- which schedule is most relevant [35, 74, 108]. Once these
tunity for the investigator to learn the optimal biological data are available, schedules of administration for phase
response modifying dose and the toxic dose, and per- II studies can be more rationally designed.
haps to gain therapeutic information, all in the context of
a single trial. Theoretically, rational maintenance regi-
mens could be designed based on the observations made
Route
during the escalating dose trial for each patient. The route of administration and delivery of biotherapy
For monoclonal antibody, this strategy may be particu- to selected target organs is critical. There are data indi-
larly important, in that the delivery of the monoclonal anti- cating that certain biologicals are inactivated, or poorly
body to the tumor site may be the most important absorbed, when given intramuscularly or subcutane-
consideration in developmental therapeutics [25, 36, 54, ously. For such biologicals intravenous administration
63, 94]. Giving a low dose and then progressively higher is quite important. When data on bioavailability are not
doses to the same patient, with subsequent determination known from preclinical models, it is probably important
of antibody localization, is a useful way of determining to conduct early clinical trials with the intravenous
the correct dose for delivery of the antibody and/or its con- route, since such trials can provide early data on serum
jugates to the appropriate target organ in the context of a pharmacokinetics and provide the best opportunity for
toxicity study. This trial design rationally ties targe-ting broad biodistribution of the administered biological. It is
(delivery), toxicity, and therapeutic effects together in important in phase I biotherapy trials to determine if
combined phase I/II studies in a manner perfectly appro- serum levels are measurable and if biological effects are
priate for antibody and conjugates, in direct contradistinc- seen in the presence or absence of serum levels for
tion to drug development [2, 3, 17, 18, 59, 96, 98]. agents given by any route of administration. There may
In studies that employ escalating doses, a useful vari- be instances in which second mediators are involved,
ation is to enter individual patients for a limited number with useful biological effects occurring without apparent
of doses. With this trial design, the initial three or four serum bioavailability.
patients may enter at the lowest dose level and progress Effusions may contain both tumor cells and reactive
through dose level 4 or 5, at which time the second immunological elements, in which case the administra-
group of patients may be entered at dose 3 and escalated tion of a biological into a restricted space such as the
upward in a manner that allows the second group to fol- thoracic or peritoneal cavity might be appropriate. There
low the first group in dosage escalation toward the are indications that certain immunomodulators may be
MTD. A third group may be entered at dose level 6 and effective in this setting [56]. Patients with tumors that
so on. This strategy allows one to avoid administering remain confined to a single compartment for a prolonged
the full dose range to all patients, avoids most cumu- period of time may offer unique opportunities for bio-
lative toxicity, and helps avoid sub-therapeutic doses. therapy [72, 88]. Ovarian cancer, with its propensity for
48 Development therapeutics and the design of clinical trials

remaining localized in the abdominal peritoneum, may and very difficult to design. It would be virtually impos-
be ideal for evaluating the antitumor activity of cells sible to investigate a significant number of biologicals in
[72], biologicals, and BRM in a relatively closed space. phase II or III trials utilizing this type of clinical trial
Interleukin-2 with activated cells is active when infused design. The number of patients required for trials in
into selected anatomical sites and visceral cavities where the adjuvant setting is enormous, in that the recurrence
tumor is present [44, 88]. It seems clear that targeting of rate cannot be precisely predicted without a concurrent
monoclonal antibody and its conjugates may be improved random control group. The lack of certainty for recurrence
by selective organ or region perfusion or infusion. prompts ethical questions in designing phase II or III trials
Considerations for the design of early-phase trials in for these patients, since dose, route, schedule, OBRM and
these spaces differ markedly from those using systemic therapeutic efficacy have not been determined.
administration. It has now been well demonstrated that certain bio-
logicals have activity in patients with bulky and resistant
Patient Selection disease [4, 7, 13–16, 27, 57, 58, 67–68, 76, 89, 94, 108].
Even though biotherapy may be more effective, as
Historically, patient selection for phase I drug trials has chemotherapy and radiotherapy are, when the tumor
been broad, and patients bearing many tumor types have burden is small, that does not mean that it is totally inef-
been entered into the trials. While this may be appropriate fective in patients with bulky disease. Given the large
for drugs, and for selected lymphokines that act broadly number of biologicals available to the clinic, there is a
on immunological responses or may act in a general anti- need to develop methods of rapid clinical testing in
proliferative manner across a broad spectrum of tumors, early-phase trials, and this will necessitate testing in
it is not appropriate for those biologicals that act more patients with apparent disease [55, 57, 75, 108]. Indeed,
specifically. Thus, a monoclonal antibody that has been the initial clinical trials with interferons, interleukins,
specifically designed to recognize a particular type of monoclonal antibodies, and cellular therapies generally
cancer can be appropriately tested only in patients with selected such patients with a good performance status
that type of cancer [45, 59]. and a reasonably functional immune system. These
In vitro determinations of specificity and activity may patients have shown evidence of biological response
play a greater role in the selection of the patient popula- modification, and they have shown antitumor responses.
tions for phase I biotherapy trials. The escalating-dose Thus, such clinical trials can be used as indicators for
phase I trials, with the dose being escalated within indi- biological activity of new agents [57, 67, 94].
vidual patients, preselected on the basis of in vitro speci-
ficity and/or activity, appear to be most relevant and
efficient for determining the distribution, biological
effects, and toxicity of monoclonal antibodies. The same
Future Prospects
considerations may apply to certain lymphokines/cytok- Developmental therapeutics for biotherapies is well
ines, in which case selection as to activity may be on underway. From the inception of this field in 1980 to the
an individual patient basis. These trials may be more present, a great number of new approaches have become
appropriately termed phase I/II trials. In fact, given the available in biotherapy [42, 57, 71, 94, 108]. Unlike the
heterogeneity of cancer, both with respect to the specificity field of drugs, where very large numbers of compounds
of recognition by antibody and with respect to activity by are screened in a preclinical testing program and very
immunoconjugates and lymphokines/cytokines, in vitro few reach the clinic, a much higher percentage of the
determinations may play a major role in clinical trial biologicals selected rationally and tested actually go on
design. Biological systems are diverse, and single patients to clinical trials. This is due to selection based on the
may require specifically designed treatments when spec- known physiologic activities of biologicals as opposed
ificity and activity restrict the applicability of biotherapy. to random testing of chemicals in drug development.
The design of early-phase [biotherapy] clinical trials needs The strategies for these clinical trials need to be cost effi-
to be radically changed [55, 58, 71]. cient, advantageous to the patient, ethical [71, 97],
There has been much speculation concerning the and scientifically interpretable to the clinician/scien-
types of patients most appropriate for trials with bio- tist [64, 75]. Phase I strategies are most important in
logicals. Many believe that these agents should be tested that they give the initial leads on which the design of
primarily in the adjuvant setting, where the tumor early phase II and III trials are based. With proper selec-
burden is quite small and the biological responses to be tion clinical trial strategies, new biologicals can be more
modified are still healthy [75]. Although this sort of effectively screened and evaluated as potential anti-
strategy may be optimal, it is also prohibitively expensive cancer agents [59, 71].
Robert K. Oldham 49

Biotherapy is the fourth modality of cancer treatment emerge for other forms of biotherapy [42, 57, 59, 67, 71,
[57, 58, 84, 85, 86, 94]. These agents and this technol- 104, 108, 109]. This should give pause to those who
ogy have far broader applications in medicine than can- believe the immunological dogma that biotherapy can
cer therapeutics. Given the large number of cloned only be active in minimal residual disease. Like chemo-
biologicals and the virtually unlimited number of recom- therapy and radiotherapy, biotherapy may be more
binant molecules that can be produced therefrom, along active with lesser tumor burdens, but current data indi-
with unlimited variety of monoclonal antibodies that cate that activity and hence selection of compounds for
can now be produced, it is apparent that the coming further study can be assessed in patients with advanced
years will produce new challenges for those involved disease.
in developmental therapeutics [33]. This tremendous The potential for the use of biotherapy is great. We
expansion of agents and approaches available for cancer are in a new era in cancer therapeutics. To develop bio-
therapeutics will increase our opportunities and amplify therapy, we must begin to approach this new era in phar-
the complexity in the preclinical evaluation of these maceutical research and to contemplate novel methods
agents and their translation to the clinic. The numbers of for the efficient and timely development of biotherapy
biologicals to be evaluated and their inherent selectivity rather than simply continuing in old paradigms (Fig. 1)
call for new methods in the selection of the “most likely [33, 55, 71, 82, 83]. Some would still say no new treat-
to succeed.” Additionally, the methods used historically ment can be judged without randomized, clinical trials.
by pharmaceutical manufacturers and the regulations Such trials are valid in searching for small differences
established by governmental agencies for approving between a new treatment and an ethically acceptable
anticancer drugs are inhibitory to the rapid development control (standard treatment) [101]. Whether placebo
of biotherapy [47, 55, 83]. Novel approaches will be controls are ever acceptable in cancer treatment is debat-
necessary if these biologicals are to be effectively and able; however, pilot studies without controls can be very
rapidly translated to clinical trials [47, 55, 64, 95]. useful even to the point of defining efficacy if the treat-
Alpha-interferon, IL-2 and monoclonal antibodies ment effect is large and obvious. A recent and innova-
and their conjugates have anti-tumor effects, even in tive “n of 1” trial also bears consideration in trials of
patients with bulky disease. This finding is likely to biotherapy [35].

Preclinical Testing (in vitro/animal model)

Phase I Clinical Trial

Phase II Clinical Trial

Phase III Clinical Trials

Unresponsive or poorly responsive neoplasm;


Less than partial responses

Partial responses to newer active single agents

Increased partial responses and rare complete responses


To some single and combined agents
Figure 1. Although radically different,
chemotherapy and biotherapy share a Increased complete responses to more effective
systemic approach to cancer treatment. For Combination therapy
those cancers that chemotherapy has been
able to cure, reaching that goal required a Increased survival in complete responders with or
sequence of steps in which new and increas- Without continued treatment during remission
ingly more effective agents and combinations
of agents had to be identified. Biotherapy can
be expected to travel a similar path A portion of long-term survivors are cured
50 Development therapeutics and the design of clinical trials

We must not just continue to simply do what has been 14. Dillman RO, Oldham RK, Tauer KW, et al. Continuous interleu-
done historically [71]. Development therapeutics for kin-2 and lymphokine activated killer cells for advanced cancer: an
NBSG trial. J Clin Oncol 1991; 9:1233–1240.
biologicals certainly represents the kind of science 15. Dillman RO, Church C, Oldham RK, et al. Inpatient continuous
Lewis Thomas described: infusion Interleukin-2 in 788 cancer patients: the NBSG Experience.
Cancer 1993; 71:2358–2370.
It is hard to predict how science is going to turn out, and if it is 16. Dillman RO. The clinical experience with Interleukin-2 in cancer
really good science it is impossible to predict. This is in the nature therapy. Cancer Biother 1994; 9(3):179–182.
of the enterprise. If the things to be found are actually new, they are 17. Dillman RO, Dillman JB, Halpern SE, et al. Toxicities and side
by definition unknown in advance, and there is no way of telling in effects associated with intravenous infusions of murine monoclo-
advance where a really new line of inquiry will lead. You cannot nal antibodies. J Biol Response Mod 1986; 5:73–84.
make choices in this matter, selecting things you think you’re going 18. Dillman RO. Human antimouse and antiglobulin responses to
to like and shutting off the lines that make for discomfort. You monoclonal antibodies. Antibody, Immunoconj and Radiopharm
either have science or you don’t, and if you have it you are obliged 1990; 3:1–15.
to accept the surprising and disturbing pieces of information, even 19. Dillman RO Monoclonal antibodies for treating cancer. Ann Inten
the overwhelming and up heaving ones, along with the neat and Med 1989; 111:593–603.
promptly useful bits. It is like that. 20. Dillman RO. Antibodies as cytotoxic therapy. J Clin Oncol 1994;
12:1497–1515.
21. Dillman RO. Magic bullets at last! Finally – approval of a mono-
clonal anbitody for the treatment of cancer!!! Cancer Biother
Radiopharm 1997; 12:223–225.
References 22. Dillman RO. Radiolabeled anti-CD20 monoclonal antibodies
for the treatment of B-cell lymphoma. J Clin Oncol 2002; 20:
1. Avner B, Swindell L, Sharp E, et al. Evaluation and clinical rele- 3545–3557.
vance of patient immune responses to intravenous therapy with 23. Ellenberg SS. Studies to compare treatment regimens: the random-
murine monoclonal antibodies conjugated to Adriamycin. Mol ized clinical trial and alternative strategies. J Am Med Assoc 1982;
Biother 1991; 3(1):14–21. 246:2481–2482.
2. Bernhard MI, Foon KA, Oeltman TN, et al. Guinea pig line 10 hepa- 24. Fidler IJ, Berendt M, Oldham RK. Rationale for and design of a
tocarcinoma model: characterization of monoclonal antibody and screening procedure for the assessment of biological response modi-
in vivo effect of unconjugated antibody and antibody conjugated to fiers for cancer treatment. J Biol Response Modif 1982; 1:15–26.
diphtheria toxin A chain. Cancer Res 1983; 43:4420–4428. 25. Foon KA, Bernhard KA, Oldham RK. Monoclonal antibody ther-
3. Bernhard MI, Hwang KM, Foon KA, et al. Localization of 111In- apy: assessment by animal tumor models. J Biol Response Modif
and 125I-labeled monoclonal antibody in guinea pigs bearing line 1982; 1:277–304.
10 hepatocarcinoma tumors. Cancer Res 1983; 43:4429–4433. 26. Foon KA, Schroff RW, Bunn PA, et al. Effects of monoclonal anti-
4. Bunn PA Jr., Foon KA, Ihde DC, et al. Recombinant leukocyte body serotherapy in patients with chronic lymphocytic leukemia.
alpha interferon: an active agent in advanced cutaneous T-cell lym- Blood 1984; 64:1085–1093.
phomas. Ann Intern Med 1982; 101:484–487. 27. Foon KA, Sherwin SA, Abrams PG, et al. Treatment of advanced
5. Cantell K. In: Burke D, Cantell K, DeMaeyer E, Landy M, Revel non-Hodgkin’s lymphoma with recombinant leukocyte alpha
M, Vilcek J, eds. Interferon 1 New York: Academic Press, 1981. interferon. N Engl J Med 1984; 311:1148–1152.
6. Carter SK. The clinical trial evaluation strategy for interferons and 28. Fost N. Consent as a barrier to research [editorial]. N Engl J Med
other biological response modifiers – not a simple task [editorial]. 1979; 300:1272–1273.
J Biol Response Modif 1982; 1:101–105. 29. Giardina SL, Schroff RW, Woodhouse CS, et al. Detection of two
7. Cheever MA, Greenberg PD, Fefer A. Potential for specific cancer distinct malignant B-cell clones in a single patient using anti-idiotype
therapy with immune T lymphocytes. J Biol Response Modif monoclonal antibodies and immunoglobulin gene arrangement.
1984; 3:113–127. Blood 1985; 66:1017–1021.
8. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus 30. Ghielmini M, Schmitz SF, Cogliatti SB, et al. Prolonged treat-
rituximab compared with CHOP alone in elderly patients with dif- ment with rituximab in patients with follicular lymphoma signi-
fuse large-B-cell lymphoma. N Engl J Med 2002; 346:235–242. ficantly increases event-free survival and response duration
9. Curran WJ, Reasonableness and randomization in clinical trials: compared with the standard weekly × 4 schedule. Blood 2004; 103:
fundamental law and governmental regulation [editorial]. N Engl J 4416–4423.
Med 1979; 300:1273–1274. 31. Goeddel D, Yelverton E, Ullrich A, et al. Human Leukocyte
10. DeVita VT Jr, Hellman S, Rosenberg SA, eds. Biologic therapy of Interferon Nature (Lond) 1980; 287:411–416.
cancer, second edition. Philadelphia, PA: J.B. Lippincott, 1995; 32. Guisti RM, Shastri KA, Cohen MH, et al. FDA drug approval sum-
553–607. mary: panitumumab (Vectibix). Oncologist 2007; 12:57–83.
11. DeVita VT Jr, Hellman S, Rosenberg SA, eds. Biologic therapy of 33. Grillo-Lopez AJ. Regularity process for approval of biologicals
cancer, second edition. Philadelphia, PA: J.B. Lippincott, 1995; for cancer therapy. Chapter in: principles of cancer biotherapy, 5th
553–565. ed. Dordrecht, The Netherlands: Kluwer.
12. DeVita VT Jr, Hellman S, Rosenberg SA, eds. Biologic therapy of 34. Gutterman JU, Rosenblum MG, Rios A, et al. Pharmacokinetic
cancer, second edition. Philadelphia, PA: J.B. Lippincott, 1995; study of partially pure interferon in cancer patients. Cancer Res.
879–890. 1984; 44:4164–4171.
13. Dillman RO, Oldham RK, Barth NM, et al. Continuous interleu- 35. Guyatt G, Sackett D, Taylor DW, et al. Determining optimal ther-
kin-2 and tumor infiltrating lymphocytes as treatment of advanced apy – randomized trials in individual patients. N Engl J Med 1986;
melanoma. Cancer 1991; 68(1):1–8. 314:889–892.
Robert K. Oldham 51

36. Hanna MG, Jr., Key ME, Oldham RK. Biology of cancer therapy: 60. Oldham RK. Biologicals and biological response modifiers: new
some new insights into adjuvant treatment of metastatic solid strategies for clinical trials. In: Finter NB and Oldham RK, eds.
tumors. J Biol Response Modif 1983; 4:295–309. Interferons, IV. Amsterdam: Elsevier, 1985; 235–249.
37. Herberman RB, Thurman GB. Summary of approaches to the 61. Oldham RK. Interferon: a model for future biologicals. In: Burke D,
immunological monitoring of cancer patients treated with natural or Cantell K, Gresser I, De Maeyer E, Landy M, Revel M, Vilcek J,
recombinant interferons. J Biol Response Modif 1982; 2:548–562. eds. Inf. VI. New York: Academic Press, 1985; 127–143.
38. Horwitz RI, Feinstein AR. Improved observational method for 62. Oldham RK. Biologicals for cancer treatment: interferons. Hospital
studying therapeutic efficacy. JAMA 1981; 246:2455–2459. Practice 1985; 20:72–91.
39. Hurvitz SA, Timmerman JM. Current status of therapeutic vaccines 63. Oldham RK, Foon KA, Morgan AC, et al. Monoclonal antibody
for non-Hodgkin’s lymphoma. Curr Opin Oncol 2005; 17:432–440. therapy of malignant melanoma: in vivo localization in cutaneous
40. Hwang KM, Foon KA, Cheung PH, et al. Selective antitumor metastasis after intravenous administration. J Clin Oncol 1984;
effect of a potent immunoconjugate composed of the A chain of 2:1235–1242.
abrin and a monoclonal antibody to a hepatoma-associated anti- 64. Oldham RK. Therapeutic monoclonal antibodies: effects of
gen. Cancer Res 1984; 44:4578–4586. tumor cell heterogeneity. In: Present status of nontoxic concepts
41. Kavvinavar FF, Hambleton J, Mass RD, et al. Combined analysis in cancer therapy. Cancer symposium (Germany). Basel: Karger,
of efficacy: the addition of bevacizumab to fluorouracil/leucovorin 1986.
improved survival for patients with metastatic colorectal cancer. 65. Oldham RK. Monoclonal antibody therapy. In: Chiao JW, ed.
J Clin Oncol 2005; 23:3706–3712. Biological response modifiers and cancer research. New
42. Kirkwood JM, Ernstoff MS. Interferons in the treatment of human York:Marcel Dekker, 1988; 40:3–16.
cancer. J Clin Oncol 1984; 2:336–352. 66. Oldham RK. Set my factors free. Mol Biother. 1990; 2(4):194–195.
43. Knost JA, Sherwin SA, Abrams PG, et al. The treatment of cancer 67. Oldham RK. Cancer biotherapy: principles and practice. New York:
patients with human lymphoblastoid interferon: a comparison of Marcel Dekker, 1991.
two routes of administration. Cancer Immunol Immunother 1983; 68. Oldham RK, Dillman RO, Yannelli JR, et al. Continuous infusion
15:144–151. interleukin-2 and tumor derived activated cells as treatment of advanced
44. Lembersky B, Baldisseri M, Seski J, et al. Phase IB study of intra- solid tumors. An NBSG trial. Mol Biother 1991; 3(2):68–73.
peritoneal (IP) interleukin-2 (IL-2) for refractory ovarian cancer 69. Oldham RK. Custom tailored drug immunoconjugates in cancer
(OC). Proc Am Assoc Cancer Res 1990; 31:277. therapy. Mol Biother 1991; 3(3):148–162.
45. Lewko WM, Ladd PA, Pridgen D, et al. Tumor acquisition propa- 70. Oldham RK. The cure. Franklin, TN: Pulse Publications, 1991.
gation and preservation: culture of human colorectal cancer. 71. Oldham RK. BioEthics: opportunities, risks and ethics: the priva-
Cancer 1989; 64:1600–1608. tization of cancer research. Franklin, TN: Media America, 1992.
46. Liao SK, Meranda C, Avner BP, et al. Immunohistochemical phe- 72. Oldham RK, Greco FA. Brief intensive chemotherapy and second
notyping of human solid tumors with monoclonal antibodies in look laparotomy in advanced ovarian carcinoma. In: William CJ,
devising biotherapeutic strategies. Cancer Immunol Immunother Whitehouse M, eds. Recent advances in clinical oncology. London:
1989; 28:77–86. Churchill Livingstone, 1982; 165–185.
47. Madden BJ. A clinical trial for the FDA’s clinical trial process. 73. Oldham RK, Morgan AC, Woodhouse CS, et al. Monoclonal anti-
Cancer Biother Radiopharm 2005; 20(6):569–578. bodies in the treatment of cancer: preliminary observations and future
48. Maluish AE, Leavitt R, Sherwin SA, et al. Effects of recombinant prospects. Med Oncol Tumor Pharmacother 1984; 1:51–62.
alpha interferon on immune function in cancer patients. J Biol 74. Oldham RK, Sherwin SA, Maluish A, et al. A phase I trial of immune
Response Modif 1983; 2:470–481. interferon: a preliminary report. In: Goldstein AL, ed. Thymic hor-
49. Maluish AE, Ortaldo JR, Sherwin SA, et al. Changes in immune mones and lymphokines. New York: Plenum Press, 1984; 497–506.
function in patients receiving natural leukocyte interferon. J Biol 75. Oldham RK, Smalley RV. Immunotherapy: the old and the new.
Response Modif 1983; 2:418–427. J Biol Response Modif 1983; 2:1–37.
50. Maluish AE, Ortaldo JR, Conlon JC, et al. Depression of natural 76. Oldham RK, Smalley RV. The role of interferon in the treatment of
killer cytotoxicity following in vivo administration of recombinant cancer. In: Zoon KC, Noguci PD, Lui TY, eds. Interferon: research,
leukocyte interferon. J Immunol 1983; 131:503–507. clinical application and regulatory consideration. New York: Elsevier,
51. Miller RA, Maloney DG, Warnke R, et al. Treatment of B cell 1984; 191–206.
lymphoma with monoclonal anti-idiotype antibody. N Engl J Med 77. Oldham RK, Thurman GB, Talmadge JE, et al. Lymphokines,
1982; 306:517–522. monoclonal antibodies and other biological response modifiers in
52. Nowinski RC, Tam MR, Goldstein LC, et al. Monoclonal antibodies the treatment of cancer. Cancer 1984; 54:2795–2810.
for diagnosis of infectious disease in humans. Science 1983; 219:637. 78. Oldham RK, Patient-funded cancer research. N Engl J Med 1987;
53. Oldham RK. Toxic effects of interferon. Science 1982; 219:902. 316:46–47.
54. Oldham RK. Monoclonal antibodies in cancer therapy. J Clin 79. Oldham RK. Drug development: who foots the bill? Bio/technol-
Oncol 1983; 1:582–590. ogy 1987; 5:648.
55. Oldham RK. Biologicals: new horizons in pharmaceutical devel- 80. Oldham RK. Who pays for new drugs? Nature 1988; 332(28):795.
opment. J Biol Response Modif 1983; 2:199–206. 81. Oldham RK, Avent RA. Clinical research: who pays the bills?
56. Oldham RK. Guest editorial: biological response modifiers. J Natl Oncology Issues 1989; 4(2):13–14.
Cancer Inst 1983; 70:790–796. 82. Oldham RK. Clinical research in cancer: a time for consensus.
57. Oldham RK. Biologicals and biological response modifiers; fourth Pharm Exec 1989; July.
modality of cancer treatment. Cancer Treat Rep 1984; 68:221–232. 83. Oldham RK. Regulatory hierarchies (editorial). Mol Biother 1988;
58. Oldham RK. Biologicals and biological response modifiers: new 1(1):3–6.
approaches to cancer treatment. Cancer Invest 1985; 3:53–70. 84. Oldham RK. Biotherapy: the fourth modality of cancer treatment.
59. Oldham RK. Biologicals and biological response modifiers: the Cancer: perspective for control symposium. J Cell Physiol Suppl
design of clinical trials. J Biol Response Modif 1985; 4:117–128. 1986; 4:91–99.
52 Development therapeutics and the design of clinical trials

85. Oldham RK. Biotherapy: the fourth modality of cancer treatment. 99. Romond EH, Perez EA, Bryan J, et al. Trastuzumab plus adju-
In: Mak TW, Sun TT, eds. Cancer: perspective for control sympo- vant chemotherapy for operative HER2-positive breast cancer. N
sium. New York: Alan R. Liss, 1986; 91–99. Engl J Med 2005; 353:1673–1684.
86. Oldham RK. The government-academic “industrial” complex. J Biol 100. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone
or with bevcizumab for non-small-cell lung cancer. N Engl J Med
Response Modif 1986; 5:109–111.
2006; 355:2542–2550.
87. Oldham RK. The cure for cancer. J Biol Response Modif 1985; 101. Schafer A. The ethics of randomized clinical trials. N Engl J Med
4:111–116. 1982; 307:719–724.
88. Oldham RK, Bartal AH, Yannelli JR, et al. Intra-arterial and intra- 102. Sears HF, Herlyn D, Steplewski Z, Koprowski H. Effects of mono-
cavitary administration of lymphokine activated killer cells in clonal antibody immunotherapy on patients with gastro-intestinal
patients with advanced cancer: feasibility and laboratory results. adenocarcinoma. J Biol Response Modif 1984; 3:138–150.
Proc AACR (abstr) 1988; 29:396. 103. Sherwin SA, Knost JA, Fein S, et al. A multiple dose phase I trial
89. Oldham, RK, Lewko W, Good R, et al. Growth of tumor derived of recombinant lymphocyte alpha interferon in cancer patients.
activated T cells for the treatment of cancer. Cancer Biother 1994; JAMA 1982; 248:2461–2466.
9(3):211–224. 104. Slamon DJ, Leyland-Jones B,Shak S, et al. Use of chemothe-rapy
90. Oldham RK. Fundamentally different. Cancer Biother Radiopharm plus a monoclonal antibody against HER2 for metastatic breast
1999; 14:413–15. cancer that overexpresses HER2. N Engl J Med 2001; 344:
91. Oldham RK. Cracking the FDA code. Wall Street J Op/Ed. 783–92.
2.2.2001. 105. Smalley RV, Oldham RK. Interferon as a biological response
92. Oldham RK. Informal opinions: editorial introduction. Cancer modifying agent in clinical trials. J Biol Response Modif 1983;
Biother Radiopharm 2002; 17:247. 2:401–409.
93. Oldham RK. FDA trials cost lives. Wall Street J Op/Ed. 3.3.2002. 106. Sylvester RJ, Pinedo J, De Pauw M, et al. Quality of institutional
94. Oldham RK, Dillman RO. Monoclonal antibodies in cancer ther- participation in multicenter clinical trials. N Engl J Med 1981;
apy: twenty-five years of progress. JCO. 2008; 26(11):1774–1777. 305:852–855.
95. Oldham RK, Dillman RO. Gold standard or wrong standard? 107. Weiss DG, Williford WO, Collins JF, Binham SF. Planning multi-
Cancer Biother Radiopharm 2004; 17(6):271–272. center clinical trials; a biostatistician’s perspective. Controlled
96. Orr DW, Oldham RK, Lewis M, et al. Phase I trial of Mitomycin-C Clin Trials 1983; 4:53–64.
immunoconjugate cocktails in human malignancies. Mol Biother 108. West WH, Tauer KW, Yannelli JR, et al. Constant infusion recom-
1989; 1(4):229–240. binant interleukin-2 in adoptive immunotherapy of advanced
97. Relman AS. The ethics of randomized clinical trials: two perspec- cancer. N Engl J Med 1987; 316(15):898–905.
tives [editorial]. N Engl J Med 1979; 300:1272. 109. Vitetta ES, Krokick KA, Miyama-Inaba M, et al. Immunotoxins:
98. Ricart AD, Tolcher AW. Technology insight: cytotoxic drug immu- a new approach to cancer therapy. Science 1983; 219:644–649.
noconjugates for cancer therapy. Nat Clin Prac Oncol 2007; 110. Zelen M. A new design for randomized clinical trials. N Engl J
4:1379–1389. Med 1979; 300:1242–1246.
4 Recombinant proteins and genomics
in cancer therapy
KAPIL MEHTA, BULENT OZPOLAT, KISHORCHANDRA GOHIL,
AND BHARAT B. AGGARWAL

Abbreviations ADA, adenosine deaminase; AML, acute because of the lack of methods for producing adequate
myeloid leukemia; bFGF, basic fibroblast growth factor; amounts of the pure protein. Interest in interferon beyond
CML, chronic mylogenous leukemia; CSF, colony-stimu- the field of virology began in the early 1960s, when
lating factor; EGF, epidermal growth factor; EPO, eryth- workers began to recognize its growth-inhibitory and
ropoietin; FDA, Food and Drug Administration; HIV, immune-activation properties. During the 1960s and
human immunodeficiency virus; IFN, interferon; IL, early 1970s, reports of interferon’s antiviral and antitumor
interleukin; LIF, leukemia inhibitory factor; MAb, activity in laboratory animals and humans stirred up this
monclonal antibody; PDGF, platelet-derived growth interest and several groups decided to purify human inter-
factor; PE, Pseudomonas exotoxin; PEG, polyethylene feron for clinical use.
glycol; TGF, transforming growth factor; TNF, tumor The first practical method of producing sufficient
necrosis factor quantities of interferon was developed by Cantell et al.
[31]. They were able to isolate 100–200 mg of interferon
from 1,000 l of starting material that contained 2–5 kg of
other contaminating proteins. The purified material had
Introduction a specific activity of greater than 108 U/mg protein [69]
Recently published sequence of the complete human and was sufficient to treat only a few patients [282], but
genome represents a major milestone in the era of the the initial clinical results stimulated wider interest in
modern molecular biology [318, 132]. The sequencing expanding production of interferon for more extensive
of approximately 3.2 billion nucleotides of the human clinical trials. Enthusiasm intensified when interferon-
genome that is estimated to contain about 20,000–25,000 alpha was successfully cloned and the purified recombi-
protein-encoding genes signifies the first step down the nant protein became available [225, 226].
long road. Gene identification does not necessarily trans- The first trial to test dose levels and side effects of the
late into an understanding of gene function. Although purified bacterial product in human beings began in 1981
mapping and cloning of several genes have linked them [227, 112]. The availability of pure recombinant pro-
to heritable genetic disorders, the normal function of a tein led researchers to crystallize interferon, the first step
majority of these genes remains unknown. Recombinant toward analysis of the protein’s three-dimensional struc-
DNA technology has made it possible to generate large ture by X-ray crystallography. This permitted the produc-
amounts of many biologically active proteins and to deli- tion of individual molecular species of interferon free
neate their functions. The novelty of recombinant techno- from other species and other proteins that were simulta-
logy is the precision and efficiency with which scientists neously induced in human cell cultures. By using this
can manipulate single gene. The ability to isolate human technique, our knowledge of the varied biological proper-
genes and insert them into microorganisms, which then ties of interferons, previously determined with relatively
produce human proteins, thereby serving as biological crude preparations, has been confirmed and extended.
factories, has revolutionized the field of biology.
Interferons have special significance to recombinant
DNA technology as paradigm modifiers of immune Isolation, Cloning and Expression
response. The interest in the therapeutic potential of inter-
feron against cancer and viral diseases has served as
of Genes
catalyst to the emerging recombinant DNA industry. A gene is a defined region of a chromosome comprising
Despite its great promise as an antiviral agent, the clinical a specific sequence or part of a long polynucleotide.
application of interferon had been rather slow, mainly It codes for some specific function or characteristic

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 53


© Springer Science + Business Media B.V. 2009
54 Recombinant proteins and genomics in cancer therapy

GENE

-A-G-A a b c d
C-T-A-G- -
-T-C-T G-A-T-C- - Eukaryotic DNA Genome
Intron exon
Transcription

a b c d
Primary RNA transcript

a b c d
Splicing

a b c d
mRNA

Translation

Protein
NH2 ------ COOH

Protein processing, folding

Mature functional protein

Figure 1. The organization of a eukaryotic gene and processing of the RNA transcript

(phenotype) of a cell. The eukaryotic genome contains mRNA molecule is then translated to produce the cor-
up to 109 nucleotides in 50–100,000 genes [40, 97]. To responding protein (Fig. 1).
study the events in such a complex system it is neces- The organization of bacterial genes is simpler. They
sary to be able to isolate and study a single gene in a do not produce the enzymes necessary for RNA splicing,
purified form. Molecular cloning provides a method for so a eukaryotic gene containing introns, if introduced
isolating a single discrete segment of DNA from a popu- into a bacterial cell as in recombinant techniques, will
lation of genes and amplifying the DNA segment to pro- not be properly expressed. This problem can be circum-
duce enough pure material for chemical, genetic, and vented by making a DNA copy (cDNA) from the appro-
biological analysis. Cloning and expression of foreign priate mRNA by using the enzyme reverse transcriptase
genes has permitted access to such complex biological (Fig. 2). As an alternative, the DNA can be fragmented
mechanisms as RNA splicing, oncogene dynamics and at specific target sequences with the help of restriction
antibody diversity. It has also been the foundation for endonucleases [249]; the DNA fragment that contains
the new biotechnology industry. the gene of interest is inserted into the purified DNA
The organization of genes in higher organisms is genome of a self-replicating genetic element, generally
more complex than in bacteria and viruses. The linear a virus or a plasmid. A DNA fragment containing a human
array of information in a complex gene contains one or gene, for example, when inserted into such a virus or
more stretches of non-coding sequences, called introns. plasmid, can be joined in a test tube to the chromosome
The remaining sequences, which encode information of a bacterial cell. Starting with only one such recombi-
for proteins, are called exons. When these genes pro- nant DNA molecule which can infect only a single
duce a protein, the DNA is transcribed into a large RNA cell, the normal replication mechanism of the virus
molecule from which the introns are removed; this can produce more than trillion identical molecules in
Kapil Mehta et al. 55

Genomic DNA
GENE Restriction nuclease digestion
------ ------

Transcription
mRNA

Reverse Transcriptase

DNA Fragments
Alkali degradation
Single-stranded DNA

DNA Polymerase

Complementary DNA (cDNA)

cDNA molecules are joined to


pBR322
vector DNA for propagation in
bacteria

Selection of clone(s) carrying desired cDNA


sequence, using a nucleic acid or antibody probe

Expression of cloned gene in host organisms such as


bacteria, yeast or mammalian cells

Figure 2. Gene cloning from mRNA or genomic DNA

less than a day, thereby amplifying the amount of the common and convenient host organisms. They are simple
inserted human DNA fragment by the same factor, making to grow, have short generation times, provide large yields,
it possible to infect millions of cells. The virus or plas- and are most cost-effective. Particularly in the case of
mid used in this way is known as a cloning vector, and B. subtilis, the cells can be induced to secrete the product
the DNA propagated by insertion into it is said to have into culture medium, which facilitates the purification
been cloned. of the cloned protein.
Cloning the gene or cDNA encoding a particular However, there are some disadvantages of using bacte-
protein is only the first of many steps needed to produce rial cells for gene expression (Table 1). Though most pro-
a recombinant protein for medical and industrial use. teins are expressed in large amounts in bacteria, some
Expression of foreign genes in a host organism requires of them fail to fold properly, leading to formation of inso-
vectors which contain specific control sequences gov- luble ‘inclusion bodies.’ Protein extracted from these
erning transcription and efficient splicing of RNA. The inclusion bodies is sometimes biologically inactive. Small
most popular expression systems used for this purpose proteins can be refolded into their native form, but larger
are the bacteria Escherichia coli and Bacillus subtilis, ones containing several cysteine residues, in general, stay
yeast, cultured insect cells and mammalian cells. The inactive, most likely because of the improper formation
choice of expression system depends on the properties of disulfide bridges. Moreover, the expressed foreign pro-
of the protein to be produced. Bacterial cells are most tein is sometimes toxic to the bacteria, so that the culture
56 Recombinant proteins and genomics in cancer therapy

Table 1. Posttranslational processing of proteins in various using mammalian expression systems [61], but this pro-
expression systems cess is often lengthy and costly.
Insect Mammalian Expression of foreign proteins in cultured insect cells
Event Bacteria Yeast cells cells by baculovirus vectors is an alternative to this problem.
Proteolytic +/− + + This relatively new system is becoming the system of
Cleavage choice for expressing mammalian and viral proteins.
Glycosylation − + + + Baculovirus promoters are among the strongest known
Secretion +/− + + + and can drive the expression of target genes at high levels
Folding +/− +/− + + (1–500 mg/l). The baculovirus expression system offers
Phospho- − + + + several other advantages over prokaryotic, yeast, and
rylation
mammalian expression systems [178]. Some of the advan-
Acylation − + + +
Amidation − − + +
tages include: high-level expression, correct folding, the
% yield 1–5% 1% 30% <1% ability to catalyze post-translational modifications like
mammalian cells (Table 1), quick and easy growth in
monolayers or suspension cultures without CO2, and safest
producing the protein cannot be grown to high cell density. – they do not infect humans, animals or plants. More than
This problem can be overcome by using an inducible 400 different proteins have been expressed by using bacu-
promoter that is turned on to begin the transcription of the lovirus vectors, and in most cases the protein produced is
foreign gene only after the culture has been grown. Unlike similar in structure, biological activity and immunological
eukaryotic cells, bacterial cells lack enzymes that cata- reactivity to the authentic protein [177]. Although the cost
lyze posttranslational modifications of proteins such as of culturing insect cells is currently more than that of
phophorylation, acylation and glycosylation. These post- culturing bacteria or yeast, it is still significantly less than
translational modifications are sometimes essential for that of culturing mammalian cells.
the normal functioning of a protein. Despite the significant advantages of producing
Yeast cells may be preferable to bacteria for the human proteins in heterologous host cells, in some cases
production of some proteins biological by recombinant the best method for producing a mammalian protein is
DNA procedures. Yeast is a simple eukaryote that resem- a mammalian cell system. Transient expression in
bles mammalian cells in many ways but, like bacterial cells, mammalian cells is often used to check the function of
can be grown conveniently and economically. Yeast cells a newly cloned gene and as a quick method to assess
are capable of catalyzing many post-translational modi- the function of engineered proteins. The extracellular
fications that are found on mammalian proteins [126]. domains of cell-surface receptors have been engineered
Also, they process the signal peptides needed for the for secretion from cells by introduction of a stop codon
secretion of protein and thus can be induced to secrete into the gene before the sequence of the transmembrane
certain proteins into the growth medium for harvesting. domain. These soluble receptors, as we will discuss later
Moreover, unlike bacteria, yeast cells do not have endo- in this chapter, are valuable reagents for the study of
toxins. However, yeasts do produce active proteases ligand binding and for screening receptor agonists or
that can degrade the foreign proteins, thereby reducing antagonists that may eventually be used as therapeutics
the yield of the final product. To overcome this problem, themselves. Although transient transfections yield enough
the construction of yeast strains with deleted protease protein for laboratory experiments, stably integrated
genes is being attempted. amplified genes in mammalian cells are necessary for
Until recently, the expression of a biologically active large-scale production of proteins [2]. With this goal in
protein from a complex eukaryotic gene in large amounts mind, great improvements have been made in recent
was a problem. The problems with prokaryotic expres- years in identification of appropriate promoters, vectors,
sion systems have already been described. Even in yeast transformation protocols and host cell systems.
(eukaryotic) expression systems, the low biological
activity of complex eukaryotic proteins remains a com-
mon problem. Mammalian expression systems provide Recombinant Proteins as Cancer
all the post-translational modifications that may be
necessary for full activity of eukaryotic proteins, but the
Therapeutics
yields from these systems are generally much lower Biotherapy, as an alternative to the rigorous cytotoxic
than can be obtained from E. coli or yeast. Recently, regimens used in the treatment of various malignant dis-
protein yields as high as 10 mg/l have been obtained by orders, offers distinct and attractive advantages over
Kapil Mehta et al. 57

Antiproliferative
Anti-angiogenic

IFN
TNF
Anti-VEGF antibody
TGF
Endostatin
Amphiregulin
Angiostatin
Oncostatin M
Lymphotoxin
FAS/APO ligand Approaches to the
Biological Therapy of
Cancer

Differentiation Immunomodulatory Cytotoxic


CSFs
Immunotoxins
IFN-gamma IFNs
antibodies
TNF TNF
Fas/APO
LIF
EPO

Figure 3. Model for recombinant DNA-derived protein therapeutics for cancer

conventional chemotherapy. Biological therapy is based on target cells and regulate the proliferation, differentia-
on the principle of stimulating the body’s own immune tion and metabolism of either the same cell (autocrine)
response and/or using biological substances against a dis- or another cell (paracrine). Cytokines are different from
ease. A perception of the malignant cell as one whose dif- endocrine hormones in that they are produced by any
ferentiation has been blocked due to the lack, deficiency or number of different cells rather than by specialized
mutation of some key element led to the emergence of a glands. Different cytokines exhibit considerable overlap
biological therapy. The strategy of biological therapy con- in their biological activities. Because of the advent of
trasts with the immediate cell death induced by cytotoxic recombinant DNA technology, cytokines have become
drugs, where there is no attempt to restore homeo-stasis. available in highly pure form and in sufficient quanti-
Biotherapy may offer the opportunity to use relatively ties. This has accelerated the elucidation of in vitro and
nontoxic agents, the body’s own elements, to correct the in vivo biological activities of these proteins and led to
underlying problem. It has long been recognized that the their rapid testing in the patients as therapeutic agents
immune system plays a pivotal role in the patient’s response for the treatment of cancer. The cytokines that have been
to disease. Therefore, recent cancer therapies have been shown to have therapeutic potential in cancer include
directed at modulation of components of the immune the interferons (IFNs), interleukins (ILs), colony- stim-
system (Fig. 3). In particular, “immune messengers” or ulating factors (CSFs), and tumor necrosis factors
cytokines may play a vital role in regulating host antitumor (TNF). Table 2 lists some recombinant cytokines that
defense mechanisms. Knowledge of cytokines and their are currently being used for the treatment of malignant
functions is expanding rapidly [3, 6, 15, 122, 123, 126, 135, diseases.
179]. In the following section we will discuss the potential
role of cytokines in the treatment of malignant disease.
Interferons
Interferons are a family of regulatory glycoproteins pro-
Cytokines duced by many cell types in response to viral infections
In general, cytokines are low molecular weight (10– and a variety of mitogenic and antigenic stimuli. Three
50 kDa) proteins secreted by cells of the immune system major classes of IFNs have been described: IFN-α and
that bind with great specificity and affinity to receptors IFN-β, which share components of the same receptor
58 Recombinant proteins and genomics in cancer therapy

Table 2. Cytokines Approved or in Development for Human EPO Anemia associated with chronic USA
usea renal failure
Countries Malignancy, chemotherapy
Cytokine Target disease approved AIDS, AZT treatment
rheumatoid arthritis
r.Protein biotherapeutics approved by FDA for human use anemia of prematurity
IFN-α Chronic myelogenous leukemia USA, Europe, Autologous blood donation prior to
Japan surgery
Hairy cell leukemia USA, Europe, Compensation of surgical blood
Japan loss
AIDS-related Kaposi’s sarcoma USA Hepatitis B hepatomas surface
Chronic non-A/non-B/C USA antigen
hepatitis
Condylomata acuminata USA r.Protein biotherapeutics currently under clinical trials
Lymphoma IL-1αβ Malignant disorders
Essential thrombocythemia Bone marrow transplantation
Melanoma Severe aplastic anemia
Certain solid tumors Allotransplant patients
Multiple myeloma IL-3 Advanced neoplasms
Acute hepatitis B Secondary hematopoietic failure
IFN-β Renal cell carcinoma Europe Bone marrow recovery
Advanced solid tumors Breast cancer
Soft tissue carcinoma IL-4 Metastatic renal cell carcinoma
Adult T cell leukemia Metastatic breast carcinoma
IFN-γ Chronic granulomatous USA Metastatic melanoma
disease (CGD) Disseminated cancer
Advanced solid tumors Advanced malignancies
Renal cell carcinoma IL-7 Refractory solid tumors
Adult T cell leukemia Metastatic melanoma
Chronic myelogenous leukemia Metastatic kidney cancer
Lepromatous leprosy IL-11 Relapsed non-Hodgkin lymphoma
IL-2 Renal cell carcinoma Europe Leukemia
Metastatic melanoma Japan, Europe Melanoma
Advanced malignancies IL-12 Lymphoma
G-CSF Non-myeloid malignancies USA Advanced solid tumors (mela-
associated with chemo- noma, neuroblastoma, breast
therapy-induced cancer)
myelosuppression IL-18 Ovarian carcinoma
Myelodysplastic syndromes IL-21 Melanoma
Severe chronic neutropenia Ovarian carcinoma
(cyclic, idiopathic, congenital) IL-29 Hepatitis C (associated with
Acute myelogenous leukemia hepatocellular carcinoma)
Bone marrow transplantation CSF-1 Endometrial carcinoma
GM-CSF Autologous bone marrow USA, Japan IFN-α Metastatic renal carcinoma,
transplantation for non- Metastatic melanoma
Hodgkin’s lymphomas, Multiple myeloma
Hodgkin’ s disease, acute Lymphoma
lymphocytic leukemia TNF-α Advanced neoplasms
Allogeneic bone marrow trans- Reduction of ovarian ascites
plantation for leukemias M-CSF Solid tumors
Myelodysplastic syndromes/ Breast cancer
aplastic anemia
Fungal infections
Cancer chemotherapy associated
Acute myelogenous leukemia
myelosuppression
IL-1 AML, CML, sepsis, septic shock
Acute myelogenous leukemia
Receptor
AIDS, anti-AIDS drug treatment
antagonist
Associated myelosuppression
a
Disease in bold indicates approved use of cytokine.
Kapil Mehta et al. 59

and are referred to as type I; and IFN-γ, which uses a shown to induce remissions in some patients with well-
separate receptor system and is referred to as type II differentiated B-cell tumors [282].
(Table 3). Additional IFNs have been discovered, but The most remarkable effects of recombinant IFN-α
they are not well characterized [259]. Crude IFN was were observed in patients with hairy cell leukemia
probably the first cytokine used in patients and was [237]. Treatment with partially purified or recombinant
shown to delay recurrent growth of tumors in patients IFN-α suppressed peripheral blood cell production and
who had undergone surgery for osteogenic sarcoma rapidly increased platelet counts in these patients [112].
[282, 283]. Pharmacological doses of partially purified Their immune status improved and the number of leuke-
IFN-α were reported to induce regression of tumors in mia cells in the bone marrow and blood declined. These
significant numbers of patients with metastatic breast patients stopped having opportunistic infections and
cancer, low-grade lymphoma, or multiple myeloma required no further platelet and erythrocyte transfusions.
[110]. In 1981, IFN-α was successfully cloned by two These studies led to the approval of IFN-a for treatment
groups, and the purified protein was immediately tested of hairy cell leukemia in June 1986 by the United States
in the clinic. This was the first study of a recombinant Food and Drug Administration (FDA), an action adopted
cytokine in patients with cancer and, for the most part, by regulatory agencies from 31 other countries. The mecha-
the biological activity seen with the partially pure natu- nisms of IFN-α action in the compromised survival of
ral form was reproduced with the recombinant DNA- the patients with hairy cell leukemia are not fully under-
derived form [111]. Treatment with IFN-α was also stood but may include differentiation, cell-cycle arrest
and/or apoptosis.
Table 3. Recombinant DNA-derived proteins relevant to Other B-cell neoplasms also show variable degrees of
treatment of cancer sensitivity to IFN-α [110, 111, 282]. In patients with
Cytokine Receptor(s) used Reference multiple myeloma or low-grade lymphoma, the cytokine
often has demonstrated a positive clinical impact on
Interferon-α (IFNα) IFNα/β-R [2, 225]
Interferon-ß (IFN-β) IFNα/β-R [293]
survival when combined with chemotherapy [182, 272,
Interferon-γ (IFN-γ) IFNγ-R [103, 241, 261] 275]. Clinical results with IFN-α in patients with chronic
Transforming growth TGF-β -R1 and -RII [2, 56] myelogenous leukemia (CML) have been rather inter-
factor-β (TGF-β) esting. During the chronic phase of CML, IFN-α treatment
Thymosin α1 [329] causes hematologic remission [112]. Approximately 75%
Epidermal growth EGF-R [2, 101, 261] of the patients in the benign phase of the disease achieve
factor (EGF) complete normalization of blood counts. Moreover,
Platelet derived growth PDGF-RI and RII [116]
IFN-α had the astonishing capacity to suppress selec-
factor (PDGF)
Fibroblast growth factor FGF-R [98] tively the cells bearing the Philadelphia chromosome,
(Basic) (bFGF) resulting in partial or complete restoration of the normal
Fibroblast growth factor FGF-R [98] clone [290].
(Acidic) (aFGF) While showing great promise for leukemia, IFN-α
Transforming growth EGF-R [57] therapy of solid tumors has been rather discouraging.
factor-a (TGF-α) Only 10–15% of patients with renal cell carcinoma or
Insulin like growth IGF-RI and IGF-RII [54]
factor(IGF-I & II)
malignant melanoma undergo regression in response to
Hepatocyte growth HGF-R [199] IFN-α. However, the responses of carcinoid tumors to
factor (HGF) the cytokine were encouraging. A majority of patients
Macrophage colony- M-CSF-R [2, 145] showed improvement in symptoms, and a smaller frac-
stimulating factor tion experienced tumor regression [210]. Both squamous
Granulocyte colony- G-CSF-R [2, 120, 276] and basal cell carcinomas of the skin show sensitivity to
stimulating factor
IFN-α, as a single agent or in combination with retin-
GM-colony stimulating GM-CSF-R [2, 335]
factor oids [170, 229]. Mycosis fungoides, a malignancy of the
Leukemia inhibitory LIF-R [2, 99] T helper cells, is also sensitive to IFN-α alone or with
factor (LIF) other modalities including retinoids [140, 247]. Kaposi’s
Stem cell factor (SCF) SCF-R [2, 331] sarcoma, an angioproliferative disease that commonly
Erythropoietin (EPO) EPO-R [2, 78] develops in individuals infected with human immuno-
B cell growth factor BCGF-R [260] deficiency virus (HIV), has responded well to IFN-α
Hepatitis B surface ? [65, 314] therapy: 40% of patients experienced significant regres-
antigen
sion of lesions [282]. This work led to the approval of
60 Recombinant proteins and genomics in cancer therapy

IFN-α in the U.S. and 21 other countries for the treat- Table 5. Interleukins with relevance to cancer treatment
ment of AIDS-related Kaposi’s sarcoma. Interleukin Receptor Reference
After isolation of its gene and 10 years’ work identif-
Interleukin-1-α (IL-1α) IL-1R type I; IL-1R [59]
ying several of its biological activities, IFN-α was tested type II
in human subjects for a wide variety of diseases includ- Interleukin-1-β (IL-1ß) IL-1R type I; IL-1R [59]
ing cancer. It was found to have impressive effects against type II
chronic granulomatous disease, leading to its approval Interleukin-1 receptor IL-1R type I; IL-1R [17]
by the FDA for human use [298]. More recently, IFN-β antagonist type II
was approved for treatment of ambulatory patients with Interleukin-2 (IL-2) IL-2R. α, IL-2R. β, [162]
relapsing/remitting multiple sclerosis [4]. Clinical toxic
IL-2R.γ
Interleukin-3 (IL-3) IL-3R. α, IL-3γ [131]
effects associated with administration of IFNs and other Interleukin-4 (IL-4) IL-4R, IL-2R . γ [205]
cytokines are summarized in Table 4. The IFNs, like Interleukin-5 (IL-5) IL-5R. α, IL-5R. γ [288]
most other cytokines, are produced by the body to act Interleukin-6 (IL-6) IL-6R, gp130 [8]
locally. When used as a systemic pharmaceutical, they Interleukin-7 (IL-7) IL-7R (CDw127), [46]
can have substantial toxic effects [282]. IL-2R. γ
Interleukin-8 (IL-8) IL-8R, Type I & [128, 193]
Type II
Interleukins Interleukin-9 (IL-9) IL-9R, IL-2R . γ;
R [239, 240]
Interleukin-10 (IL-10) IL-10R1; IL-10R2 [188]
The interleukins are a family of cytokines that are essen-
Interleukin-11 (IL-11) IL-11R, gp130 [64, 222]
tial to both cellular and humoral immune responses. Interleukin-12 (IL-12) IL-12R [304]
To date, at least 34 interleukins have been identified Interleukin-13 (IL-13) IL-13R . α, IL-4R, [187, 347]
and cloned (Table 5). Many of these molecules exhibit IL-2R. γ
Interleukin-15 (IL-15) IL-15Rα, IL-2R . β, [90]
IL-2R. γ
Interleukin-16 (IL-16) CD4 [49]
Table 4. Common toxic effects associated with administra- Interleukin-17 (IL-17) IL-17R [166]
tion of cytokines Interleukin-18 (IL-18)/ IL-18R [200, 201]
Fever IGIF
Chills Interleukin 19 (IL-19)a IL-19R [86]
Nausea Interleukin-20 (IL-20) IL-20Ra. & IL-20Rβ [24]
Vomiting Interleukin-21 (IL-21) IL-21R [217, 353]
Headache Interleukin-22 (IL-22) IL-22R [149,
Anorexia 336–349]
Fatigue Interleukin-23 (IL-23) IL-12Rα1, IL-12Rβ2 [213]
Myalgias Interleukin-24 (IL-24) IL-20R1,IL-20R2, [347, 354,
Arthralgias IL-22R, IL-22R2 355]
Bone pain Interleukin-27 (IL-27) IL27RA [349, 350]
Flush
Interleukin-32 (IL-32) [351, 352]
Local erythema
Inflammation at the site of injection a
IL-19 is a novel homologue of human IL-10.
Capillary leak syndrome
Granulocytopenia
Thrombocytopenia
Anemia
Hypotension antineoplastic activity. Currently, IL-2, IL-12, IL-15
Liquid accumulation in the lung, spleen, kidneys and IL-18 are the most potent inducers of anti-tumor
Reversible increase in body weight
activity in preclinical and clinical studies. Recently, new
Reversible increase in the serum creatinine
Oliguria interleukins have been characterized, including IL-21,
Malaise IL-23, IL-24 and IL-27, which exert potent immuno-
Asthenia modulatory and anti-tumor effects in vitro and in vivo
Rigors settings, indicating that they may have considerable
Diarrhea
Hepatocytotoxicity
potential as future cancer therapeutics [348].
Lethargy, depression Interleukin 1 (IL-1): IL-1 was originally described as
Mental confusion an “endogenous pyrogen” in 1940 because of its ability
EEG-abnormalities to cause fever when injected into animals. Two forms of
Kapil Mehta et al. 61

IL-1 are now recognized: IL-1a is cell-associated and is observed that lasted for 3–14 months in 29% patients
involved in antigen presentation, whereas IL-1β, the with renal cell cancer and 23% of those with melanoma.
predominant form, is readily secreted by macrophages. Further potential of IL-2 in cancer therapy has been
Though the two forms have limited amino acid homol- demonstrated by using recombinant IL-2 in combination
ogy, IL-1α and IL-1β bind to the same receptor and with other cytokines [244, 306]. For example, recombi-
share several biological properties. Other cell types that nant IL-2 in combination with IFN-α elicited a potent
produce IL-1 are endothelial cells, keratinocytes, neu- antitumor response in several animal tumor models [175].
trophils and B lymphocytes. Constitutive expression of The most significant antitumor activity seen with IL-2
IL-1 occurs in cells lining the external environment, i.e., therapy has been in malignant melanoma and renal cell
skin and mucosal surfaces [58]. The cDNAs coding for carcinoma. Other tumors treated with IL-2 include
both human IL-1s were reported in 1985 [39, 83]. glioma, bladder carcinoma, ovarian carcinoma, neuro-
Recombinant IL-1s induced fever, hepatic protein syn- blastoma, lung carcinoma, head and neck carcinoma,
thesis, production of prostaglandin E2, cartilage break- breast carcinoma, lymphoma, colon carcinoma and
down, bone resorption, and elevated ACTH and mesothelioma [123]. As shown in Table 4, IL-2 adminis-
augmented the T lymphocyte response to antigens and tration in patients is associated with a wide range of toxic
mitogens [58]. Recombinant IL-1 exhibits cytostatic effects, the most common being fluid retention, anemia,
activity toward human melanoma tumor cells in vitro thrombocytopenia and hypotension [175]. IL-2 is the
and direct cytotoxic effects against human melanoma first cytokine that has been employed so widely in clinical
cell line A375 [157]. Recently, recombinant IL-1 was trials. However, because of its toxic effects, its clinical use
shown to enhance the recovery of platelets in ovarian has been limited.
cancer patients following carboplatin therapy, suggesting Interleukin-3 (IL-3): Recombinant IL-3 is a 15–17-
a potential role for IL-1 in attenuating thrombocytopenia kDa polypeptide that is known to stimulate mast cells,
associated with chemotherapy [309]. neutrophils, macrophages and megakaryocytes. No direct
Interleukin-2 (IL-2): IL-2 is a 15.5-kDa glycoprotein antitumor activity has been observed for IL-3, but this
produced by peripheral blood lymphocytes and is a cytokine has a role in increasing platelet and neutrophil
potent growth factor for activated T lymphocytes. It acts counts in patients with advanced malignancy [87]. Phase
as a cofactor in development of cytotoxic T lymphocyte I and II clinical trials with recombinant IL-3 have been
activity against tumors and has been shown to participate carried out in patients with advanced malignancy. A dose-
in tumoricidal activity by inducing the growth of natural dependent increase in platelet counts and substantial
killer (NK) cells and lymphokine-activated killer (LAK) increases in the numbers of circulating neutrophils,
cells [242]. Several cancers show sensitivity toward eosinophils, monocytes and lymphocytes were observed
recombinant IL-2, both in animal models and in the in these patients [87]. Hematopoietic failure caused by
patients. LAK cells are peripheral blood lymphocytes prolonged chemotherapy, radiotherapy or infiltration of
that can be generated in vitro by incubation with high bone marrow by tumor cells could be restored by recom-
doses of IL-2. They have the ability to kill tumor cells binant IL-3 treatment. The side effects of rIL-3 therapy
specifically while leaving normal cells unharmed. IL-2 in patients include fever, bone pain and headache [87].
has been used in combination with LAK cells to achieve Thus, recombinant IL-3 is a multilineage hematopoietic
more potent antitumor response [242–245]. In 1985, cytokine with promising effects on platelet and neutro-
Rosenberg and associates published the results of their phil counts.
first study documenting tumor regression in patients with Interleukin-4 (IL-4): IL-4 is a T cell-derived glyco-
melanoma following administration of IL-2 and LAK protein of 20 kDa. The gene for human IL-4 has been
cells [243]. An update of the results in 180 patients was cloned [16]. The antitumor functions of IL-4 include
published in 1991 [242]. Antitumor responses were seen increased T cell, NK cell and monocyte proliferation.
in patients with advanced melanoma, renal cell cancer, IL-4 has also been shown to enhance the generation of
colon cancer or non-Hodgkin’s lymphoma. cytotoxic T lymphocytes and to participate in induction
Like LAK, tumor-infiltrating lymphocytes (TILs), the of LAK cell activity, and to synergize with I-2 in this
lymphoid cells that infiltrate solid tumors, can be grown activity [194]. Furthermore, IL-4 can stimulate the gen-
in vitro in the presence of IL-2. These cells have unique eration of TILs in human melanoma, increase the anti-
lytic activity against autologous tumors. Treatment with gen-presenting ability of mouse and human monocytes
TILs in combination with IL-2 was shown to mediate and augment the expression of tumoricidal activity
substantial tumor regression in some patients with advanced in murine macrophages. It appears to inhibit the release
malignant melanoma [174]. Objective responses were of TNF, IL-1 and IL-6 by human monocytes [296].
62 Recombinant proteins and genomics in cancer therapy

Interleukin-4 has been shown to exert potent antitumor recovery of bone marrow in patients with myelosup-
activity against several transplantable tumors in a pression that usually follows aggressive chemotherapy
murine model. Using IL-4-transfected tumor cells, the regimens. The results of phase I clinical studies of IL-6
potential of transfecting lymphokine genes into tumor have recently been reported [325]. IL-7 is a 22 to 25-kDa
cells as a method of cancer therapy has been demon- glycoprotein that was originally characterized on the
strated [93]. Because of its antitumor effects in vitro and basis of its ability to promote the growth of precursor B
in murine models, IL-4 may be useful in inhibiting the lymphocytes [96]. Recombinant IL-7 induces the prolif-
growth of solid tumors and cell lymphomas. eration of both thymocytes and mature T cells and is
known to activate macrophages for tumor cell killing. In
human monocytes, IL-7 induces the expression of IL-8,
Other Interleukins IL-6, IL-1a, IL-1.b and TNF-α. IL-12, a product of B
The family of interleukins has continued to grow and cells and mononuclear phagocytes, has multiple effects
new members have been included (Table 5). IL-18 is the on both T cells and NK cells. It induces IFN-γ produc-
most recently cloned member of this family [200], tion in T and NK cells and sustains the cell-mediated
which is synthesized as a 24 kDa proform that is pro- immune response [304].
cessed into an 18 kDa mature form. The mature form of Interleukin-21 (IL-21) is a cytokine with structural
IL-18 induces IFN-gamma secretion and plays an and sequence homology to IL-2 and IL-15 that has anti-
important role in antitumor immunity [219]. IL-18- tumor activity alone in experimental tumor models and
transgenic mice show increased CD8+ CD44 high T a tolerable safety profile in phase I trials in patients with
cells and macrophages, while B cells are decreased in metastatic melanoma and renal cell carcinoma [353].
these mice [129]. Similarly, IgE, IgG1, IL-4 and IFN-γ Several monoclonal antibodies targeted at tumor-asso-
levels are substantially increased in the sera of IL-18- ciated antigens also have improved antitumor activities
transgenic mice. In contrast, the IL-18 knock out mice in mice when used in combination with IL-21, suggest-
exhibit impaired clearance of neurovirulent influenza A ing that the use of IL-21 in adjuvant settings induces a
virus-infected neurons from the brain [190]. Another strong T cell-mediated immune responses [353].
relatively newer member of the interleukin family is Interleukin-24 (IL-24) is a cytokine belonging to the
IL-15 that acts as a strong cofactor for Th1 T cell devel- IL-10 family of cytokines that signals through two het-
opment and as a growth factor for T lymphocytes and erodimeric receptors (IL-20R1/IL-20R2 and IL-22R1/
TILs [112]. Like IL-2, it binds to the beta and gamma IL-20R2). IL-24, a recently identified cancer gene thera-
chains of the IL-2 receptor to exert its action [100]. peutic, is melanoma differentiation associated gene-7/
However, specificity for IL-15 versus IL-2 resides in the IL-24 (mda-7/IL-24) that has the unique ability of induc-
unique private α-chain receptors that complete the ing apoptosis in a variety of cancer cells with no toxicity
IL-15Rαβ.g and IL-2αβγ heterotrimeric high-affinity to normal cells or tissues. It exerts immunomodulatory,
receptor complexes and thereby allow differential anti-angiogenic, and radiosensitizing effects when applied
responsiveness depending on the ligand and high affin- as adenovirus expressing mda-7/IL-24 (Ad.mda-7) in
ity receptor expressed [323]. A recent review discusses human xenograft animal models and has been success-
the role of IL-15 in human diseases and its potential fully used in a Phase I clinical trial in patients with
clinical implications as a therapeutic target [70]. Other advanced cancers [347, 354].
important members of the interleukin family include
IL-5, IL-6, IL-7, IL-12, IL-21 and IL-24. IL-5 is an
18-kDa product of T lymphocytes that has been cloned
Colony-Stimulating Factors
and shown to be a lineage-specific eosinophil growth The colony-stimulating factors (CSF) are a family of
and differentiation factor [30, 250]. Murine IL-5 induces glycoproteins that have the ability to induce prolifera-
antibody-dependent killing of tumor cells by blood tion and differentiation of progenitor hematopoietic
eosinophils and enhances phagocytosis by eosinophils. cells and have effects on the functional status of their
Interleukin-6, initially described as β2-interferon, is a mature progeny. Multi-colony stimulating factor (IL-3),
19–28-kDa protein produced by a variety of cells, macrophage colony-stimulating factor (M-CSF), gran-
including mononuclear phagocytes, fibroblasts, kerati- ulocyte colony-stimulating factor (G-CSF), granulo-
nocytes and endothelial cells. The experimental data cyte-macrophage colony-stimulating factor (GM-CSF),
support a potential clinical role for IL-6. Its hematopoi- erythropoietin (EPO), stem cell factor (SCF), and leuke-
etic activity and thrombopietic activity, in particular, mia inhibitory factor (LIF) are some of the clinically
may make this cytokine a useful agent for inducing the important members of this family (Table 3). All the
Kapil Mehta et al. 63

known CSFs have been produced by recombinant DNA dependent increases in hematocrit and hemoglobin
methods and tested in human subjects (Table 2). The levels, and in most cases eliminated the need for regular
potential for using hematopoietic growth factors in the blood transfusions [67]. The major side effect reported
treatment of disease is enormous. Their ability to con- is increased blood pressure. EPO also increases the abil-
trol the production of blood cells has been realized, and ity of patients undergoing elective surgery to donate
the results of clinical trials to date suggest that the side autologous blood [94]. Double-blind placebo-controlled
effects of these growth factors are relatively minor studies with recombinant EPO suggested that it is an
[329]. Three recombinant hematopoietic growth factors, effective treatment for predialysis patients [168].
G-CSF (filgrastim), GM-CSF (Sargramostim), and EPO Stem cell factor (SCF) has recently been used in the
(epoetin alfa), are now commercially available for clinic as a single agent following chemotherapy. SCF by
clinical use in the United States. itself appears to have limited efficacy and significant
Extensive clinical and preclinical data on recombi- toxicity-mainly due to mast cell stimulation at higher
nant human G-CSF and GM-CSF indicate that both doses. However, Tong et al. [300] showed that patients
these cytokines are effective in accelerating neutrophil receiving CSF have an increase in primitive progenitor
recovery and shortening the duration of neutropenia fol- cells suggesting that SCF might be highly effective in
lowing chemotherapy with or without bone marrow combination with later acting hemopoietins. From these
transplantation [48, 77, 185, 193, 206]. Administration data it is clear that recombinant CSFs are effective in
of recombinant human G-CSF as an adjunct to cyclo- correcting hematopoietic disorders of various etiolo-
phosphamide, doxorubicin and etoposide chemotherapy gies. Whether these mediators improve morbidity and
for small cell lung carcinoma significantly reduced mortality in patients will be decided by further clinical
duration and severity of neutropenia and associated results. However, combinations of cytokines, for exam-
clinical sequelae [303]. Similarly, patients with transi- ple, those with relatively restricted biological activity
tional cell carcinoma of the bladder treated with metho- (EPO, G-CSF, M-CSF etc.) and those that have a broad
trexate, vinblastin, doxorubicin and cisplatin experienced range of action (SCF, IL-3, GM-CSF, IL-6 etc.), are
up to fourfold increases in neutrophil count on adminis- likely to show more promising effects on hematopoiesis
tration of G-CSF with few or no toxic effects [84]. than any single cytokine alone [114].
GM-CSF may also exert antitumor effect by inducing
tumoricidal activation of macrophages. Administration
of GM-CSF has been shown to decrease the tumor bur-
Tumor Necrosis Factors
den in a murine Lewis lung carcinoma model [121]. Tumor necrosis factor (TNF) is a proinflammatory
Clinical trials of M-CSF have been performed in an cytokine that is produced primarily by mononuclear
attempt to ameliorate leukopenia. A phase I trial of phagocytes in response to endotoxin. In recent years,
M-CSF in patients with metastatic melanoma showed several new members belonging to the TNF superfamily
an increase in the number and function of circulating of molecules has been described and the list has continued
monocytes [18]. In a non-randomized, controlled study to grow (Table 6). There are two most studied forms of
(32 patients with urinary tract malignancies) and a ran- TNF; TNF-α is a cytotoxic factor with a molecular weight
domized controlled study (98 patients with gynecological of 17-kDa, and TNF-β, also known as ‘lymphotoxin,’ has
malignancies), M-CSF administration reduced the period a molecular weight of 25-kDa and is released from stimu-
of post-chemotherapy leukopenia [192, 207]. lated lymphocytes [102, 203, 204, 223]. Both forms have
Among the cytokines whose role can be predicted been produced by recombinant DNA technology and
from in vitro studies, EPO is perhaps the best example. appear to have antiproliferative, cytostatic and cytolytic
EPO is produced mainly by the kidneys and is respon- effects against human tumor cells in vitro as well as in
sible for regulating the production of erythrocytes. EPO vivo when injected into nude mice [212]. TNF-α exerts
acts on erythroid precursors in the bone marrow, spleen synergistic effects with different cytokines, such as IFNs,
and fetal liver and stimulates the colony formation of IL-1, IL-2 etc., and can induce secretion of series of
the burst-forming unit-erythroid. When infused in mice, mediators, including IL-1, IL-6, prostaglandins etc. It has
EPO markedly increases both peripheral blood erythro- been implicated in both the generation and the cytotoxic-
cytes levels and the number of erythroid progenitor ity of LAK cells and cytotoxic T lymphocytes [37].
cells present in bone marrow. These results led to the Based on these observations, a large number of phase
clinical trials with a human recombinant EPO, the find- I and II studies were initiated to investigate the antitu-
ings suggested that EPO can reverse anemia in patients mor properties of TNF-α. Unfortunately, in clinical
with end-stage renal cell disease. EPO produced dose- settings the efficacy of TNF-α has been very limited,
64 Recombinant proteins and genomics in cancer therapy

Table 6. Members of the TNF family and their receptors for its use. In a recent study, Lienard and co-workers
Cytokine Other names Receptor [167] used an intra-arterial route to administer of high
doses of TNF-α in conjunction with melphalan, hyper-
TNF-α TNFR1 (CD120a);
TNFR2(CD120b) thermia and IFN-γ. Of the 23 patients treated (19 with
LT-α3 (TNF-β) TNFR1,TNFR2 melanoma and four with sarcoma), all responded, 21
LT-β1α2 LTβR with complete remission and two with partial remission.
LIGHT (HVEML) HVEM (TR2, ATAR, Eleven of these patients were previously unresponsive
LIGHTR), LtbR, DcR3
to melphalan alone and one had failed to respond to cis-
OX40L CD134L OX40 (CD134)
4-1BBL CD137L 4-1BB, TNFRSF9, CD137, platin therapy. The toxic effects observed (neutropenia,
ILA hypotension, thrombocytopenia and kidney failure)
CD27L CD70) CD27 were reversible. The overall rate of survival was 70%
CD30L CD153 CD30 and of disease-free survival over 12 months, 76%.
CD40L CD154, TRAP, gp39) CD40
FasL Apo-1L, CD95L Fas (Apo-1, CD95); DcR3
These results suggested that further understanding of
TRAIL Apo-2L TRAIL-R1 (DR4, Apo-2) the mechanisms of TNF-α’s antitumor action could help
TRAIL-R2 (DR5, KILLER) improve its clinical efficacy. For example, decreasing the
TRAIL-R3 (DcR1, TRID) agent’s systemic toxicity without reducing its anticancer
TRAIL-R4 (DcR2, effects could lead to substantial therapeutic advantages.
TRUNDD)
OPG (OCIF) TNF-α mediates its effects by interacting with two dif-
RANK TRANCE, OPGL RANK (TRANCE-R) ferent surface receptors, p55 and p75 [296]. Studies have
OPG (OCIF) suggested that TNF-α’s interaction with p75 may be
TWEAK Apo-3L Fn14 (TWEAK-R), DR3 responsible for its systemic toxicity [118, 296]. Thus,
APRIL TALL-2 BCMA mutant TNF molecules that interact with p55 but not p75
TACI
BAFF (BlyS, THANK, could induce antitumor effects with reduced systemic
TALL-1, zTNF4) TACI toxicity, permitting higher doses of TNF-α. Indeed, such
MCMA mutant human TNF molecules that specifically bind to
GITRL AITRL GITR p55 have already been described and shown to exert
EDA-A1 EDAR
cytotoxic effects against transformed cells in vitro [214].
EDA-A2 XEDAR
? TAJ (TROY); TRAIN In addition, concomitant use of drugs that are able to
VEGI ? decrease TNF-α systemic toxicity could permit use of
? DR6 higher, more effective doses of this cytokine in cancer
75NTR NGFR (p75, NTR) therapy. Combination regimens of TNF-α with other
? RELT
cytokines, concomitant use of TNF toxicity inhibitors
APRIL, A proliferation-inducing ligand; BCMA, B cell maturation
and use of mutant TNF molecules may provide better
antigen; BAFF, B cell activating factor; BlyS, B lymphocyte stimu- clinical outcomes. Moreover, regional therapy with
lator; THANK, TNF homologue that activates apoptosis, NF-kB TNF-α requires further exploration in view of the fact
and JNK; TACI, Transmembrane activator and CAML-interactor; that such regimens have already produced some very
TWEAK, a weak homologue of TNF; HVEM, herpes virus enetery promising results [167].
mediator; LIGHT, homologous to lymphotoxin, shows inducible
expression and competes with HSV glycoprotein D for HVEM;
TRANCE, TNF-related activation-induced cytokine; RELT, receptor Soluble Cytokine Receptors
expressed in lymphoid tissues.
For details, see references [5, 52, 109, 262, 269, 324, 339, 340]. Certain membrane receptors are enzymatically cleaved
from the cell-surface and released into the extracellular
and its use is associated with serious toxic effects [125]. medium in the form of soluble fragments. Soluble recep-
Of 127 eligible patients enrolled in nine different phase tors corresponding to the ligand-binding domains of
II protocols between 1988 and 1990 for the treatment of many polypeptide hormones and cytokine receptors
diverse malignancies, including breast, colon, gastric, have been described (Table 7). The function of soluble
pancreatic, endometrial, and bladder cancers, multiple receptors is not yet known [73, 115]. However, it is
myeloma and sarcomas, only one patient responded likely that this process represents an important mecha-
(response rate, 0.8%), whereas 13% experienced grade nism for regulation of surface expression of such recep-
four or fatal toxic effects. Despite the initial disappointing tors and may determine the effects of cytokines and
results with TNF-α as an antitumor agent, investigators growth hormones on the target cells. For example, the cell
have continued working on new and improved approaches growth, differentiation and immunomodulatory effects
Kapil Mehta et al. 65

Table 7. List of soluble receptors identified of activation of specific proteolytic enzyme or enzymes.
for various cytokines The identity of enzymes involved in proteolytic cleavage
Receptor Reference of the receptors is not known; however, it is a highly
Proteolytic cleavage: regulated process and appears to be controlled by phos-
IL-1R [115, 267, 268, 279] phatases and kinases [4]. The treatment of cells with
IL-2R [274] ligand can also lead to down modulation of the receptors
IL-4R (murine) [74] and their subsequent shedding.
IL-6R [195, 198] The significance of soluble cytokine receptors as a
IL-6R (gp 130) [195, 198, 202] therapeutic modality for treatment of cancer will be
TNFR [12, 13, 88, 139] determined by further research and evaluation. Since
IFN-α R [208] soluble receptors can provide highly specific biological
IFN-γ R [209] inhibitors for cytokines and growth factors, and because
NGFR [60]
the majority of transformed cells require cytokines for
M-CSFR [62]
Hergulin R [161]
their growth and survival, soluble receptors may have
EGFR [108] therapeutic potential as antagonists to cytokine action.
EPOR [21, 196] For example, hematopoietic growth factors are known
PDGFR [299] to maintain the viability of hematopoietic cells through
Alternative splicing: the prevention of apoptosis [332]. Several investigators
IL-4R [191] have reported that autocrine production of hematopoi-
IL-5R (murine) [178] etic growth factors such as IL-1β [82, 104] or GM-CSF
IL-7R [95] [344] supports the growth and survival of acute myeloid
LIFR [159] leukemia (AML) cells in vitro. In contrast, their soluble
GM-CSFR [251]
receptors and receptor antagonist could inhibit the
G-CSFR [79]
growth of leukemic cells including AML [343], chronic
EGFR [228]
c-erbB3 [143] myelocytic leukemia (CML) [68], and juvenile CML
[257]. Receptor proteins for most of the cytokines have
been cloned and expressed [2]. However, the informa-
of cytokines are exerted in response to their binding to tion available on their therapeutic potential in cancer is
specific cell-surface receptors. The presence of soluble very limited.
receptors in the biological milieu may thus promote Like the soluble and membrane-bound forms of cytokine
direct binding of the ligand to the soluble receptor, neu- receptors, the cytokine ligands also exist in these two
tralizing and preventing its action. forms. For example, the cytokines IL-1, TNF, FGF, TGF-
The in vivo relevance of soluble cytokine receptors is α, TGF-β and SCF have been reported to exist in both the
well illustrated by several viruses. Vaccinia and cowpox soluble and membrane-bound forms. This process, com-
viruses encode a protein that displays homology with monly initiated by cell stimulation, may regulate the sur-
soluble IL-1 receptor and is able to bind IL-1β [279]. face expression of such cytokines and play an important
Furthermore, proteins that bind to TNF have been identi- role in determination of cytokine activity.
fied in the open reading frame of pox viral strains [274].
Herpes and myxoma viruses encode proteins that can Genomics and Proteomics in Cancer
effectively bind IL-8 and IFN-γ ligands, respectively [7,
307]. Such soluble receptors assist virus infection by sup-
Therapeutic
pressing host defense mechanisms. From the studies on Advances in the analytical tools and recombinant
induction of antiviral soluble LDL receptors by IFNs, it DNA technology have improved our understanding of
seems that host organisms make use of a similar mecha- the signaling pathways encoded in cytokine, growth
nism for the opposite role of controlling viral infection. factors and hormones [158, 85]. In most cases, the
During the release of cell-surface receptors, it is usu- focus of such investigation is often a single molecular
ally the extracellular domain of the receptor that is shed; target. Compilation and integration of this voluminous
thus, soluble receptors act as inhibitors of cytokines. The literature demonstrates a high level of complexity in
soluble receptors may originate via two separate mecha- cell’s repertoire for the detection and processing of
nisms, one involving alternate splicing in which a receptor molecular signals. Despite all this information, the
gene lacks a transmembrane domain. As an alternative, relative contribution of each and more importantly,
the receptors can be shed from the cell-surface as a result the cross-talk among different pathways that influence
66 Recombinant proteins and genomics in cancer therapy

the process of differentiation, proliferation or apoptosis signal transduction pathways [277]. It is now abundantly
remains unclear. clear that single transcription factors such as NF-κB
Recent advances in the techniques for quantitative which is translocated to the nucleus from cytoplasm in
and comprehensive analysis of all the mRNAs (tran- response to binding of a cytokine to its receptor, acti-
scriptome, ∼40,000 unique molecules [53, 317]) or all vates a large number of genes [19, 255]. Similarly intra-
the proteins (proteome, >40,000 unique proteins) of the nuclear p53 [234, 345] and MYC [41] proteins also
cell, offer a promise for defining the cellular functions affect the expression of large numbers of genes that are
in terms of causative molecular changes. In the follow- important determinants of cell proliferation and cell
ing sections we define some of the basic concepts of death. Such large-scale gene profiling studies begin to
these recent techniques and their early contribution in address the relative importance of individual genes in
our understanding of cancer cells. These early data suggest the determination of cell’s destiny.
considerable molecular heterogeneity in an apparently
homogeneous group of tumors classified by conventional
tools, such as immuno- and histochemistry [92, 9].
Oligonucleotide Arrays for Quantitative
Interpretation of such results offers possible explanation Analysis of >30,000 Unique mRNAs
for the observed differences in the outcome of a specific GeneChips commercialized by Affymetrix offer a reli-
anticancer therapy. In addition, the spectrum of molecu- able tool for the evaluation of distinct mRNAs present in
lar targets for a therapeutic agent can also be defined by extracts of total RNA or mRNA by selection with oligo
the application of global gene expression analysis [181]. dT. The GeneChips are unique with respect to at least
Such treatment specific mRNA profiles offer the oppor- two notable features: the first that they have oligonucle-
tunity to predict diverse effects of anticancer drugs and otide probes prepared by a combination of photolitho-
contribute towards selection and design of tumor spe- graphic technologies and combinatorial chemistry [169].
cific therapies. The second unique feature of GeneChips is the distribu-
The Fig. 4 represents some essential molecular players tion of the probes on the glass slide. There are 16–20
that are involved in gene expression pathways and are pairs of antisense oligonucleotides arrayed on a glass
recruited during changes in cell proliferation, differen- slide for each of ∼30,000 unique mRNAs. Each pair of
tiation and apoptosis initiated by extracellular signals. oligonucleotides contains a set of perfectly matching
mRNAs and proteins are obligatory intermediates in the antisense sequence of 23 nucleotides and a set of identi-
cal sequence of nucleotides with one “mismatch” base at
Cytokines
the 13th nucleotide. Hence each mRNA is probed 16–20
times for specific binding and an equal number of times
Growth factors Micronutrients
for non-specific binding. A comprehensive statistical
analysis of 16–20 pairs of data for each target then evalu-
ates the specific mRNA to be either present or absent and
Inactive activated gives a numerical value for absolute hybridization. A major
transcription factors transcription factors disadvantage of the technique is its inability to detect
Translocation to
low abundance mRNA. For example, in authors’ experi-
?
histone acetylases nucleus ence, the GeneChips were unable to detect mRNAs for
histonedeacetylases
nitric oxide synthase genes that could be detected by
heterochromatin Euchromatin RT-PCR. The application of oligonucleotide arrays to
(inactive) (active)
human cancers have initiated a more detailed compilation
RNA
of molecular changes that are cancer specific [285–328].
Splicing protein
Such a data-base will prove useful in diagnosis and treat-
mRNAs ment of cancers.
Proteins

cDNA arrays
Oligonucleotide arrays
Proteomic analysis cDNA Arrays are Dotted Arrays of PCR-
(2D separation-Mass Spec)
SAGE
Differential display
Amplified Products of Cloned Genes
Subtraction hybridization
cDNA arrays of large numbers of genes were invented
Figure 4. Molecular players involved in gene expression and pioneered in laboratories of Patrick Brown and
pathways David Botstein [28, 256]. Unlike oligonucleotide arrays,
Kapil Mehta et al. 67

cDNA arrays are constructed by robotic dotting of PCR methods for structural determination, and to improve
amplified fragments of cDNA on to glass slides or nylon methods for predicting protein structure. While efforts to
membranes. The cDNA arrays of human genes have improve the crystallographic and NMR techniques have
been used to define disease specific gene profiles. Such continued to receive a major share of intellectual effort,
analyses have contributed towards a better understand- structural prediction studies have received new impetus
ing of cancer and normal cells [9, 10]. The techniques from the development of new models and methods [273].
for constructing cDNA arrays are more readily available The molecular modeling serves several functions: (1) it
to laboratories and tissue or cell-specific arrays can be provides the tool to visualize protein structure in three
constructed in a specialized and dedicated core facility dimensions; (2) it permits protein-to-protein comparisons;
[254]. The application of cDNA arrays to various can- (3) predicts the structure of proteins; and (4) facilitates the
cers cells ex vivo [254] and in vivo [92] have resulted in computer-aided drug design. Such integrated information
a better classification of cancers and will lead to that can define the transcriptome and the proteome will
improved paradigms of anti-cancer drug development help identify novel molecular targets for therapeutic inter-
in the near future. ventions of malignant growth.

Proteomic Analysis and Cancer Antibodies and Conjugates


One major aim of scientists is to understand mutations Antibodies are highly selective proteins that can bind to
that can cause cancer. To accomplish this aim, a thorough a single target among millions of irrelevant sites. Because
understanding of the workhorse of all biological systems, of this specificity, the antibodies have been used exten-
the proteins, is required. Currently, a major effort is being sively to target drugs, prodrugs, toxins and other agents
made to understand protein–protein interactions. There is to particular sites in the body. It is this use of antibodies
an equal emphasis on understanding the structural fea- as targeting devices that led to the concept of “magic
tures of proteins and the structural basis of protein–pro- bullets,” a treatment that could effectively seek and
tein, protein–ligand, and protein–drug interactions. selectively destroy tumor cells wherever they resided.
However, initially lack of techniques that enable compre- The major problem in the therapeutic use of antibodies
hensive and quantitative labeling and separation technol- was their production in large quantities, but the develop-
ogy for proteins, have been limiting factors these efforts ment of ‘monoclonal antibody’ technology changed the
[1]. The separation of cellular proteins by two dimen- situation dramatically [113, 186, 333]. Monoclonal anti-
sional, denaturing gel electophoresis combined with bodies (MAb) are already widely used for the diagnosis
determination of peptide sequence to identify proteins and treatment of cancer and for imaging of tumors for
has been used to define changes in proteins in response to radiotherapy.
oxidative stress in yeast [91]. More recent developments Despite the rapid progress being made in application
of analytical techniques that use novel methods of label- of MAbs as therapeutic agents, their use has been limited
ing and separating large numbers of proteins [1] are because of their immunogenicity problem. MAbs are
beginning to define the pathways of flow of molecular usually mouse proteins; when injected into patients they
information from the genome to the proteome [130]. are eventually recognized as foreign proteins and cleared
Various stable isotope labeling techniques have recently from the circulation. To overcome this problem, research-
emerged to improve the efficiency and accuracy of pro- ers set out to engineer fully “humanized MAbs” that will
tein quantitation by mass spectrometry. We have devel- be indistinguishable from natural proteins [32, 334].
oped a mass-tagging strategy to incorporate stable isotope Humanizing MAbs is a technology that uses recombi-
tagged amino acids into cellular proteins in a residue- nant DNA techniques to improve or change the function
specific manner during cell growth [356]. of these antibodies [233]. The first fully humanized MAb
The major emphasis for new drug development in the recognizes an antigen on the surface of human lympho-
modern era is based on thorough understanding of the cytes and is being evaluated as an immunosuppresant
relationship between the drug and its target protein, and for treatment of lymphoid tumors. Another poten-
whether it is an enzyme, receptor, structural, or transport tially useful humanized MAb recognizes a growth factor
protein. Detailed information regarding the physical, receptor in large numbers on the surface of several breast
chemical, biochemical, genetic, and physiological charac- tumor cells. This MAb successfully inhibited tumor cell
teristics of that relationship must be well understood. growth in culture and is currently being evaluated in
Hence, a major efforts is being devoted to enhance the patients [322]. In the following section, we will briefly
pace and resolution of classical crystallographic and NMR discuss the potential use of Mab-based immunotherapies
68 Recombinant proteins and genomics in cancer therapy

that have been used for the treatment of malignant dis- stroma, or matrix that are necessary for tumor growth or
eases. Detailed aspects of this approach will be discussed metastases. For example, injection of anti-CD44 MAb
somewhere else in this book. or its F(ab’)2 fragment 1 week after inoculation of
human melanoma cells in mice with severe combined
immunodeficiency (SCID) prevented metastases but not
Monoclonal Antibodies as Agonists the development of primary tumor [78]. The antibody
Antibodies directed against cell-surface molecules on had no effect on growth of tumor cells in vitro. MAbs
many types of tumor cells can act as ligands, resulting in against growth factors or their receptors can also exert
powerful antitumor effects mediated by signal transduc- significant antitumor effects. For example, antibodies
tion [320]. For example, MAb 4D5 against erbB-2, against IL-6 and IL-6 receptor were effective in the
when added to breast or ovarian carcinoma cells that treatment of human myeloma in SCID mice [287] and
overexpress erbB-2, induces a strong antiproliferative produced transient responses in patients bearing IL-6-
effect [164]. The erbB-2 protein product is a member of dependent tumors [146]. MAbs against the IL-2 receptor
the EGF receptor family and is shown to act as a signal- have been used to treat adult T-cell leukemia with some
ing receptor for a recently identified ligand, heregulin, partial or complete remissions [322]. Thus MAbs
in regulation of growth and differentiation of breast cancer selected against tumor surface antigens to exert either
and other cell types [47]. Herceptin is a humanized potent growth-inhibitory effects or host-tumor interactions
monoclonal antibody directed against Her-2/neu protein should lead to new strategies for selecting the antitumor
that is abnormally abundant in 25–30% breast cancer activity of Mabs.
patients [141]. In view of the encouraging results
observed in Her-2/neu positive breast cancer, Herceptin
(trastuzumab) was recently approved by the FDA for
Monoclonal Antibodies-Conjugated Drugs
treatment of metastatic breast cancer [165]. Data from The clinical progress with conjugates of Mabs and cyto-
pivotal trials suggested that trastuzumab is active when toxic drugs has been rather slow. An important factor
added to chemotherapy in patients with advanced meta- that has limited the use of this approach for treatment of
static breast cancer [271]. In particular, the combination cancer is the relatively low potency of standard chemo-
significantly prolonged the median time to disease pro- therapeutic agents. The potency of these compounds is
gression, increased overall response rate, increased the further reduced by their conjugation with MAbs [148].
duration of response, and improved median survival However, a recent report that such a conjugate, BR96-
time by approximately 25% compared with chemother- doxorubicin (BR96-Dox), is highly effective in curing
apy alone [271, 22]. As a single agent, trastuzumab xenografted human carcinoma-bearing mice [301], has
induces durable objective response in women with rejuvenated great interest in Mab-drug conjugates. BR96
HER-2 positive metastatic breast cancer and is rapidly is a chimeric monoclonal antibody that contains a frame-
becoming a standard of care for these patients [23]. work region of human immunoglobulin and the binding
Similarly, the ligation of CD95 (APO-1/Fas) protein region of a murine antibody. The antibody binds to an
with an anti-Fas MAb resulted in apoptosis (pro- antigen that is expressed on the surface of many human
grammed cell death) of malignant cells. Using MAbs, carcinomas. Treatment of tumor-bearing athymic mice
the human Fas and APO-1 proteins were identified as with BR96-Dox induced complete regressions and cures
cell-surface proteins of 200- and 48-kDa molecular of xenografted human lung, breast, and colon carcino-
mass, respectively, in two different laboratories. Both mas and cured 70% of mice with extensive metastases
induced apoptosis in a variety of cell types upon binding. from a human lung carcinoma [301]. Clinical trials with
Subsequent isolation of cDNAs encoding the two proteins BR96-Dox were recently initiated to determine its safety
revealed that they were identical despite a difference in in patients. Similar results with Mab-vinblastine conju-
apparent molecular weight [133, 211]. Apoptosis triggered gates were reported years ago, but evaluation of this
by the anti-CD95/Fas/APO-1 Mab has been successfully Mab-drug conjugate was discontinued because of unac-
used for the treatment of mice bearing human hematopoi- ceptable gastrointestinal toxic effects [14, 230].
etic tumors [302, 151]. The clinical use of anti-Fas/ Recently, several groups have concentrated their
APO-1 therapy for cancer treatment will be based on research on more potent immunotoxins conjugated with
further studies. agents such as calicheamicins, maytansines and trichoth-
Besides negative signaling, Mabs have other poten- ecenes. Calicheamicin is a family of antibiotics that pro-
tial uses in tumor therapy. Some MAbs can block inter- duce double-stranded DNA breaks; when conjugated
actions between tumor cells and neighboring cells, with Mab CT-M-01, which recognizes PEM antigen and
Kapil Mehta et al. 69

is located on the surface of human cancerous epithelial invariably fatal leukemia of these mice [307]. It remains
cells, and injected into breast carcinoma-xenografted to be seen whether antibody-drug conjugates will be
mice, it significantly inhibited tumor growth and pro- effective anti-cancer agents in clinical settings as well.
duced long-term tumor free survivors [124]. In a more
recent study, 40 patients with relapsed or refractory
CD33+ AML were treated with humanized anti-CD33
Immunotoxins
antibody linked to the calicheamicin [266]. In eight of Immunotoxins are chimeric molecules in which antibod-
the 40 patients, leukemia was eliminated from the blood ies or the ligand that interacts with the cell-surface mol-
and marrow whereas, in 3 additional patients the blood ecules are coupled to toxins or their subunits (Fig. 5).
counts returned to normal. Similarly, patmaytansinoids, The antibody or growth factor binds with high selectivity
which are 100 to 1,000-fold more potent than doxorubi- to the target cells. The toxins are derived from plants or
cin and vinblastine, when conjugated to Mab A7, which bacteria. DNA sequences encoding the bacterial toxins;
recognizes an antigen expressed on human colon cancer Pseudomonas exotoxin (PE) and diphtheria toxin and
cell lines, showed high antigen-specific in vitro cytotox- the plant toxins; ricin and gelonin have been cloned and
icity against cancer cells and low systemic toxicity in expressed in E. coli, [2, 221]. The topic of immunotoxins
mice [34]. Similar specificity and potency have been is discussed in detail elsewhere in this book, therefore,
observed in Mab-trichothecenes conjugates (protein syn- we will concentrate only on the therapeutic potential of
thesis inhibitors) has been observed in terms of their recombinant immunotoxins in this section.
tumor cell-killing ability [189]. Initially, the toxins produced in bacteria were chemi-
More recently, Mabs have been used to deliver enzyme cally linked to antibodies to make immunotoxins. In recent
inhibitors to tumor cells. Thus, conjugation of Geninstein years, significant progress has been made in engineering
(an inhibitor of Src protooncogene family protein tyrosine recombinant immunotoxins by fusing the cell-binding
kinases) to MAb B43, which recognizes the B cell- ligand genes to modified toxin genes [221]. For example,
specific receptor CD19, selectively bound to B-cell pre- a truncated form of Pseudominas exotoxin (PE40) has
cursor leukemia cells, inhibited CD19-associated tyrosine been produced by deleting the first 252 amino acids; this
kinases and triggered rapid apoptotic cell death [307]. toxin has extremely low toxicity because of its inability to
Treatment of B-cell precursor leukemia-xenografted bind to cellular receptors [136]. However, when chemically
SCID mice with less than one tenth the maximum toler- conjugated to an antibody [76, 156] or the recombinant
ated dose of B43-Geninstein resulted in more than chimeric toxin generated by fusing the PE40 gene to DNA
99.999% killing of human leukemia cells, which led to fragments encoding growth factors, antibody-binding
100% long-term even-free survival from an otherwise sites, or other target-recognition elements, Pseudomonas

Immunotoxin (IT)

Toxin
Receptor Ligand (Ricin, Pseudomonas
exotoxin, gelonin etc.)
Cell surface MAb or
antigen or cytokine A chain B chain
Cytokine hormone
Receptor
Killing Translocation Native
Domain enhancing domain binding
domain

TUMOR CELL NORMAL CELL

Figure 5. Schematic representation of immunotoxin


70 Recombinant proteins and genomics in cancer therapy

exotoxin becomes highly specific and potent in killing that express IL-6 receptors at high numbers and also
target cells [220, 221]. The gene encoding this chime- several other carcinomas [150, 263, 264]. The first clinical
ric toxin is expressed in E. coli. Table 8 lists some of trial of a genetically engineered immunotoxin (B3LysPE38)
the recombinant immunotoxins that have been pro- was initiated in April 1993 in breast and colon carci-
duced by fusing the PE40 gene to cDNAs encoding noma patients. Some responses were evident, but toxic
different targeting molecules. Anti-Tac(Fv)-PE40 is effects appeared to be greater in human patients than
one such recombinant immunotoxin that was gener- seen in mice and primates [270].
ated by fusing the truncated form of the Pseudomonas In contrast to Pseudomonas exotoxin-immunotoxins,
exotoxin (PE40) gene with the variable region of an the immunotoxins composed of ricin or its A subunit and
antibody against the IL-2 receptor [36, 155]. This MAbs have generally been constructed using chemical
immunotoxin is highly toxic to cells from patients with cross-linking reagents [89, 173]. Recombinant ricin-
adult T-cell leukemia and induced regression of IL-2 based chimera molecules have been difficult to produce,
receptor-bearing carcinoma tumors in athymic mice because the A chain of the plant toxins must be attached
[66, 153, 154]. TGFα-PE40 recombinant chimera toxin to the cell recognition domain by a disulfide bond, and
targets PE40 to cells with EGF [66, 258]. Although disulfide-linked subunits are difficult to produce in bac-
many normal cells contain EGF receptors, tumor cells teria [238]. Many reviews have already described the
often have extremely large number of receptors because activities and properties of immunotoxins made with
of amplification and overexpression of the EGF recep- ricin and other plant toxins [89, 142, 249, 308, 315, 320].
tor gene [238]. When administered systemically, Ricin-containing immunotoxins have been used to elimi-
TG&Fbdot;α-PE40 caused regression of subcutaneous nate selected populations of lymphocytes. Vitetta, Uhr
epidermoid carcinoma and prostate carcinoma tumors and associates have produced ricin conjugates of anti-
in mice [221]. bodies to B cell-specific antigens and shown such conju-
Interleukin-2-PE40 is a recombinant chimeric protein gates to cause complete regression of B-cell lymphomas
designed to deliver the toxin to cells with IL-2 receptors in mice [80]. Significant antitumor activity of ricin A
[172]. Normal resting lymphocytes do not express IL-2 chain immunoconjugates has been observed against solid
receptors, but when they are activated with an antigen or or ascites tumors in animal models [75, 105].
IL-2, the receptors are induced. IL-2-PE40 has been Because of encouraging results in preclinical studies,
shown to be highly toxic to activated mouse and rat T several ricin-containing immunoconjugates have been
cells and had some therapeutic effect against mouse developed and approved for human trials, and two kinds
lymphoma [150]. Similarly, IL-6-PE40 chimeric toxin of human trials have been conducted. The first involves
killed many human myeloma and hepatoma cell lines the ex vivo treatment of harvested bone marrow to elimi-
nate contaminating tumor cells prior to re-infusion in
patients undergoing autologous bone marrow transplan-
tation. The second kind of trial involves the parenteral
Table 8. Recombinant toxins derived from Pseudomonas administration of immunotoxins [231, 308, 315, 320].
exotoxin Some patients with B-cell lymphoma responded to immu-
Immunotoxin Target Reference notoxin therapy [89]. Currently, an anti-CD22 dgA immu-
notoxin is being evaluated in phase II clinical studies in
PE40
lymphoma patients.
Anti-Tac (Fv)-PE40 Human IL-2 receptor [36,
(leukemia) 153–155] The side effects observed with administration of immu-
TGF-α-PE40 EGF receptor, [66, 113, notoxins are different from those of conventional chemo-
epidermoid carcinomas 258, 252] therapy; immunotoxins do not exert cytotoxic effects
adenocarcinomas, against normal rapidly dividing cells. Immunotoxins such
glioblastomas smooth as the bacterial toxins, Pseudomonas exotoxin and diph-
muscle cells theria toxin, induce hepatotoxicity, whereas the ricin-
IL-2-PE-40 IL-2 receptor (leukemia) [150, 172]
based immunotoxins cause reversible vascular leak and
IL-6-PE40 IL-6 receptor myelomas, [264, 265]
hepatomas, prostate myalgias [320]. Several groups of researchers are cur-
B3(dsFv)-PE38KDEL Many carcinomas [25, 26] rently working on second- and third-generation immuno-
e23(Fv)PE40 erbB2 toxins to eliminate the immunogenicity and side effects
lung, breast, ovary and [20] of the first generation immunotoxins. Continued refine-
stomach ment in design of these pharmaceuticals may eventually
adenocarcinomas prove useful in the treatment of cancer.
Kapil Mehta et al. 71

Monoclonal Antibodies and elicits the biological effects of both its component cytok-
ines [29, 311, 312]. Thus, PIXY321 became the first
Radioimmunotherapy
recombinant fusion of two hematopoietins to enter the
The use of MAbs to deliver radioisotopes for treatment of clinic. Early clinical experience has shown great potential
certain cancers has yielded some encouraging results [106, in the prevention and treatment of hematopoietic suppres-
137, 138]. Some of the most promising of these results sion. The other recombinant fusion proteins have been
radioimmunoconjugates in bone marrow transplantation. made, including chimeric toxins constructed by fusion of
In the treatment of leukemia, radioiodine-MAb conjugates genes encoding human cytokines and Pseudomonas exo-
ablate marrow safely, delivering up to fourfold more radi- toxin [35, 55, 284]. For example, IL-4-PE, a chimera of
ation to the marrow than to other normal tissues. Responses human IL-4 and Pseudomonas exotoxin proteins, is
have been less frequent in solid tumors treated with radio- highly potent against many cancer cells [55], suggesting
immunoconjugates in clinical trials [183, 184]. Objective that it might be useful in the therapy of many cancers.
tumor regressions, however, were seen in four patients Generation of homologues and analogues of natural
with non-bulky disease who were treated with rhenium- biotherapeutic proteins is another potentially important
186-labeled Mab NR-LU-10 [134]. Because of their size application of recombinant DNA technology. The tech-
and high molecular weight, diffusion of MAbs within nology involves alteration or deletion of key nucleotide
bulky tumors can be a problem, as illustrated by the results sequences in the gene that will result in the modification
of a clinical trial that used radiolabeled anti-CD20 and of only a few amino acid sequences in the resultant pro-
anti-CD21 MAbs to treat patients with B-cell lymphomas tein, compared with the natural protein. Synthesis of
[236, 253]. Among the most promising approaches to novel human TNFα, IFNα and IF&Nbdot;γ homologues
overcoming this problem is the use of genetically engi- has already been reported [11, 214]. All three of these
neered, low molecular weight F(ab’)2, Fab and single- homologues are distinct from their parent protein.
chain Fv fragments that may diffuse better at tumor sites. Prolonged activity of insulin has been achieved by various
Early results of clinical trials employing humanized MAbs substitutions that increase the isoelectric point. The pro-
appear encouraging. In addition to reduced immunogenic- longed activity seems to occur because the novel homo-
ity, F(ab’)2, Fab and Fvs, with their improved ability to logues precipitate when they encounter the neutral pH of
penetrate tumors, may prove useful carriers for radiother- the body [180]. These examples indicate an interesting
apy. Newer regimens combining radioantibody-based way by which protein modifications can be exploited for
therapy with other treatment modalities may prove even therapeutic potential.
more effective. Furthermore, the ability afforded by newer techniques
in recombinant DNA technology such as “phage display”
to correlate protein structure and function in a systematic
Chimeric Proteins
way makes it possible to design novel drugs [38, 51,
One interesting aspect of recombinant DNA technology 176]. Such a technique could be used to design or even
is the potential for producing of new proteins with novel select a small peptide that binds to the receptor with
properties. For example, the hybrid proteins formed by the same affinity as the larger protein. And then, using
fusion of two or more genes (chimera) offer several computer modeling to display the molecular contacts
advantages in terms of their stability, affinity, efficacy and between ligand and receptor, small nonprotein mole-
pharmacology over the individual component proteins. cules that make the same contacts could be designed
A chimera protein formed by fusion of the IFN-γ and LT and synthesized. The end product would be a small
genes was shown to have better antiproliferative activity organic molecule that could be produced more cheaply
than IFN-γ or LT alone [71]. Similarly, PIXY321, a than the recombinant protein, yet would retain the full
genetically engineered hybrid of the GM-CSF and IL-3 biological activity of the protein hormone. What’s more,
proteins exhibits greater colony-stimulating effects such molecules could be administered orally, eliminat-
in vitro than the combination of GM-CSF and IL-3 [50]. ing the major disadvantage of most recombinant protein
In preclinical studies, PIXY321 has been shown to accel- therapeutics, which must be delivered directly into the
erate both neutrophil and platelet recovery in rhesus mon- blood stream by injection.
keys subjected to sublethal irradiation [330]. Because of
the preclinical observations, PIXY321 was tested in
patients for its ability to ameliorate disease- or treatment-
Cancer Vaccines
related bone marrow suppression. The clinical results The issue of genetically engineered vaccines for cancer
were encouraging and suggested that the hybrid protein treatment will be discussed in detail in another part of
72 Recombinant proteins and genomics in cancer therapy

this book. We will, therefore, focus only on the novel specific cell-surface antigens, and monoclonal antibody
aspects of recombinant vaccines that may have some technology permitted identification of tumor-associated
potential as cancer therapeutics. The function of a vaccine antigens, their characterization and tissue distribution.
is to give the immune system a boost, thus helping it The polymerase chain reaction (PCR) brought the tech-
recognize and destroy the ‘non-self’ antigens on the sur- nology of cloning and expressing gene products. These
face of cancer cells. Though the idea of inciting the technologies jointly led to the development of the sub-
immune system to fight cancer has been around for a unit vaccines of the 1990s.
long time, recent developments in biotechnology and The human melanoma antigen MAGE-1 was the first
better understanding of the immune-system network tumor-specific antigen identified [315]. Poxvirus con-
have caused an explosion of research and development taining the MAGE-1 gene is currently being evaluated
in the field of cancer vaccines. Table 9 lists some cancer as a candidate vaccine for treatment of melanoma and
vaccines that are currently undergoing clinical trial. breast cancers in humans [163]. Similarly, the recombi-
Prior to the advent of recombinant DNA technology, nant vaccinia virus containing carcinoembryonic anti-
two types of vaccines were used, inactivated vaccines gen (CEA) is being evaluated for treatment of certain
(chemically killed derivatives of actual infectious agents) cancers. CEA is expressed on the surface of virtually all
and attenuated vaccines (actual infectious organisms colorectal cancers, 70% of lung cancers, and 50% of
altered so that they do not multiply). However, these breast cancers. Clinical results if these phase I trials of
types of vaccines are potentially dangerous, as they can this vaccine in late-stage cancer patients were promising
carry over the infectious contaminations. For example, a [152]. A recombinant fusion of a tumor-derived idio-
small number of children each year contract polio from type and GM-CSF, yielded a strongly immunogenic
their live polio vaccinations. Thus, one of the most prom- protein that was capable of inducing idiotype-specific
ising applications of recombinant DNA technology is the antibodies and protected the recipient animals from
production of subunit vaccines, consisting solely of the challenge with an otherwise lethal dose of B-cell lym-
surface protein to which the immune system responds phoma [295]. These results can be applied not only to
and thus eliminating the risk of infection [27]. The arrival B-cell lymphoma but perhaps can be generalized to
of biotechnology in the late 1970s enabled targeting of other classes of tumor antigens as well.
The discrete peptide fragments from certain tumor-
Table 9. Cancer vaccines under clinical trials specific oncoproteins (such as mutant p53 and the pro-
tein products of the ras and HER-2/neu genes) are rapidly
Immunogen Target Cancer Reference
progressing as potential vaccine candidates for cancer
Recombinant vaccinia Colorectal, lung, breast [152] treatment [72, 342]. This strategy is based on the princi-
encoding for CEA ple that intracellular proteins are degraded and presented
Gene therapy using Renal cell, carcinoma, [152, 287]
back on the cell surface as small peptides in the groove
patient’s own cells melanoma prostate,
colorectal of class I major histocompatibility complex (MHC) anti-
Recombinant poxvirus Melanoma [315] gens. A nine amino acid peptide from the HER-2/neu
encoding for MAGE oncoprotein was shown to be recognized by both breast
antigen and ovarian cytotoxic T lymphocytes, and this small
Heat shock protein Melanoma [163] peptide was able to induce a tumor-specific immune
Naked DNA Lymphoma, Cervical [163, 232, response [224]. It is now possible to isolate and custom
prostate, 346]
Prostate, Melanoma,
synthesize tumor-specific immunogenic oncopeptides
Renal, Colorectal by using the patients’ own mutation to prepare an autolo-
Synthetic peptides Melanoma, Cervical [72, 224, gous peptide vaccine that is selectively targeted to tumor
342] cells containing the mutant gene product [342]. The abil-
Synthetic antigens Ovarian, Breast, [44, 163] ity of human oncopeptide vaccines to generate a peptide-
Melanoma, specific CD8+ cytotoxic T-lymphocyte response in
Colorectal
animal models has formed the rationale for clinical trials
Anti-idiotypic Melanonoma, Colorectal, [278, 295]
antibodies gastric, ovarian of autologous peptide immunizations in patients with
Inactivated tumor cells Colon [163] diverse epithelial malignancies (breast, lung, gastroin-
with the cytokine IL-2 testinal) that are commonly accompanied by p53 and ras
Recombinant antigens Colorectal, lung [63, 119, mutations [142]. It is likely that similar considerations
prostate 278] will apply to other recently identified oncoproteins such
Gene transfer Melanoma [163] as the product of the BRCA-1 gene in breast cancer.
Kapil Mehta et al. 73

Recently, there have been several attempts to gener- Ultimately, as we discussed earlier in this chapter, devel-
ate tumor cell vaccines engineered to secrete various opment of multiple strategies that could be applied in
cytokines [42, 43, 127, 235]. The strategy seeks to alter synergy are most likely to yield beneficial results in cancer
the local immunologic environment of the tumor cell so treatment and control. It remains to be seen what place
as to enhance either the antigen presentation of tumor- the immunotherapy will have in this armamentarium.
specific antigens to the immune system or the activation However, it is obvious that availability of recombinant
of tumor-specific lymphocytes. Many cytokine genes DNA technology has made it possible to design vaccines
have been introduced into tumor cells with varying on a molecular basis.
effects on both tumorigenicity and immunogenicity.
Some of these cytokines, when produced by tumor,
induce a local inflammatory response that results in
elimination of the injected tumor. The local inflamma- Problems Unique to
tory response is, in general, dependent on leukocytes
other than classical T cells. Many cytokine genes have
Recombinant Biotherapeutics
been introduced into tumor cells, including IL-1α, IL-2, The development of protein therapeutics by using recom-
IL-4, IL-5, IL-6, IL-7, IL-12, GM-CSF, IFNα, IFNγ and binant DNA technology has presented many new and
TNFα [42, 215]. Preclinical data from various animal interesting challenges to pharmacologists and drug deliv-
models suggest that tumor cells engineered to produce ery scientists. Many of these biotherapeutics have multiple
cytokines indeed provide a novel approach for tumor biological effects [6]; thus, an important priority in their
therapy [42, 107, 216]. Clinical trials are currently in development is the evaluation of their potency, pharmaco-
progress to assess the therapeutic efficacy of cytokine- logical profile and toxic effects. One strategy to under-
transduced tumors as vaccines for the treatment of stand the potency and toxicity of anticancer agents is the
established solid tumors. use of appropriate animal models. However, many animal
A remarkably straightforward and potentially useful models that have been developed for testing conventional
approach in the field of cancer vaccines was recently low molecular-weight drugs may not be useful for testing
developed. It involves the direct in vivo delivery of rDNA-derived therapeutic proteins. Species specificity of
MHC-associated tumor antigens to provoke a tumor- protein therapeutics further narrows down the choice for
directed immune response. This approach for treatment appropriate animal models. Certain in vitro biological prop-
of diverse malignant diseases is currently under clinical erties of the interferons for example, did not translate to their
investigation (Table 9). In essence, this approach is efficacy in intact animals or in patients [280].
based on the ability of some viral and human ‘naked The lack of information on preclinical pharmacological
DNA’ genes to transfect certain cells without the need behavior also limits a general analysis of the toxic effects
for elaborate genetic engineering maneuvers, sometimes of therapeutic proteins. For example, agents such as IFÑγ,
using modified DNA/liposome complexes to deliver IFN-α and IL-2 are produced in E. coli and are nongly-
genes by direct injection at the tumor site or by systemic cosylated. EPO, in contrast, is produced in Chinese ham-
administration [232, 346]. For example, injection of ster ovary cells and is glycosylated. Rats, dogs, hamsters
naked DNA or mRNA transcripts encoding human CEA and monkeys were used to study the toxicity of these
in mice has been shown to elicit strong cytotoxic agents. Comparison of results on gross morphology,
T-lymphocytes and antibody responses against this anti- histology, blood chemistry and hematology obtained
gen [44, 45]. The development of this simple and direct from these studies demonstrated no general responses
approach for in vivo gene therapy-based immunotherapy that might be attributed to the use of biotherapeutic
is an extremely important avenue for continued clinical proteins. The effects observed were primarily related to
and preclinical investigations. the known or anticipated biological effects of the agent
Most tumor vaccines must be employed subsequent and, as expected, occurred only in the species in which
to the development of cancer. However, in some instances, the agents were known to be biologically active.
for example, in those geographic areas in which human Considering these results, it is apparent that toxicologic
papilloma virus infection is highly endemic and thus findings in animals essentially reflect the pharmacologi-
rates of cervical carcinoma are high, it may be useful to cal effects of biotherapeutic agents. Therefore, the use
vaccinate children prophylactically. In China, where of biotherapeutic agents in species in which they are
endemic hepatitis virus is causally related to hepatic active should be most informative. However, observa-
cancer, broad immunization to prevent hepatitis and tions of the lack of toxic effects in other species may be
subsequent cirrhosis and hepatic cancer is underway. important in addressing some concerns about nonspecific
74 Recombinant proteins and genomics in cancer therapy

toxic effects. Cross-species activity may be seen for These considerations have led to the development of dif-
some if not all biological effects. For example, human ferent strategies to prolong the bioavailability of thera-
IFN-α has pyrogenic activity in rabbits. Clearly, the peutic proteins.
pharmacological effects observed with materials that Conjugation of certain proteins to synthetic polymers
lack pronounced species specificity are likely to be more can circumvent the problems of rapid clearance from the
dependable, especially if different species have mani- circulation, immunogenicity and instability. The general
fested the same toxicity profile. Similarly, human TNF requirements of any polymer used for this purpose are
was found to be less toxic in mice than in human sub- that it be water-soluble, biocompatible, nonimmuno-
jects because one of the receptor subunits with which genic and devoid of biological activity. Zoladex® and
TNF interacts is species-specific and the other is not. In Nafarelin®, the decapeptide agonists of luteinizing hor-
contrast, the pharmacological effects of agents like EPO mone releasing hormone (LHRH), have been formulated
are highly cell type and species specific. in slow-releasing polymer base and used effectively in
There have been relatively few preclinical studies of clinical studies [81, 117]. Attempts have also been made
the immunogenicity of recombinant therapeutic pro- to stabilize the proteins against degradation at the site of
teins. The useful information has come from the clinical injection as well as in the circulation, but these studies
studies. The generation of antibodies to proteins admin- are still preliminary in nature [160].
istered over long periods may result in formation of Covalent conjugation of certain proteins with water-
soluble immune complexes. These immune complexes soluble polyethylene glycol (PEG) enhances their solu-
can induce vascular and tissue injury, particularly glom- bility and permits the design of stable formulations
erulonephritis [281]. As an alternative, immune com- suitable for clinical use. This is particularly important for
plexes can elicit the release of inflammatory mediators recombinant proteins produced in E. coli that are usually
from cells. However, to date there have been no exam- recovered as insoluble refractile bodies, and unlike many
ples of antibody responses to any recombinant therapeu- of their native counterparts, are not glycosylated. For
tic protein that have been shown to clearly cause clinical example, conjugation of both IL-2 and IFN-β with PEG
pathologic effects. increased their solubility, and aqueous solutions were
Another potential problem is incorporation of the stable for long periods of time [144]. Moreover, a recently
wrong amino acids when a high level of expression of published study revealed that PEGlated IFN-α is better
recombinant proteins is enforced. Such errors are gener- tolerated and may be more effective in treating CML
ally difficult to pinpoint, since current analytical meth- patients [291]. Similarly, PEGylation of TNF-α alters its
ods for amino acid composition and sequence are not pharmacokinetics and reduces its in vivo toxicity [144].
really amenable to detecting variations below 10% of G-CSF conjugated to PEG has a four times slower clear-
the major constituents. It is conceivable that such altered ance rate in rats than unmodified G-CSF. In addition,
sequences may resemble a toxic peptide or a protein PEGylated G-CSF administration exerted a sustained
with different biological functions. Also, the altered biological effect on peripheral blood neutrophils [294].
sequences in the protein may render them immunogenic PEGylated adenosine deaminase (ADA), an enzyme
and may provoke enhanced immunogenic responses. unrelated to cancer, is now approved for use as replace-
ment therapy for severe combined immunodeficiency
diseases that are due to inherited ADA deficiency.
Delivery of Therapeutic Proteins Patients who received PEG-ADA did not develop neu-
The potential use of therapeutic proteins in medicine is tralizing antibodies to ADA activity [33].
severely limited because of their poor activity when The key factor with any drug delivery system is to
administered orally. Proteins are rapidly degraded by achieve adequate concentrations of the drug at the desired
proteolytic enzymes in the gastrointestinal tract and have sites while avoiding significant concentrations at sites
been primarily administered by injection. Moreover, that mediate toxic effects. Novel delivery systems such
proteins are generally characterized by short biological as liposomes may prove to be useful in achieving this.
half-lives in the circulatory system, so that the repeated The ability of IFN-γ to stimulate the tumoricidal activity
injections are generally needed. Even after intramuscular of monocytes was increased 1,000-fold by its encapsula-
or subcutaneous administration, their bioavailability is tion in liposomes [147]. Encapsulation of TNF-α in lipo-
often low because of their small size and the widespread somes ameliorated the systemic toxicity of this cytokine
distribution of proteolytic enzymes. In addition, most in dogs [171]. Anti-HER2 immunoliposomes, consisting
proteins pass through biological barriers rather poorly of long circulating liposomes linked to anti-HER2 MAb
because of low diffusion and a low partition coefficient. fragments proved highly efficient for intracellular delivery
Kapil Mehta et al. 75

of the drugs and showed superior anti-tumor activity Acknowledgements The research in authors’ laboratories
in animal models [218]. Delivery can also be modified was supported in part by a grants from Food and Drug
by a combination of the biotherapeutic protein with an Administration, Clayton Foundation, and Department of
Defense.
antibody [61]. For example, the in vivo clearance of
human IFN-α in rats is threefold slower when it is
combined with a specific MAb [246]. However, it
remains to be seen whether such delivery systems will
confer any advantages to biotherapeutic proteins in vivo References
in terms of local delivery, reduced toxicity or altered 1. Aebersold R, Rist B, Gygi SP. Quantitative proteome analysis:
pharmacokinetics. methods and applications. Ann N Y Acad Sci 2000;919:33–47.
2. Aggarwal BB, Gutterman JU, eds. Human Cytokines: Handbook
for Basic and Clinical Research, Boston, MA, Blackwell, 1992.
3. Aggarwal BB, Pocsik E. Cytokines: From clone to clinic. Arch
Biochem Biophys 1992;292:335–359.
4. Aggarwal BB, Puri R. Common and uncommon features of cytokines
Conclusions and cytokine receptors: an overview. In: Human Cytokines: Their
Role in Disease and Therapy (Aggarwal BB, Puri R, eds), Cambridge,
It is clear that recombinant DNA technology has pro- MA, Blackwell, 1994:pp3–24.
5. Aggarwal BB.Tumour necrosis factors receptor associated signal-
duced a revolution in the field of cancer therapeutics. ling molecules and their role in activation of apoptosis, JNK and
The dream of biological therapy, thought to have a great NF-kappaB. Ann Rheum Dis. 2000;59 Suppl 1:i6–16.
potential for cancer, can now be realized. The ability to 6. Aggarwal BB, Puri R. Common and uncommon features of cytok-
ines and cytokine receptors: an overview. In: Human Cytokines:
manufacture cancer drugs by using genetically engi- Their Role in Disease and Therapy (Aggarwal BB, Puri R, eds),
neered organisms has given rise to a novel biotechnol- Cambridge, MA, Blackwell, 1994:pp3–24.
7. Ahuja SK, Murphy PM. Molecular piracy of mammalian interleu-
ogy industry within the last decade that has earned as kin-8 receptor type B by herpes virus saimiri. J Biol Chem
much as $15 billion. The revolution has been not only in 1993;268:20691–20694.
the industrial sector; but also in the academic sector. It 8. Akira S, Taga T, Kishimoto T.Interleukin-6 in biology and medicine.
Adv Immunol, 1993;54: 1–78.
has enabled scientists to discover new molecules, rede- 9. Alizadeh AA, Ross DT, Perou CM, van De Rijn M. Towards a
sign molecules for lower toxicity and more efficacy, and novel classification of human malignancies based on gene expression
investigate the pathology of disease at the molecular patterns. J Pathol 2001;195:41–52.
10. Alizadeh A, Eisen M, Davis RE. The lymphochip: a specialized
level. This technology, however, has given rise to new cDNA microarray for the genomic-scale analysis of gene expression
set problems in the area of drug manufacturing and in normal and malignant lymphocytes. Cold Spring Harb Symp
delivery. One of the major problems in the manufactur- Quant Biol 1999;64:71–78.
11. Alton K, Stabinsky Y, Richards R, et al. Production, characteriza-
ing area is ensuring that the recombinant biological tion and biological effects of recombinant DNA derived human
product is identical or very similar to the natural coun- IFN-α and IFN-α analogues. In: The Biology of Interferon System
terpart. Another problem is the rapid degradation of pro- (De Maeyer E, Schellekens H, eds), Elsevier, North Holland,
Amsterdam 1983;pp119–127.
teins in the circulation and their short circulating 12. Ambrus JL, Pippin J, Joseph A, et al. Identification of a cDNA for
half-lives, which restrict oral delivery of biotherapeutic a human high molecular weight B-cell growth factor. Proc Natl
proteins and may require continuous injection. Acad Sci USA 1993;90:6330–6334.
13. Andus T, Gross V, Holstege A, et al. High concentrations of soluble
It is now possible to design from the crystal structures tumor necrosis factor receptors in ascites. Hepatology 1992;16:
of the ligand and the receptor small molecules that can 749–755.
14. Apelgren LD, Zimmerman DL, Briggs SL, Bumol TF. Antitumor
mimic these large proteins and thus may help to over- activity of the monoclonal antibody-vinca alkaloid immunoconju-
come delivery problems. Some of these problems might gate LY203725 in a nude mouse model of human ovarian cancer.
be circumvented by directly injecting the gene for a Cancer Res 1990;50:3540–3544.
15. Arai K, Lee F, Miyajima A, et al. Cytokines: Coordinators of
given protein: although this approach seems attractive, immune and inflammatory responses. Annu Rev Biochem 1990;
it also suffers from the delivery and organ/cell-specific- 59:783–836.
ity problems. Treatment with antisense DNA and RNA 16. Arai N, Nomura D, Villaret D, et al. Complete nucleotide sequence
of the chromosomal gene for human interleukin 4 and its expres-
to inhibit the expression of oncogenes in tumor cells and sion. J Immunol 1989;142: 274–282.
the new technologies based on viral vectors for the 17. Arend WP. Interleukin-1 receptor antagonist. Adv Immunol.
delivery of vaccines and genes may find widespread 1993;54:167–227.
18. Bajorin DF, Cheung NKV, Houghton AN. Macrophage colony
application in the near future. Many of these approaches stimulating factor: biological effects and potential applications for
work effectively in the test tube, and the main challenge cancer therapy. Semin Hematol 1991;28:42–48.
now is to translate these laboratory techniques into com- 19. Baltathakis I, Alcantara O, Boldt DH. Expression of different
NF-kappaB pathway genes in dendritic cells (DCs) or mac-
mercially viable processes to produce active, effective rophages assessed by gene expression profiling. J Cell Biochem
biotherapies with acceptable toxicity. 2001;83:281–290.
76 Recombinant proteins and genomics in cancer therapy

20. Batra JK, Kasprzyk PG, Bird RE, Pastan I, King RC. Recombinant 42. Colombo MP, Forni G. Cytokine gene transfer in tumor inhibition
anti-erbB2 immunotoxins containing Pseudomonas exotoxin. and tumor therapy: where are we now? Immunol Today
Proc Natl Acad Sci USA 1992;89:5867–5871. 1994;15:48–51.
21. Baynes RD, Reddy GK, Shih YJ, Skikne BS, Cook JD. Serum 43. Colombo MP, Modesti A, Parmiani G, Forni G. Local cytokine avail-
form of the erythropoietin receptor identified by a sequence-specific ability elicits tumor rejection and systemic immunity through granu-
peptide. Blood 1993;82:2088–2095. locyte-T lymphocyte cross talk. Cancer Res 1992;52:4853–4857.
22. Baselga J. Clinical trials of Herceptin (R) (trastuzumab). Eur J 44. Conry RM, LoBugilio A, Wright M, et al. Charaterization of a
Cancer 2001; 37:S18–S24. messenger RNA polynucleotide vaccine vector. Cancer Res
23. Bell R. Duration of therapy in metastatic breast cancer: manage- 1995;55:1397–1400.
ment using Herceptin. Anti-Cancer Drugs 2001;12:561–568. 45. Conry RM, LoBuglio AF, Kantor J, et al. Immune response to a
24. Blumberg H, Conklin D, Xu WF et al. Interleukin −20: discovery, carcinoembryonic antigen polynucleotide vaccine. Cancer Res
receptor identification, and role in epidermal function. Cell. 1994;54:1164–1168.
2001;104:9–19. 46. Cosman D, Lyman SD, Idzerda RL, Beckmann MP, Park LS,
25. Brinkmann U, Pai LH, FitzGerald DH, Willingham MC, Pastan I. Goodwin RG, March CJ. A new cytokine receptor superfamily.
B3(Fv)-PE38KDEL, a single chain immunotoxin that causes com- Trends Biochem Sci. 1990;7:265–270.
plete regression of human carcinoma in mice. Proc Natl Acad Sci 47. Coussens L, Yang-Feng TL, Liau YC, et al. Tyrosine kinase recp-
USA 1991;88:8616–8620. tor with extensive homology to EGF receptor shares chromosomal
26. Brinkmann U, Reiter Y, Jung SH, Lee B, Pastan I. A recombinant location with neu oncogene. Science 1985;230:1132–1139.
immunotoxin containing a disulfide-stabalized Fv fragment. Proc 48. Crawford J, Ozer H, Stoller R. Reduction by granulocyte colony
Natl Acad Sci USA 1993;90:7538–7542. stimulating factor of fever and neutropenia induced by chemo-
27. Brown F. From Jenner to genes-the new vaccines. Lancet therapy in patients with small cell lung cancer. N Engl J Med
1990;335:587–590. 1991;325:164–170.
28. Brown PO, Botstein D. Exploring the new world of the genome 49. Cruikshank WW, Kornfeld H, Center DM. Interleukin-16. J
with DNA microarrays. Nat Genet 1999;21:33–37. Leukoc Biol. 2000;67:757–766.
29. Broxmeyer HE, Benninger L, Cooper S, Huge N, Benjamin RS, 50. Curtis BM, Williams DE, Broxmeyer HE, et al. Enhanced
Vadhan-Raj S. Effects of in vivo treatment with PIXY321 on pro- hematopoietic activity of a human granulocyte/macrophage colony
liferation kinetics of bone marrow and blood myeloid progenitor stimulating factor-interleukin-3 fusion protein. Proc Natl Acad Sci
cells in patients with sarcoma. Exp Hematol 1995;23:335–340. USA 1991;88:5809–5813.
30. Campbell HD, Tucker WQJ, Hort Y, et al. Molecular cloning, 51. Cwirla SE, Peters EA, Barrett RW, Dower WJ. Peptides on phage:
nucleotide sequence, and expression of gene encoding human a vast library of peptides for identifying ligands. Proc Natl Acad
eosinophil differentiation factor (IL-5). Proc Natl Acad Sci USA Sci USA 1990;87:6378–6382.
1987;84:6629–6633. 52. Darnay BG, Aggarwal BB.Signal transduction by tumour necrosis
31. Cantell K, Hirvonen S, Mogensen KT, Pyhala L. Human leukocyte factor and tumour necrosis factor related ligands and their receptors.
interferon: production, purification, stability, and animal experi- Ann Rheum Dis. 1999;58 Suppl 1:I2-I13.
ments. Monograph. 1977;3:35–139. 53. Das M, Burge CB, Park E, Colinas J, Pelletier J. Assessment of
32. Carter P, Presta L, Gorman CM, et al. Humanization of anti- the total number of human transcription units. Genomics
p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci 2001;77:71–78.
USA 1992;89:4285–4289. 54. Daughaday WH, Rotwein P. Insulin-like growth factors I and II.
33. Chaffee S, Mary A, Stiehm ER, Girault D, Fischer A, Hershfield Peptide, messenger ribonucleic acid and gene structures, serum,
MS. IgG antibody response to ethylene glycol-modified adenosine and tissue concentrations. Endocr Rev 1989;10:68–91.
deaminase in patients with ADA deficiency. J Clin Invest 55. Debinski W, Puri RK, Kreitman RJ, Pastan I. A wide range of
1992;89:1643–1651. human cancers express interleukin-4 receptors that can be targeted
34. Chari RVJ, Martell BA, Gross JL, et al. Immunoconjugates con- with chimeric toxin composed of IL-4 and Pseudomonas exotoxin.
taining novel maytansinoids: promising anticancer drugs. Cancer J Biol Chem 1993;268:14065–14070.
Res 1992;52:127–131. 56. Derynck R. The physiology of transforming growth factor-β. Adv
35. Chaudhary VK, FitzGerald DJ, Adhaya S, Pastan I. Activity of a Cancer Res 1992; 58:27–52.
recombinant fusion protein between transforming growth factor- 57. Derynck R, Jarrett JA, Chen EY, et al. Human transforming growth
alpha and Pseudomonas toxin. Proc Natl Acad Sci USA 1987; factor-beta cDNA sequence and expression in tumor cell lines.
84:4538–4542. Nature 1985;316:701–705.
36. Chaudhary VK, Queen C, Junghans RP, Waldman TA, FitzGerald 58. Dinarello CA. The interleukin-1 family: 10 years of discovery.
DJ, Pastan I. A recombinant immunotoxin consisting of two anti- FASEB J 1994;8:1314–1325.
body variable domains fused to Pseudomonas exotoxin. Nature 59. Dinarello CA. Interleukin-1 and interleukin-1 antagonism. Blood.
1989;339:394–396. 1991 Apr 15;77(8):1627–52.
37. Chong CF, Scuderi P, Grimes WJ, et al. Tumor tragets stimulate 60. DiStefano PS, Johnson EM. Identification of a truncated form of
IL-2 activated killer cells to produce IFN-gamma and TNF. J nerve growth factor receptor. Proc Natl Acad Sci USA
Immunol 1989;142:2133–2139. 1988;85:270–274.
38. Clackson T, Hoogenbbom HR, Griffiths AD, Winter G. Making 61. Dorai H, McCartney JE, Hudziak RM, Tai M-S, Laminet AA,
antibody fragments using phage display libraries. Nature 1991;352: Houston LL, Huston JS, Oppermann H. Mammalian cell expres-
624–628. sion of a single chain Fv (sFv) antibody protein and their C-terminal
39. Clark BD, Collins KL, Gandy MS, Webb AC, Auron PE. Genomic fusions with interleukin-2 and other effector domains. Biotechnology
sequence for human interleukin-1 beta gene. Nucleic Acid Res 1995,12:890–897.
1986;14:7897–7914. 62. Downing JR, Rousesel MF, Sherr CJ. Ligand and protein kinase C
40. Claverie JM. Gene number. What if there are only 30,000 human downmodulate the colony stimulating factor 1 receptor by inde-
genes? Science 2001; 291: 1255–1257. pendent mechanism. Mol Cell Biol 1989;9:2890–2896.
41. Coller HA, Grandori C, Tamayo P, Colbert T, Lander ES, Eisenman 63. Dranoff G, Mulligan C. Gene transfer as cancer therapy. Adv
RN, Golub TR. Expression analysis with oligonucleotide microarrays Immunol 1995;58:417–454.
reveals that MYC regulates genes involved in growth, cell cycle, 64. Du XX, Williams DA.Interleukin-11: a multifunctional growth
signaling, and adhesion. Proc Natl Acad Sci USA 2000;97: factor derived from the hematopoietic microenvironment. Blood
3260–3265. 1994;83:2023–2030.
Kapil Mehta et al. 77

65. Edman JC, Halliwell RA, Valenzuela P, Goodman HM, Rutter WJ. 88. Gatanaga T, Hwang C, Kohr W, et al. Purification and character-
Synthesis of hepatitis B surface and core antigens in E. coli. Nature ization of an inhibitor (soluble TNF receptor) for TNF and lym-
1981;291: 503–506. photoxin obtained from the serum infiltrates of human cancer
66. Edwards GM, DeFeo-Jones D, Tai JY, et al. Epidermal growth fac- patients. Proc Natl Acad Sci USA 1990;87:8781–8784.
tor receptor binding is affected by structural determinants in the 89. Ghetie MA, Vitetta ES. Recent developments in immunotoxin
toxin domain of transforming growth factor-alpha-Pseudomonas therapy. Curr Opin Immunol 1994;6:707–714.
exotoxin fusion protein. Mol Cell Biol 1989;9:2860–2867. 90. Giri JG, Anderson DM, Kumaki S, Park LS, Grabstein KH,
67. Eschbach JW, Egrie JC, Downing MR, Browne JK, Adamson JW. Cosman D. IL-15, a novel T cell growth factor that shares activi-
Correction of the anemia of end-stage renal disease with recombi- ties and receptor components with IL-2. J Leukoc Biol.
nant human erythropoietin. N Engl J Med 1987;316:73–78. 1995;57:763–766.
68. Estrov Z, Kurzrock R, Wetzler M, et al. Suppression of chronic 91. Godon C, Lagniel G, Lee J, Buhler JM, Kieffer S, Perrot M,
myelogenous leukemia colony growth by IL-1 receptor antagonist Boucherie H, Toledano MB, Labarre J. The H2O2 stimulon in
and soluble IL-1 receptors: a novel application for inhibitors of Saccharomyces cerevisiae. J Biol Chem 1998;273:22480–22489.
IL-1 activity. Blood 1991;78:1476–1484. 92. Golub TR, Slonim DK, Tamayo P et al. Molecular classification
69. Fantes KH, Allen GJ. Specific activity of pure human interferons of cancer: class discovery and class prediction by gene expres-
and a non-biological method for estimating the purity of highly sion monitoring. Science 1999;286: 531–537.
purified interferon preparations. Interferon Res 1981;1:465–472. 93. Golumbek PT, Lazenby AJ, Levitsky HI, et al. Treatment of
70. Fehniger TA, Caliguiri MA. Interleukin-15; biology and relevance established renal cell cancer by tumor cells engineered to secrete
to human diseases. Blood 2001;97:14–32. interleukin-4. Science 1991;254:713–716.
71. Feng GS, Gray PW, Shepard HM, Taylor MW. Antiproliferative 94. Goodnough LT, Rudnick S, Price TH, et al. Increased preopera-
activity of a hybrid protein between interferon- and tumor necrosis tive collection of autologous blood with recombinant human
factor-ß. Science 1988;241:1501–1503. erythropoietin therapy. N Engl J Med 1989;321:1163–1168.
72. Fenton RG, Taub DD, Kwak LW, Smith MR, Longo DL. Cytotoxic T 95. Goodwin R, Friend D, Ziegler SF, et al. Cloning of the murine
cell response and in vivo protection against tumor cells harboring acti- and human interleukin receptors: demonstration of a soluble
vated ras prot-oncogenes. J Natl Cancer Inst 1993;85:1294–1302. form and homology to a new receptor superfamily. Cell
73. Fernandez-Botran R. Soluble cytokine receptors: their role in 1990;60:941–951.
immunoregulation. FASEB J 1191;5:256–2574. 96. Goodwin RG, Lupton S, Schmierer A, et al. Human interleu-
74. Fernadez-Borton R, Vitetta ES. A soluble, high affinity interleu- kin-7: molecular cloning and growth factor activity on human
kin-4 binding protein is present in the biological fluids of mice. and murine B-lineage cells. Proc Natl Acad Sci USA 1989;
Proc Natl Acad Sci USA 1990;87:4202–4206. 86:302–306.
75. FitzGerald DJ, Bjorn MJ, Ferris R, et al. Antitumor activity of an 97. Gorman CM. Mammalian cell expression. Curr Opin Biotech
immunotoxin in nude mouse model of human ovarian cancer. 1990;1:36–43.
Cancer Res 47:1407–1410. 98. Gospodarowicz D. Fibroblast growth factor. Crit Rev Oncog
76. FitzGerald DJ, Pastan I. A recombinant immunotoxin consisting 1989;1:1–26.
of two antibody variable domains fused to Pseudomonas exotoxin. 99. Gough NM, Gearing DP, King JA, et al. Molecular cloning and
Nature 1989;339:394–397. expression of the human homologue of the murine gene encoding
77. Fletcher FA, Williams DE. Recent progress in the discovery and myeloid leukemia inhibitory factor. Proc Natl Acad Sci USA
invention of novel hematopoietic cytokines. Crit Rev Oncol 1988;85:2623–2627.
Hematol 1992;13:1–15. 100. Grabstein KH, Eisenman J, Shanebeck K, et al. Cloning of a
78. Fujiwara K, Yoshino T, Miyake K, Ohara N, Akagi T. Expression novel T cell growth factor that interacts with the ß chain of the
of lymphocyte adhesion molecule (CD44) in malignant lympho- interleukin-2 receptor. Science 1994;264:965–968.
mas. Relevance to primary site, histological subtype and clinical 101. Gray A, Dull TJ, Ullrich A. Nucleotide sequence of epidermal
stage. Acta Med Okayama 1993;47:215–222. growth factor cDNA predicts a 128,000-molecular weight protein
79. Fukunaga R, Seto Y, Mizushima S, Nagata S. Three different precursor. Nature 1983;303:722–725.
mRNAs encoding human granulocyte colony-stimulating factor 102. Gray PW, Aggarwal BB, Benton C, et al. Cloning and expression
receptor. Proc Natl Acad Sci USA 1990;87:8702–8706. of cDNA for human lymphotoxin, a lymphokine with tumor
80. Fulton RJ, Uhr JW, Vitetta ES. In vivo therapy of the BCL1 tumor: necrosis factor activity. Nature 1984;312:721–724.
effect of immunotoxin valency and deglycosylation of the ricin A 103. Gray PW, Goeddel DV. Structure of the human immune inter-
chain. Cancer Res 1988;48;2626–2631. feron cDNA. Nature 1982;298:859–863.
81. Furr BJA, Hutchinson FG. A biodegradable delivery system for 104. Griffin JD, Rambaldi A, Vallenga E, Young DC, Ostapovicz D,
peptides: preclinical experience with the gonadotropin-releasing Cannistra SA. Secretion of interleukin-1 by acute myeloblastic
hormone agonist Zoladex. J Controlled Release 1992;21:117–128. leukemia cells in vitro induces endothelial cells to secrete colony
82. Furukawa Y, Ohata M, Miura Y, Saito M. Interleukin-1 production stimulating factors. Blood 1987;70:1218–1221.
by monocytic leukemia cells and its role in coagulation abnormali- 105. Griffin TW, Richardson C, Houston LL, LePage D, Bogden A,
ties. Leuk Res 1991;15:1133–1137. Raso V. Antitumor activity of intraperitoneal immunotoxins in a
83. Furutani Y, Notake M, Fukui T, et al. Complete nucleotide nude mouse model of human malignant mesothelioma. Cancer
sequence of the gene for human interleukin-1 alpha. Nucleic Acid Res 1987;47:4266–4270.
Res 1986;143:3167–3179. 106. Grossbard ML, Press OW, Appelbaum FR, Bernstein ID, Nadler
84. Gabrilove J, Jakubowski A, Grous J, et al. Initial results of a study LM. Monoclonal antibody-based therapies of leukemia and lym-
of rhG-CSF in cancer patients. Exp Hematol 1987;15:461–465. phoma. Blood 1992;80:863–878.
85. Gadina M, Hilton D, Johnston JA et al. Signaling by type I and II cytokine 107. Gubler U, Chua AO, Schoenhaut DS, et al. Coexpression of two
receptors: ten years after. Curr Opin Immunol 2001;13: 363–373. distinct genes is required to generate secreted, bioactive cytotoxic
86. Gallagher G, Dickensheets H, Eskdale J, Izotova LS, lymphocyte maturation factor. Proc Natl Acad Sci USA
Mirochnitchenko OV, Peat JD, Vazquez N, Pestka S, Donnelly RP, 1991;88:4143–4147.
Kotenko SV. Cloning, expression and initial characterization of 108. Gunther N, Betzel C, Weber W. The secreted form of the epidermal
interleukin-19 (IL-19), a novel homologue of human interleukin-10 growth factor receptor. J Biol Chem 1990;265:22082–22085.
(IL-10). Genes Immun. 2000;1:442–450. 109. Gurney AL, Marsters SA, Huang RM. Identification of a new
87. Ganser A, Lindemann A, Seipelt G, et al. Clinical effects of member of the tumor necrosis factor family and its receptor, a
recombinant interleukin-3. Am J Clin Oncol 1991;14:51–63. human ortholog of GITR. Curr Biol. 1999;9:215–218.
78 Recombinant proteins and genomics in cancer therapy

110. Gutterman JU, Blumenshein GR, Alexanian R, et al. Leukocyte ovarian carcinoma: toxicity and clinical response. Obstet Gynecol
interferon-induced tumor regression in human metastatic breast 1993;82:586–593.
cancer, multiple myeloma, and malignant lymphoma. Ann Intern 135. Jacobs K, Shoemaker C, Rudersdorf R, et al. Isolation and char-
Med 1980;93:399–406. acterization of genomic and cDNA clones of human erythropoietin.
111. Gutterman JU, Fine S, Quesada J, et al. Recombinant leukocyte A Nature 1985;313:806–810.
interferon: pharmacokinetics, single-dose tolerance, and biologi- 136. Jinno Y, Chaudhary VK, Kondo T, Adhya S, FitzGerald D, Pastan I.
cal effects in cancer patients. Ann Intern Med 1982;96:549–556. Mutational analysis of domain I of Pseudomonas exotoxin. J Biol
112. Gutterman JU. Cytokine therapeutics: lessons from interferon Chem 1988;263:13203–13207.
alpha. Proc Natl Acad Sci USA 1994;91:1198–1205. 137. Jurcic JC, Scheinberg DA, Houghton AN. Monoclonal antibody
113. Hall WA, Merill MJ, Walbridge S, Youle RJ. Epidermal growth therapy of cancer. In: Cancer Chemotherapy and Biological
factor receptors on ependymomas and other brain tumors. J Response Modifiers Annual 14 (Pinedo HM, Longo DL, Chabner
Neurosurg 1990;72:641–646. BA, eds), Amsterdam Elsevier 1993;pp129–149.
114. Han ZC, Caen JP. Cytokines acting on committed hematopoietic 138. Jurcic JG, Scheinberg DA. Recent developments in the radio-
progenitors. Clin Hematol 1994;7:65–89. therapy of cancer. Curr Opin Immunol 1994;6:715–721.
115. Heaney ML, Golde DW. Soluble hormone receptors. Blood 1993; 139. Kalinkowich A, Engelmann H, Harpaz N, et al. Elevated serum
82:1945–1948. levels of soluble tumor necrosis factor in patients with HIV infection.
116. Heldin C-H. Structural and functional studies on platelet derived Clin Exp Immunol 1992;89:351–355.
growth factor. EMBO J 1992;11:4251–4259. 140. Kaplan EH, Leslie WT. Cutaneous T-cell lymphomas. Curr Opin
117. Heller J. Polymers for controlled parenteral delivery of peptides Oncol 1993;5:812.
and proteins. Adv Drug Deliv Rev 1993;10:163–204. 141. Kaptain S, Tan LK, Chen BY. Her-2/neu and breast cancer. Diag
118. Heller RA, Song K, Fan N, et al. The p70 tumor necrosis factor Mol Pathol 2001;10:139–152.
receptor mediates cytotoxicity. Cell 1992;70:47–56. 142. Karp JE, Broder S. New directions in molecular medicine. Cancer
119. Hellstrom I, Hellstrom KE. Anti-idiotypic antibodies as tumor Res 1994;54:653–665.
vaccines. In: New Generation Vaccines (Woodrow GC, Levine 143. Katoh M, Yazaki Y, Sugimura T, Terada M. C-erbB3 gene
MM, eds), New York, Marcel Dekker, 1990;pp863–870. encodes secreted as well as transmembrane receptor tyrosine
120. Hersch EM, Metch BS, Muggia FM, et al. Phase II studies of kinase. Biochem Biophys Res Commun 1993;192:1189–1197.
recombinant tumor necrosis factor alpha in patients with malig- 144. Katre NV. The conjugation of proteins with polyethylene gly-
nant disease: a summary of the Southwest Oncology Group expe- col and other polymers: altering properties of proteins to
rience. J Immunother 1991;10:426–431. enhance their therapeutic potential. Adv Drug Deliv Rev
121. Hill ADK, Redmond HP, McCarthy J, Croke DT, Grace PA, 1993;10:91–114.
Bouchier-Hayes D. Antineoplastic effects of granulocyte mac- 145. Kawasaki ES, Ladner MB, Wang AM, et al. Molecular cloning of
rophage colony stimulating factor. Br J Surg 1992;79:459 (Abstr). a complementary DNA encoding human macrophage-specific
122. Hill ADK, Redmond HP, Croke DT, Grace PA, Bouchier-Hayes colony stimulating factor (CSF-1). Science 1985;230:291–296.
D. Cytokines in tumor therapy. Br J Surg 1992;79:990–997. 146. Klein B, Wijdenes J, Xang XG, et al. Murine anti-interleukin-6
123. Hill CS, Treisman R. Transcriptional regulation by extracellular monoclonal antibody therapy for a patient with plasma cell
signals: mechanisms and specificity. Cell 1995;80:199–211. leukemia. Blood 1991;78:1198–1204.
124. Hinman LM, Hamann PR, Wallace R, Menedez TA, Durr FE, 147. Koff WC, Paige C, Gutterman J, Fidler IJ. Efficient activation of
Upeslacis J. Preparation and characterization of monoclonal anti- human blood monocytes to a tumoricidal state by liposomes con-
body conjugates of the calicheamicins: a novel and potent family taining human recombinant gamma interferon. Cancer Immunol
of antitumor antibiotics. Cancer Res 1993;53:3336–3342. Immunther 1985;19:85–89.
125. Hirano T, Yasukawa K, Harada H, et al. Complementary DNA 148. Koppel GA. Recent advances with monoclonal antibody drug
for a novel human interleukin (BSF-2) that induces B-lymphocytes targeting for the treatment of human cancer. Bioconjug Chem
to produce immunoglobulin. Nature 1986;324:73–76. 1990;1:13–23.
126. Hitzeman RA, Chen CY, Hagie FE, Lugovoy JM, Singh A. Yeast: 149. Kotenko SV, Izotova LS, Mirochnitchenko OV, Esterova E,
an alternative organism for foreign protein production. In: Dickensheets H, Donnelly RP, Pestka S. Identification, cloning,
Recombinant DNA Products (Bollon, AP, ed), Boca Raton, FL, and characterization of a novel soluble receptor that binds IL-22
CRC Press, 1983; pp 47–65. and neutralizes its activity. J Immunol. 2001;166:7096–7103.
127. Hock H, Dorsch M, Kunzendorf U, et al. Vaccination with tumor 150. Kozak RW, Lorberboum GH, Jone L, et al. IL-2-PE40 prevents the
cells genetically engineered to produce different cytokines: effectivity development of tumors in mice injected with IL-2 receptor express-
not superior to a classical adjuvant. Cancer Res 1993;53:714–716. ing EL4 transfectant tumor cells. J Immunol 1990;145:2766–2771.
128. Horuk R. The interleukin-8-receptor family: from chemokines to 151. Krammer PH. CD95’s deadly mission in the immune system.
malaria. Immunol Today. 1994;15:169–174. Nature 2000;407:789–795.
129. Hoshino T, Kawase Y, Okamoto M et al. IL-18 transgenic mice: 152. Kreeger KY. Cancer immunotherapies: an old idea sparks new
in vivo evidence of a broad role for IL-18 modulating immune studies, industry interest. The Scientist 1995;9:1.
function. J Immunol 2001;166:1014–7018. 153. Kreitman RJ, Bailon P, Cahudhary VK, FitzGerald DJ, Pastan I.
130. Ideker T, Thorsson V, Ranish J A. Integrated genomic and pro- Recombinant immunotoxins containing anti-Tac(Fv) and deriva-
teomic analyses of a systematically perturbed metabolic network. tives of Pseudomonas exotoxin produce complete regression in
Science 2001;292:929–934. mice of interleukin-2 receptor-expressing human carcinoma.
131. Ihle JN. Signaling by the cytokine receptor superfamily in normal Blood 1994;83:426–434.
and transformed hematopoietic cells. Adv Cancer Res. 154. Kreitman RJ, Chang CN, Hudson DV, Queen C, Bailon P, Pastan I.
1996;68:23–65. Anti-Tac(Fab)-PE40, a recombinant double chain immunotoxin
132. International Human Genome Sequence Consortium. Initial which kills interleukin-2 receptor bearing cells and induces com-
sequencing and analysis of the human genome. Nature 2001; plete remission in an in vivo tumor model. Int J Cancer
409: 860. 1994;57:856–864.
133. Itoh N, Yonehara S, Ishii A, et al. The polypeptide encoded by the 155. Kreitman RJ, Chaudhary VK, Waldman T, Willingham MC,
cDNA for the human cell surface antigen Fas can mediate apoptosis. FitzGerald DJ, Pastan I. The recombinant immunotoxin anti-Tac
Cell 1991;66:233–243. (Fv)-Pseudomonas exotoxin 40 is cytotoxic towards peripheral
134. Jacobs AJ, Fer M, Su FM, et al. A phase I trial of a Rhenium blood malignant cells from patients with adult T cell leukemia.
186-labeled monclonal antibody administered intraperitoneal in Proc Natl Acad Sci USA 1990;87:8291–8299.
Kapil Mehta et al. 79

156. Kreitman RJ, Hansen HJ, Jones AL, FitzGerald D, Goldenberg 178. Luckow VA, Summers MD. Trends in the development of bacu-
DM, Pastan I. Pseudomonas exotoxin-based immunotoxin con- lovirus expression vectors. Biotechniques 1988;6:47–55.
taining the antibody LL2 or LL2 Fab’ induce regression of sub- 179. Malik N, Kallestad JC, Gunderson NL, et al. Molecular cloning,
cutaneous human B-cell lymphoma in mice. Cancer Res 1993; sequence analysis, and functional expression of a novel growth
53:819–825. regulator, oncostatin M. Mol Cell Biol 1989;9:2847–2853.
157. Lachman LB, Dinarello CA, Llansa LD, Fidler IJ. Natural and 180. Markussen J, Diers I, Engesgaard A, et al. Soluble, prolonged
recombinant human interleukin-1 is cytotoxic for human mela- acting insulin derivatives. II Degree of protraction and crystalliz-
noma cells. J Immunol 1986;136: 3098–3102. ability of insulins substituted in positions A17, B8, B27 and B30.
158. Lalli E, Sassone-Corsi P. Signal transduction and gene regulation: Protein Eng 1987;1:215–223.
the nuclear response to cAMP. J Biol Chem 1994;269:17359–17362. 181. Marton MJ, DeRisi JL, Bennett HA et al. Drug target validation
159. Layton MJ, Cross BA, Metcalf D, Ward LD, Simpson RJ, Nicola and identification of secondary drug target effects using DNA
NA. A major binding-protein for human leukemia inhibitory fac- microarrays. Nat Med 1998;4:1293–1301.
tor in normal mouse serum: identification as a soluble form of the 182. McLaughlin P, Cabanillas, F, Hagemeister FB, et al. CHOP-Bleo
cellular receptor. Proc Natl Acad Sci USA 1992;89:8616–8620. plus interferon for stage IV low-grade lymphoma. Ann Oncol
160. Lee VHL. Peptide and protein drug delivery: opportunities and 1993;4:205–211.
challenges. Pharm Int 1986;208–212. 183. Meredith RF, Khaxaeli MB, Lui T, et al. Dose fractionation of
161. Leitzel K, Teramoto Y, Sampson E, et al. Elevated soluble c-erb-2 radiolabeled antibodies in patients with metastatic colon cancer.
antigen levels in the serum and effusions of a proportion of a J Nucl Med 1992;33:1648–1653.
breast cancer patients. J Clin Oncol 1992;10:1436–1443. 184. Meredith RF, Khazaeli MB, Plott WE, et al. Phase I trial of
162. Leonard WJ, Shores EW, Love PE. Role of the common cytokine iodine-131 chimeric B72.3 (human IgG4) in metastatic colorectal
receptor gamma chain in cytokine signaling and lymphoid devel- cancer. J Nucl Med 1992;33:23–29.
opment. Immunol Rev. 1995 Dec;148:97–114. 185. Miller L. Current status of G-CSF in support of chemotherapy
163. Lewis R. End of century marks dawn of clinical trial era for and radiotherapy. Oncology 1993;7:67–88.
cancer vaccines. The Scientist 1995;9:15. 186. Milstein C. Monoclonal antibodies (review). Sci Am
164. Lewis GD, Figari L, Fendly B, Carter P, Gorman C, Shepard M. 1980;243:66–74.
Differential responses of human tumor cell lines to anti-p185HER2 187. Minty A, Chalon P, Derocq JM, et al. Interleukin 13 is a new
monoclonal antibodies. Cancer Immunol Immunother human lymphokine regulating inflammatory and immune
1993;37:255–263. responses. Nature 1993;362:248–250.
165. Lewis R. Herceptin earns recognition in breast cancer arsenal. 188. Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A.
Scientist 2001;15:10–11. Interleukin-10 and the interleukin-10 receptor. Annu Rev
166. Li H, Chen J, Huang A, Stinson J, Heldens S, Foster J, Dowd P, Immunol. 2001;19:683–765.
Gurney AL, Wood WI. Cloning and characterization of IL-17B 189. Morgan AC, Comezoglu FT, Manger R, Jarvis B, Abrams PJ,
and IL-17C, two new members of the IL-17 cytokine family. Proc Sivam G. Immunoconjugates of a protein-synthesis-inhibiting
Natl Acad Sci U S A 2000;97:773–778. drugs. In: Therapeutic Monoclonal Antibodies (Borrebaeck C,
167. Lienard D, Ewalenko P, Delmotte JJ, et al. High dose recombi- Larrick J, eds), New York, Stockton Press, 1990:pp143–158.
nant tumor necrosis factor alpha in combination with interferon 190. Mori I, Hossain MJ, Takeda K, Okamura H, Imai Y, Koshaka S,
gamma and melphalan in isolation perfusion of the limbs for Kimura Y. Impaired microglial activation in the brain of IL-18
melanoma and sarcoma. J Clin Oncol 1992;10:52–60. gene-disrupted mice after neurovirulent influenza A virus infec-
168. Lim VS, DeGowin RL Zavala D, et al. Recombinant human tion. Virol 2001;287:163–170.
erythropoietin treatment in pre-dialysis patients. Ann Intern Med 191. Mosley B, Beckman MP, March CJ, et al. The murine interleu-
1989;110:108–118. kin-4 receptor: a molecular cloning and characterization of
169. Lipshutz RJ, Fodor SP, Gingeras TR, Lockhart DJ. High density secreted and membrane-bound forms. Cell 1989;59:335–348.
synthetic oligonucleotide arrays. Nat Genet 1999;21:20–24. 192. Motoyshi K, Takaku F. Human monocytic colony stimulating
170. Lippman SM, Parkinson DR, Itri LM, et al. 13-Cis retinoic acid factor, phase I/II clinical studies. In: Hematopoietic Growth
and interferon-α-2a: effective combination therapy for advanced Factors in Clinical Applications. (Metelsmann R, Hermann F,
squamous cell carcinoma of the skin. J Natl Cancer Inst eds), New York: Marcel Dekker, 1990; pp 161–175.
1992;84:235–241. 193. Mukaida N, Shiroo M, Matsushima K. Genomic structure of the
171. Lodato RF, Feig B, Akimaru K, Soma GI, Klostergaard J. human monocyte-derived neutrophil chemotactic factor IL-8.
Hemodynamic evaluation of recombinant human tumor necrosis J Immunol 1989;143:1366–1371.
factor (TNF), TNF-SAM2 and liposomal TNFSAM2 in an anes- 194. Mule JJ, Smith CA, Rosenberg SA. Interleukin 4 (B cell stimula-
thetized dog model. J Immunother 1995;17:19–29. tory factor 1) can mediate the induction of lymphokine activated
172. Lorberboum GH, FitzGerald DJ, Chaudhary VK, Adhya S, killer activity directed against fresh tumor cells. J Exp Med
Pastan I. Cytotoxic activity of an interleukin 2-Pseudomonas 1987;166:792–798.
exotoxin chimeric protein produced in Escherichia coli. Proc 195. Mullberg J, Schooltink H, Stoyan T, et al. The soluble interleu-
Natl Acad Sci USA 1988;85:1922–1926. kin-6 receptor is generated by shedding. Europ J Immunol
173. Lord MJ, Roberts LM, Robertus JD. Ricin: structure, mode of 1993;23:473–480.
action, and some current applications. FASEB J 1994;8:201–208. 196. Nagao M, Masuda S, Abe S, Ueda M, Sasaki R. Production and
174. Lotze MT, Custer MC, Bolton ES, Wiebke EA, Kwakami Y, ligand-binding characteristics of the soluble form of murine
Rosenberg SA. Mechanisms of immunological antitumor activ- erythropoietin receptor. Biochem Biophys Res Commun
ity: lessons from the laboratory and clinical application. Hum 1992;188:888–897.
Immunol 1990;28:198–207. 197. Nagata S, Tsuchiya M, Asano S, et al. Molecular cloning and
175. Lotze MT, Matory YL, Rayner AA. Clinical effects and toxicity of expression of cDNA for human granulocyte colony-stimulating
interleukin-2 in patients with cancer. Cancer 1986;58:2764–2772. factor. Nature 1986;319:415–418.
176. Lowman HB, Bass S, Simpson S, Wells JA. Selecting high affin- 198. Nakajima T, Yamamoto S, Cheng M, et al. Soluble interleukin-6
ity binding proteins by monovalent phage display. Biochemistry receptor is released from receptor-bearing cell lines in vitro.
1991;30:10832–10838. Japan J Cancer Res 1992;83:373–378.
177. Luckow VA. Recombinant DNA Technology and Applications 199. Nakamura T, Nishizawa T, Hagiya M, et al. Molecular cloning
(Prokop A, Bajpai RK, Ho CS, eds), New York, McGraw Hill, and expression of human hepatocyte growth factor. Nature
1991;pp97–152. 1989;342:440–443.
80 Recombinant proteins and genomics in cancer therapy

200. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 224. Peoples GE, Goedegebuure PS, Smith R, Linehen DC, Yoshino I,
is a unique cytokine that stimulates both Th1 and Th2 responses Eberlein TJ. Breast and ovarian cancer-specific cytotoxic T lym-
depending on its cytokine milieu. Cytokine Growth Factor Rev. phocytes recognize the same HER1/neu-derived peptide. Proc
2001;12(1)57–72. Natl Acad Sci USA 1995;92:432–436.
201. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 225. Pestka S. The human interferons-from protein purification and
regulates both Th1 and Th2 responses. Annu Rev Immunol. sequence to cloning and expression in bacteria: before, between
2001;19:423–474. and beyond. Arch Biochem Biophys 1983;221:1–37.
202. Narazaki M, Yasukawa K, Saiton T, et al. Soluble form of IL-6 226. Pestka S. The purification and manufacture of human interferons.
signal transducing receptor component gp130 in human serum Sci Am 1983;249:36–43.
processing a potential to inhibit signals through membrane 227. Pestka S, Langer JA, Zoom KC, Samuel CE. Interferons and their
anchored gp130. Blood 1993;82:1120–1126. actions. Annu Rev Biochem 1987; 56:727–777.
203. Nedwin GE, Jarrett-Nedwin J, Smith D, et al. Structure and chro- 228. Petch L, Harris J, Raymond VW, Blasband A, Lee DC, Earp HS.
mosomal localization of the human lymphotoxin gene. J Cell A truncated secreted form of the epidermal growth factor recep-
Biochem 1985;29:171–182. tor is encoded by an alternatively spliced transcript in normal rat
204. Nedwin GE, Naylor SL, Sakaguchi AY, et al. Human lymphotoxin tissue. Mol Cell Biol 1990;10:2973–2982.
and tumor necrosis factor genes: structure, homology and chro- 229. Peters K, Vejlsgaard GL. Basal cell and squamous cell carcinoma
mosomal localization. Nucleic Acid Res 1985;13:6361–6371. and Kaposi’s carcinoma. Curr Opin Oncol 1992;4:380–385.
205. Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE. The IL-4 230. Petersen BH, DeHerdt SV, Schneck DW, Bumol TF. The human
receptor: signaling mechanisms and biologic functions. Annu immune response eto KS1/4-desacetylvinblastin (LY256787) and
Rev Immunol 1999;17:701–738. KS1/4-desacetylvinblastin hydrazide (LY203728) in single and
206. Nemunaitis J, Rabinowe SN, Singer JW, et al. Recombinant gran- multiple dose clinical studies. Cancer Res 1991;51:2286–2290.
ulocyte-macrophage colony stimulating factor after autologous 231. Pietersz GA, McKenzie IFC. Antibody conjugates for the treat-
bone marrow transplantation for lymphoid cancer. N Engl J Med ment of cancer. Immunol Rev 1992;129:57–80.
1991;324:1773–1778. 232. Plautz GE, Yang ZY, Wu BY, Gao X, Huang L, Nabel GJ.
207. Niskanen E. Hematopoietic growth factors in clinical hematology. Immunotherapy of malignancy by in vivo gene transfer into
Ann Med 1991;23:615–624. tumors. Proc Natl Acad Sci USA 1993;10:4645–4649.
208. Novick D, Cohen B, Rubinstein M. Soluble interferon-alpha 233. Pluckthun A. Antibodies from Escherichia coli. Nature 1990;
receptor molecules are present in body fluids. FEBS Lett 347:497–498.
1993;314:445–448. 234. Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model
209. Novick D, Engelmann H Rubinstein M, Wallach D. Soluble for p53-induced apoptosis. Nature 1997;389:300–305.
cytokine receptors are present in normal huamn urine. J Exp Med 235. Porgador A, Tzehoval E, Katz A, et al. Interleukin 6 gene trans-
1989;170:1409–1414. fection into Lewis lung carcinoma tumor cells suppresses the
210. Obreg K, Norheim I, Lind E. Treatment of malignant carcinoid malignant phenotype and confers immunotherapeutic competence
tumors with human leukocyte interferon: long term results. against parental metastatic cell. Cancer Res 1992;52:3679–3686.
Cancer Treat Rev 1986;70:1297–1304. 236. Press OW, Eary JF, Appelbaum FR, et al. Radiolabeled antibody
211. Oehm A, Behrmann I, Falk W, et al. Purification and molecular therapy of B cell lymphoma with autologous bone marrow
cloning of the APO-1 cell surface antigen, a member of the tumor support. New Engl J Med 1993;329:1219–1224.
necrosis factor/nerve growth factor receptor superfamily; 237. Quesada JR, Hersh EM, Manning J, et al. Treatment of hairy cell
sequence identity with the Fas antigen. J Biol Chem leukemia with recombinant -interferon. Blood 1986;68:493–497.
1992;267:10932–10937. 238. Ramakrishnam S, Bjorn MJ, Houston LL. Recombinat ricin A
212. Old LJ, ed. Tumor necrosis factor and related cytotoxins. CIBA chain conjugated to monoclonal antibodies: improved tumor cell
Foundation Symp. 1987;131:131–137. inhibition in the presence of lysosomotropic compounds. Cancer
213. Oppmann B, Lesley R, Blom B, Timans JC et al. Novel p19 pro- Res 1989;49:613–617.
tein engages IL-12p40 to form a cytokine, IL-23, with biological 239. Renauld JC, Druez C, Kermouni A, et al. Expression cloning of
activities similar as well as distinct from IL-12. Immunity. the murine and human interleukin 9 receptor cDNAs. Proc Natl
2000;13:715–725. Acad Sci USA 1992;89:5690–5694.
214. Ostade XV, Vandenabeele P, Everaerdt B, et al. Human TNF 240. Renauld JC, Houssiau F, Louahed J, Vink A, Van Snick J,
mutants with selective activity on the p55 receptor. Nature Uyttenhove C. Interleukin-9. Adv Immunol. 1993;54:79–97.
1993;361:266–269. 241. Rinderknecht E, O’Connor BH, Rodriguez H. Natural human
215. Ostrand-Rosenberg S. Tumor immunotherapy: the tumor cell as interferon gamma. J Biol Chem 1984;259:6790–6797.
an antigen-presenting cell. Curr Opin Immunol 1994;6:722–727. 242. Rosenberg SA. Immunotherapy and gene therapy of cancer.
216. Pardoll D. Cancer Vaccines. Immunol Today 1993;14:310–316. Cancer Res 1991;51:5074s–5079s.
217. Parrish-Novak J, Dillon SR, Nelson A et al. Interleukin 21 and 243. Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the
its receptor are involved in NK cell expansion and regulation of systemic administration of autologous lymphokine activated
lymphocyte function. Nature 2000;408:57–63. killer cells and recombinant interleukin-2 of high dose IL-2
218. Park JW, Kirpotin DB, Hong K et al. Tumor targeting using anti alone. N Engl J Med 1985;313:1485–1490.
her2 immunoliposomes. J Cont Release 2001;74:95–113. 244. Rosenberg SA, Lotze MT, Yang JC, et al. Combination therapy
219. Park CC, Morel JCM, Amin MA, Connors MA, Harlow LA, with interleukin-2 and alpha-interferon for treatment of patients
Koch AE. Evidence of IL-18 as a novel angiogenic mediator. with advanced cancer. J Clin Oncol 1989;7:1863–1874.
J Immunol 2001;167:1644–1653. 245. Rosenberg SA, Spiess P, Lafreniere RA. A new approach to the
220. Pastan I, FitzGerald DJ. Recombinant toxins for cancer treatment. adoptive immunotherapy of cancer with tumor infiltrating lym-
Science 1991;254:1173–1177. phocytes. Science 1986;233:1318–1321.
221. Pastan I, Chaudhary V, FitzGerald DJ. Recombinant toxins as 246. Rosenblum RG, Unger BW, Gutterman JU, Hersh EM, David
novel therapeutic agents. Annu Rev Biochem 1992;61:331–354. GS, Frincke JM. Modification of human leukocyte interferon
222. Paul SR, Bennet F, Calvetti JA, et al. Molecular cloning of a pharmacology with a monoclonal antibody. Cancer Res
cDNA encoding interleukin 11, a stromal cell-derived lymphpoietic 1985;45:2421–2424.
and hematopoietic cytokine. Proc Natl Acad Sci USA 1990;87: 247. Ross C, Tingsgaard P, Jorgensen H, Vejlsgaard GL. Interferon
7512–7516. treatment of cutaneous T-cell lymphoma. Eur J Haematol
223. Pennica D, Nedwin GE, Hayflick JS, et al. Human tumor necrosis 1993;51:63–72.
factor: precursor structure, expression and homology to lympho- 248. Samakrishnan S, Bjorn MJ, Houston LL. Recombinant ricin A
toxin. Nature 1984;312:724–729. chain conjugated to monoclonal antibodies: improved tumor cell
Kapil Mehta et al. 81

inhibition in the presence of lysosomotropic compounds. Cancer 272. Smalley RV, Andersen JW, Hawkins MJ, et al. Interferon alpha
Res 1989;49:613–617. combined with cytotoxic chemotherapy for patients with non-
249. Sambrook J, Fritsch EF, Manatis T. Molecular Cloning: A Hodgkin’s lymphoma. N Engl J Med 1992;327:1336–1341.
Laboratory Manual, New York, Cold Spring Harbor Laboratory 273. Smith CM Bioinformatics, genomics, and proteomics. Scientist
Press, 1989; pp 5.30–5.90. 2000;14:26–28.
250. Sanderson CJ. The biological role of interleukin-5. Int J Cell 274. Smith CA, Davis T, Anderson D, et al. A receptor for human
Cloning 1990;8 (suppl 1): 147–153. tumor necrosis factor defines an unusual family of cellular and
251. Sasaki K, Chiba S, Mano H, Yazaki Y, Hirai H. Identification of viral proteins. Science 1990;248:1019–1102.
a soluble GM-CSF binding-protein in the supernatant of a human 275. Solal-Celigny P, Lepage E, Brousse N, et al. Recombinant inter-
choriocarcinoma cell line. Biochem Biophys Res Commun feron alfa-2b combined with a regimen containing doxorubicin in
1992;183:252–257. patients with advanced follicular lymphoma. N Engl J Med
252. Scambia G, Panici PB, Battaglia F, et al. Receptors for epidermal 1993;329:1608–1614.
growth factor and steroid hormones in primary lyrangeal tumors. 276. Souza LM, Boone TC, Gabrilove J, et al. Recombinant human
Cancer 1991;67:1347–1351. granulocyte colony-stimulating factor: effects on normal and leu-
253. Sceinberg DA, Straus DJ, Yeh SD, et al. A phase I toxicity, phar- kemic myeloid cells. Science 1986;232:61–65.
macology, and dosimetery trial of monoclonal antibody OKB7 in 277. Spencer VA, Davie JR. Signal transduction pathways and chro-
patients with non-Hodgkin’s lymphoma: effect of tumor burden matin structure in cancer cells. J Cell Biochem (Suppl) 2000;
and antigen expression. J Clin Oncol 1990;8:792–803. 27–35.
254. Scherf U, Ross DT, Waltham M. A gene expression database for the 278. Spitler LE. Cancer vaccines: the interferon analogy (editorial).
molecular pharmacology of cancer. Nat Genet 2000;24:236–244. Cancer Biother 1995;10:1–3.
255. Shaffer AL, Rosenwald A, Hurt EM et al. Signatures of the 279. Spriggs MK, Hruby DE, Maliszewski CR, et al. Vaccinia and
immune response. Immunity 2001;15:375–385. cowpox viruses encode a novel secreted interleukin-1 binding
256. Sherlock G, Hernandez-Boussard T, Kasarskis A et al. The Stanford protein. Cell 1992; 71:145–152.
Microarray Database. Nucleic Acids Res 2001;29:152–155. 280. Stebbing N, Weck PK. Preclinical assessment of biological prop-
257. Schiro R, Longoni D, Rossi V, et al. Suppression of juvenile erties of recombinant DNA derived interferons. In: Recombinant
chronic myelogenous leukemia colony growth by IL-1 receptor DNA Products: insulin, interferon, and growth hormone (Bollon
antagonist. Blood 1994;83:460–465. AP, ed), Boca Raton, FL, CRC Press, 1984;pp75–114.
258. Seigall CB, Xu YH, Cahudary VK, Adhya S, Fitzgerald D, Pastan 281. Stills HF, Bullock BC, Clarkson TB. Increased atherosclerosis
I. Cytotoxic activities of a fusion protein compromised of TGFα and glomerulonephritis in cynomolgus monkeys given injections
and Pseudomonas exotoxin. FASEB J 1989;3:2647–2652. of BSA over an extended period of time. J Pathol
259. Sen GC, Lengyel P. The interferon system. J Biol Chem 1983;113:222–234.
1992;267:5017–5020. 282. Strander H. Interferon treatment of human neoplasia. Adv Cancer
260. Sharma S, Mehta S, Morgan J, Maizel A. Molecular cloning and Res 1986;46:1–265.
expression of a human B-cell growth factor gene in E. coli. 283. Strander H, Cantell K, Jakobsson PA, Nilsonn U, Soderberg G.
Science 1985;235:1489–1492. Exogenous interferon therapy of osteogenic sarcoma. Oerhop
261. Shoyab M, Plowman GD, McDonald VL, Bradley JG, Todaro Scand 1975;45:958–959.
GJ. Structure and function of human amphiregulin: a member of 284. Strom TB, Anderson PL, Rubin-Kelley VE, Williams DP,
the epidermal growth factor family. Science 1989; 243: Kijokawa T, Murphy JR. Immunotoxins and cytotokine toxin
1074–1076. fusion proteins. Semin Immunol 1990;2:467–479.
262. Sica GL, Zhu G, Tamada K, Liu D, Ni J, Chen L. RELT, a new 285. Su AI, Welsh JB, Sapinoso LM et al. Molecular classification of
member of the tumor necrosis factor receptor superfamily, is human carcinomas by use of gene expression signatures. Cancer
selectively expressed in hematopoietic tissues and activates tran- Res 2001;61:7388–7393.
scription factor NF-kappaB. Blood. 2001;97:2702–2707. 286. Suda T, Takahashi T, Golstein P, Nagata S. Molecular cloning
263. Siegall CB, FitzGerald DJ, Pastan I. Cytotoxicity of IL6-PE40 and expression of the Fas ligand, a novel member of the tumor
and derivatives on tumor cells expressing a range of interleukin 6 necrosis factor family. Cell 1993;75:1169–1178.
receptor levels. J Biol Chem 1990;265:16318–16323. 287. Suzuki H, Yasukawa K, Saito T, et al. Anti-human IL-6 receptor
264. Siegall CB, FitzGerald DJ, Pastan I. Selective killing of IL6 antibody inhibits human myeloma growth in vivo. Eur J Immunol
receptor bearing myeloma cells using recombinant IL6- 1992;22:1189–1193.
Pseudomonas toxin. Curr Top Microbiol Immunol 1990;166: 288. Takatsu K. Interleukin 5 and B cell differentiation. Cytokine
63–69. Growth Factor Rev. 1998 Mar;9(1):25–35.
265. Siegall CB, Kreitman RJ, FitzGerald DJ, Pastan I. Antitumor 289. Takaki S, Tominaga A, Hitoshi Y, et al. Molecular cloning and
effects of interleukin-6 Pseudoman exotoxin chimeric molecules expression of the murine interleukin-5 receptor. EMBO J
against the human hepatocellular carcinoma. Cancer Res 1990;9:4367–4374.
1991;51:2831–2836. 290. Talpaz M, Kantarjian HM, McCredie K, Trujillo JM, Keating MJ,
266. Sievers EL, Appelbaum FR, Spielberger RT et al. Selective abla- Gutterman JU. Hematological remission and cytogenetic improve-
tion of acute myeloid leukemia using antibody-targeted chemo- ment induced by recombinant human interferon alpha in chronic
therapy: A phase I study of an anti-CD33 Calicheamicin myelogenous leukemia. N Engl J Med 1986;314:1065–1069.
immunoconjugate. Blood 1999;93:3678–3684. 291. Talpaz M, O’Brien S, Rose E et al. Phase 1 study of polyethylene
267. Sims JE, Acres RB, Grubin CE, et al. Cloning of the interleukin-1 glycol formulation of interferon alpha-2B (Schering 54031) in
receptor from human T-cells. Proc Natl Acad Sci USA Philadelphia chromosome positive chronic myelogenous leukemia.
1989;86:8946–8950. Blood 2001;98:1708–1713.
268. Sims JE, March CJ, Cosman D, et al. cDNA expression cloning 292. Talapatra S, Thompson CB. Growth factor signaling in cell survival:
of the IL-1 receptor, a member of the immunoglobulin superfamily. implications for cancer treatment. J Pharmacol Exp Ther
Science 1988;241:585–589. 2001;298:873–878.
269. Singh A, Ni J, Aggarwal BB.Death domain receptors and their 293. Tanaguchi T, Ohno S, Fujii-Kuriyama Y, Murmatsu M. The
role in cell demise. J Interferon Cytokine Res. 1998;18:439–450. nucleotide sequence of human fibroblast interferon cDNA. Gene
270. Skolnick AA. First immunotoxin therapy for many common 1980;10:11–15.
solid tumors enters phase I clinical trial. JAMA 1993;270:2280. 294. Tanaka H, Satake IR, Ishikawa M, Matsuki S, Asano K.
271. Slamon D et al. Use of chemotherapy plus a monoclonal antibody Pharmacokinetics of recombinant human granulocyte colony
against Her-2 for metastatic breast cancer that overexpresses stimulating factor conjugated to polyethylene glycol in rats. Cancer
Her-2. New Eng J Med 2001;344:783–792. Res 1991;51:3710–3714.
82 Recombinant proteins and genomics in cancer therapy

295. Tao MH, Levy R. Idiotype/granulocyte-macrophage colony stim- 319. Vitetta ES, Thrope PE, Uhr JW. Immunotoxicins: magic bullets
ulating factor fusion protein as a vaccine for a B-cell lymphoma. or misguided missiles. Immunol Today 1993;14:252–259.
Nature 1993;362:755–758. 320. Vitetta ES, Uhr JW. Monoclonal antibodies as agonists: an
296. Tartaglia LA, Goeddel DV. Two TNF receptors. Immunol Today expanded role for their use in cancer therapy. Cancer Res
1992;13:151–153. 1994;54:5301–5309.
297. TeVelde AA, Huijbens RJF, DeVries JE, Figdor CG. Interleukin 321. Waldmann TA, Goldman CK, Bongiovanni KF, et al. Therapy of
4 inhibits secretion of IL-1, tumor necrosis factor and IL-6 by patients with human T-cell lymphotropic virus I-induced adult T
human monocytes. Blood 1990;76:1392–1398. cell leukemia with anti-Tac, a monoclonal antibody to the recep-
298. The International Chronic Granulomatous Disease Comparative tor for interleukin-2. Blood 1988;72:1805–1816.
Study Group. N Engl J Med 1991;324:509–516. 322. Waldmann T, Tagaya Y, Bamford R. Interleukin-2, interleukin-15,
299. Tiesman J, Hart CE. Identification of a soluble receptor for plate- and their receptors. Int Rev Immunol 1998;16:205–226.
let-derived growth factor in cell-conditioned medium and human 323. Wang H, Marsters SA, Baker T et al. TACI-ligand interactions
plasma. J Biol Chem 1993;268:9621–9628. are required for T cell activation and collagen-induced arthritis in
300. Tong J, Gordon MS, Srour EF, et al. In vivo administration of mice. Nat Immunol. 2001;2:632–637.
recombinant human stem cell factor expands the number of 324. Watson, J (ed). Recombinant DNA in medicine and industry.
hematopoietic stem cells. Blood 1993;82:784–791. 1992;453–471.
301. Trail PA, Willner D, Lasch SJ, et al. Cure of xenografted human 325. Weber J, Yang JC, Topalian SL, et al. Phase I trial of subcutane-
carcinomas by BR96-doxorubicin conjugates. Science 1993;261: ous interleukin-6 in patients with advance malignancies. J Clin
212–214. Oncol 1993;11:499–506.
302. Trauth BC, Klas C, Peters AMJ, et al. Monoclonal antibody-me- 326. Welsh JB, Sapinoso LM, Su AI et al. Analysis of gene expression
diated tumor regression by induction of apoptosis. Science identifies candidate markers and pharmacological targets in prostate
1989;245:301–305. cancer. Cancer Res 2001;61:5974–5978.
303. Trillet-Lenoir V, Green J, Manegold C, et al. Recombinant granulo- 327. Welsh JB, Zarrinkar PP, Sapinoso S et al. Analysis of gene
cyte colony stimulating factor reduces the infectious complications expression profiles in normal and neoplastic ovarian tissue sam-
of cytotoxic chemotherapy. Eur J Cancer 1993;29A:319–324. ples identifies candidate molecular markers of epithelial ovarian
304. Trinchieri G. Interleukin-12 and its role in generation of Th1 cancer. Proc Natl Acad Sci U S A 2001;98:1176–1181.
cells. Immunol Today 1993;14:335–338. 328. Wetzel R, Heyneker HL, Goeddel DV, et al. Production of bio-
305. Truitt GA, Brunda MJ, Levitt D, Anderson TD, Sherman MI. The logically active N-desacetylthymosin in E. coli through expres-
therapeutic activity in cancer of interleukin-2 in combination sion of a chemically synthesized gene. Biochemistry 1980;19:
with other cytokines. Current Surv 1989; 8:875–889. 6096–6104.
306. Uckun FM, Evans WE, Forsyth CJ, et al. Biotherapy of B-cell 329. Whetton AD. The biology and clinical potential of growth factors
precursor leukemia by targeting Geninstein to CD19-associated that regulate myeloid cell production. Trends Pharmacol Sci
tyrosine kinases. Science 1995;267:886–891. 1990;11:285–289.
307. Uckun FM, Frankel A. The current status of immunotoxins: an 330. Williams DE, Dunn JT, Park LS, Frieden EA, Seiler FR, Farese
overview of experimental and clinical studies as presented at the AM, MacVittie TJ. GM-CSF/IL-3 fusion protein promotes neu-
3rd International Symposium on Immunotoxicins. Leukemia trophil and platelet recovery in sublethally irradiated rhesus mon-
1993;7:341–348. keys. Biotechnol Ther 1993;4:17–29.
308. Upton C, Mossman K, McFadden G. Encoding of a homolog of 331. Williams DE, Eisenman J, Baird A, et al. Identification of a
the IFN-gamma receptor by myxoma virus. Science 1992; ligand for the c-kit protooncogene. Cell 1990; 63:167–174.
258:1369–1372. 332. Williams GT, Smith CA, Spooncer E, Dexter TM, Taylor DR.
309. Ushio S, Namba M, Okura T, et al. Cloning of the cDNA for Hematopoietic colony stimulating factors promote cell survival
human IFN-gamma-inducing factor, expression in Escherichia by suppressing apoptosis. Nature 1990;343:76–79.
coli, and studies on the biologic activities of the protein. J 333. Winter G, Milstein C. Man-made antibodies. Nature 1991;349:
Immunol 1996;156:4274–4279. 293–299.
310. Vadhan-Raj S, Kudelka AP, Garrison L, et al. Effect of interleu- 334. Wong GG, Witek J, Temple PA, et al. Human GM-CSF: molecu-
kin-1 on carboplatin-induced thrombocytopenia in patients with lar cloning and the complementary DNA and purification of the
recurrent ovarian cancer. J Clin Oncol 1994;12:707–714. natural and recombinant proteins. Science 1985;228:810–815.
311. Vadhan-Raj S, Papadopoulous NE, Burgess MA, et al. Effects of 335. Xie MH, Aggarwal S, Ho WH, Foster J, Zhang Z, Stinson J,
PIXY321, a GM-CSF/IL-3 fusion protein, on chemotherapy- Wood WI, Goddard AD, Gurney AL. Interleukin (IL)-22, a novel
induced multilineage myelosuppression in patients with sarcoma. human cytokine that signals through the interferon receptor-re-
J Clin Oncol 1994;12:71–724. lated proteins CRF2–4 and IL-22R. J Biol Chem. 2000;275:
312. Vadhan-Raj S. PIXY321 (GM-CSF/IL-3 fusion protein) biology 31335–31339.
and early clinical development. Stem Cells 1994;12:253–261. 336. Xu F, Lupu R, Rodriguez GC, et al. Antibody induced growth
313. Valezuela P, Medina A, Rutter WJ, Ammerer G, Hall BD. inhibition is mediated through immunologically and functionally
Synthesis and assembly of hepatitis B virus surface antigen par- distinct epitopes on the extracellular domain of the erbB-2R gene
ticles in yeast. Nature 1982;298:347–350. product p185. Int J Cancer 1993;53:401–408.
314. Vallera DA. Immunotoxicins: will their clinical promise be ful- 337. Xu W, Presnell SR, Parrish-Novak J, Kindsvogel W et al. A sol-
filled? Blood 1994;83:309–314. uble class II cytokine receptor, IL-22RA2, is a naturally occur-
315. Van Der Bruggen P, Traversari C, Chomez P, et al. A gene encod- ring IL-22 antagonist. Proc Natl Acad Sci U S A.
ing an antigen recognized by cytolytic T lymphocytes on a human 2001;98:9511–16.
melanoma. Science 1991; 254:1643–1647. 338. Yan M, Marsters SA, Grewal IS, Wang H, Ashkenazi A, Dixit
316. Velculescu VE, Madden SL, Zhang L et al. Analysis of human VM. Identification of a receptor for BLyS demonstrates a crucial
transcriptomes. Nat Genet 1999;23:387–388. role in humoral immunity. Nat Immunol. 2000;1:37–41.
317. Venter JC et al. The sequence of the human genome.Science Feb 339. Yan M, Wang H, Chan B, Roose-Girma M, Erickson S, Baker T,
16, 2001: 291. Tumas D, Grewal IS, Dixit VM. Activation and accumulation of
318. Vieira P, deWaal Malefyt R, Dang W, et al. Isolation and expres- B cells in TACI-deficient mice.Nat Immunol. 2001;2:638–643.
sion of human cytokine synthesis inhibitory factor (CSIF/IL-10) 340. Yang YC, Ciarletta AB, Temple PA, et al. Human IL-3 (multi-CSF):
cDNA clones: homology to Epstein Barr virus open reading identification by expression cloning of a novel hematopoietic
frame BCRF1. Proc Natl Acad Sci USA 1991;88:1172–1176. growth factor related to human IL-3. Cell 1986;47:3–10.
Kapil Mehta et al. 83

341. Yanuck M, Carbone DP, Pendleton CD, et al. A mutant p53 tumor 349. Imamichi T, Yang J, Huang DW, Brann TW, Fullmer BA,
suppressor protein is a target for peptide-induced CD8+ cytotoxic Adelsberger JW, Lempicki RA, Baseler MW, Lane HC. IL-27, a
T cells. Cancer Res 1993;53:3257–3261. novel anti-HIV cytokine, activates multiple interferon-inducible
342. Yin M, Gopal V, Banavali S, Gartside P, Preisler H. Effects of genes in macrophages. AIDS. 2008;22(1):39–45.
IL-1 receptor antagonist on acute myeloid leukemia. Leukemia 350. Liu LH, Shan BE, Shao LL, Wang SJ. Inhibitory effect of IL-27
1992;6:898–901. gene on tumor formation activity of Eca109 cells in nude mice
343. Young DC, Wagner K, Griffin JD. Constitutive expression of the and its mechanisms. Ai Zheng. 2008;27(1):12–17.
granulocyte macrophage colony stimulating factor gene in acute 351. Kim SH, Han SY, Azam T, Yoon DY, Dinarello CA. Interleukin-32:
myeloblastic leukemia. J Clin Invest 1987;79:100–106. a cytokine and inducer of TNFalpha. Immunity 2005;22(1):
344. Yu J, Zhang L, Hwang PM, Rago C, Kinzler KW, Vogelstein B. 131–142.
Identification and classification of p53-regulated genes. Proc 352. Marcondes AM, Mhyre AJ, Stirewalt DL, Kim SH, Dinarello
Natl Acad Sci U S A 1999;96:14517–14522. CA, Deeg HJ. Dysregulation of IL-32 in myelodysplastic syn-
345. Zhu N, Leggitt D, Liu Y, Debs R. Systemic gene expression after drome and chronic myelomonocytic leukemia modulates apopto-
intravenous DNA delivery into adult mice. Science 1993; sis and impairs NK function. Proc Natl Acad Sci U S A.
261:209–211. 2008;105(8):2865–2870.
346. Zurawski G, de Vries JE.Interleukin 13, an interleukin 4-like 353. Smyth MJ, Teng MW, Sharkey J, Westwood JA, Haynes NM,
cytokine that acts on monocytes and B cells, but not on T cells. Yagita H, Takeda K, Sivakumar PV, Kershaw MH. Interleukin 21
Immunol Today. 1994;15:19–26. enhances antibody-mediated tumor rejection. Cancer Res. 2008
347. Lebedeva IV, Emdad L, Su ZZ, Gupta P, Sauane M, Sarkar D, 5;68(8):3019–3025.
Staudt MR, Liu SJ, Taher MM, Xiao R, Barral P, Lee SG, Wang 354. Luo J, Xia Q, Zhang R, Lv C, Zhang W, Wang Y, Cui Q, Liu L,
D, Vozhilla N, Park ES, Chatman L, Boukerche H, Ramesh R, Cai R, Qian C. Treatment of the established tumors induced high
Inoue S, Chada S, Li R, De Pass AL, Mahasreshti PJ, Dmitriev IP, level of local expression of IL-24 in tumor cells and resulted in
Curiel DT, Yacoub A, Grant S, Dent P, Senzer N, Nemunaitis JJ, tumor regression. Clin Cancer Res. 2008 15;14(8):2450–2457.
Fisher PB. mda-7/IL-24, novel anticancer cytokine: focus on 355. Sarkar D, Dent P, Curiel DT, Fisher PB. Acquired and innate
bystander antitumor, radiosensitization and antiangiogenic prop- resistance to the cancer-specific apoptosis-inducing cytokine,
erties and overview of the phase I clinical experience. Int J Oncol. mda-7/IL-24: not insurmountable therapeutic problems. Cancer
2007 31(5):985–1007. Biol Ther. 2008;7(1):109–112.
348. Weiss JM, Subleski JJ, Wigginton JM, Wiltrout RH. 356. Chen C, Sun L, Yu Y, Xue Y and Yang P. Amino acid-coded tag-
Immunotherapy of cancer by IL-12-based cytokine combina- ging approaches in quantitative proteomics. Expert RevProteomics
tions. Expert Opin Biol Ther. 2007;7(11):1705–21. 2007; 4(1):25–37.
5 Current concepts in immunology
ROBERT K. OLDHAM

The main function of the immune system is to protect mediate the communication between cells of the immune
the host from certain death due to numerous potential system and other organs of the body, such as the central
pathogens present in the environment. The development nervous and endocrine systems.
and maintenance of immunity is dependent on a com- This chapter presents a summary of the structure and
plex and highly sophisticated defense organization function of the immune system with emphasis in those
functionally divided into the innate and adaptive immune areas relevant to the defense against tumors and to can-
systems. cer biotherapy. The following sections will briefly
Innate immunity provides the first line of defense describe the characteristics and functions of the major
against most pathogens. Phagocytes, including neutro- cells and mechanisms that control immune responses
phils and monocytes/macrophages, and natural killer and the clinical methodologies for assessing the general
cells are the most important cell types participating in state of immunocompetence.
this form of immunity. In addition, several soluble fac-
tors, including the complement cascade, lysozyme, and
acute-phase reactants, contribute to and reinforce the
physical barriers that prevent most infectious organisms
Current Concepts of Immunity
from penetrating the body. These factors also promote All the major cells of the immune system, that is, mono-
the activation of the inflammatory reaction that contains cytes/macrophages, B and T lymphocytes, natural killer
the injury caused by an invasive infectious agent. These cells, polymorphonuclear cells, etc., originate in the
cells and factors are suitable to accomplish this function bone marrow from pluripotent stem cells [58]. Although
because their activity does not depend on a prior encoun- the exact mechanisms and mediators involved are not
ter with the antigens in the microbial agent or tumor completely understood, it is now accepted that stem
cells, and they lack the fine specificity of the cells and cells, under the influence of growth and differentiation
humoral factors of the adaptive immune system. factors such as erythropoietin, colony-stimulating fac-
Specificity and memory are two cardinal features of tors, and interleukins, differentiate into two main pro-
the adaptive immune system. The main cellular compo- genitors: stem cells that have the potential to originate
nents are the T and B lymphocytes. Each clone of these erythrocytes, eosinophils, platelets, granulocytes and
cell populations has an extraordinary and unique antigen monocytes/macrophages (GEMM-CFC); and stem cells
specificity via the expression of genetically programmed, that are the precursors for mature lymphoid cells
antigen-specific receptors on the cell surface. The adap- (L-CFU) [37, 78, 109] (Fig. 1).
tive immune system has the ability to differentiate We now know many of the factors that induce prolif-
between foreign and self-molecules and in this way, eration of the committed stem cells. Most of these
most destructive reactions against the host’s own tissues cytokines have been purified, sequenced, and are currently
are prevented. When the immune system loses its capac- produced by recombinant techniques [37, 78, 109, 134].
ity to differentiate self from nonself, autoimmunity The availability of these recombinant cytokines has
develops. contributed significantly to defining and characterizing
The innate and adaptive systems communicate with their biological activities (Fig. 1).
each other directly by cell–cell interactions and through Four separate colony-stimulating factors (CSFs) have
soluble mediators termed cytokines. For example, mac- been recognized. They are interleukin 3 (IL-3; multi-
rophages are important not only in innate immunity but CSF), granulocyte-macrophage colony-stimulating fac-
also in specific immune responses, since they can pres- tor (GM-CSF), monocyte-colon-stimulating factor
ent antigen and activate T and B cells. In turn, T cells (M-CSF) and granulocyte-stimulating factor (G-CSF)
regulate macrophages, B cells, and natural killer cell [37, 109]. These factors, together with interleukin 1 (IL-1),
activity through the synthesis and release of cytokines. interleukin 4 (IL-4), interleukin 5 (IL-5), and interleukin 6
Some of the cytokines also behave as messengers that (IL-6), are essential for the proliferation and differentiation

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 85


© Springer Science + Business Media B.V. 2009
86 Current concepts in immunology

Bone Marrow
GEMM–CFC Pluripotent Stem Cell L-CFU

GM–CSF
IL–3
CFU–GM
GM–CSF Bursa of Fabricius
IL–3 Thymus
or equivalent
IL–5
IL–1 IL–2
IL–2 IFN–γ
IL–3 IL–3 IL–4
IL–4 Eosinophil GM–CSF
IL–3 IL–6
M–CSF B
GM–CSF
G–CSF
GM–CSF IL–1
IL–3 Basophil IL–2
IL–4 LGL IL–4
EP Mφ IL–6
BFU–E

RBC
GM–CSF Pc
IL–3
IL–4 Th/i
PMN

Platelets Ts Tc Ab
Megakaryocyte

Figure 1. Origin of the cells of the immune system. All the cells of the immune system are derived from the multipotential stem
cell in the bone marrow. Proliferation, maturation, and differentiation of the different cell lineages are driven by a multitude of
cytokines. GEMM-CFC, granulocyte/erythroid/megakaryocyte/monocyte colony-forming cell: L-CFU, lymphocyte colony-form-
ing unit; CFU-GM, granulocyte/monocyte colony-forming unit; B, B lymphocyte; Th/i, T helper inducer; Ts, T suppressor; Tc, T
cytotoxic; Mo, macrophage; LGL, large granular lymphocyte; Pc, plasma cell; Ab, antibody; PMN, polymorphonuclear cell; EP,
erythropoietin; BFU-E, burst-forming unit – erythroid; RBC, red blood cell; GM-CSF, granulocyte/monocyte colony-stimulating
factor; G-CSF, granulocyte colony-stimulating factor; M-CSF, monocyte colony-stimulating factor; IL-1, interleukin 1; IL-2,
interleukin 2; IL-3, interleukin 3; IL-4, interleukin 4; IL-6, interleukin 6; IFN-γ, interferon-γ

of committed stem cells, such as GEMM-CFC, GM-CFC, Some of these cytokines (i.e., C-CSF, GM-CSF) are
and L-CFU. For example, IL-3 and GM-CSF are required already approved for the treatment of patients with
to induce GEMM-CFC to differentiate into granulocyte/ chronic severe neutropenia associated with cancer che-
monocyte-colony forming cells (GM-CFC), while motherapy, AIDS, bone marrow transplantation, myelo-
M-CSF, GM-CSF, and IL-3 are necessary for GM-CFC dysplasia, and aplastic anemia [27, 37]. Platelet growth
to differentiate into precursors of the monocytic lineage activation factors have been approved and others, along
(Fig. 1) [37, 78, 109]. Some of these cytokines act on with receptor agonists, are in clinical testing.
targeted cell lineages; for example, IL-5 regulates Lymphocyte-precursors originating from the L-CFU
eosinophilic maturation and B-cell activity while eryth- migrate to the thymus where they differentiate into
ropoietin (EP) has its predominant effects on committed mature T lymphocytes. This cell population is com-
erythroid cells (BFU-E) (Fig. 1). Macrophages, activated prised of several subpopulation, including T helper/
T lymphocytes, fibroblasts, and endothelial cells are the inducer (Th/I) lymphocytes, which modulate the activity
major sources of GM-CSF, G-CSF, M-CSF, IL-1, IL-3, of virtually all other cell types of the immune system
IL-5, and IL-6 [16, 37, 44, 51, 78, 90, 109, 134]. (Fig. 1), T-cytotoxic (Tc) lymphocytes, which are the
Robert K. Oldham 87

effectors of the cytotoxic responses against tumors, for- cytes into specific lymphoid tissues appears to be gov-
eign tissues, virus-infected cells, etc. and T-suppressor erned by complementary adhesion molecules present on
or regulatory (Ts or Treg) lymphocytes, which are lymphocytes and on endothelial cells of high endothe-
involved in the down-regulation of the immune response. lial venules (HEV) and postcapillary venules (homing
In addition to T lymphocytes, the bone marrow receptors, specialized adhesion molecules) [115, 129].
lymphocyte-precursor cells also give rise to natural The expression of some specialized adhesion molecules
killer cells (NK), which are involved in the defense by endothelial cells is induced by cytokines produced at
against tumors, and B lymphocytes (B), which after the inflammatory sites. For example, the intracellular
maturing in the bursa of Fabricius (in birds) or bursa adhesion molecule 1 (ICAM-1), which serves as a
equivalent (bone marrow in humans), migrate to periph- receptor for the leukocyte-function-associated antigen-1
eral tissues, where in response to antigens, they differ- (LFA-1) molecule present on leukocytes [5, 23, 110], is
entiate into antibody-producing plasma cells (Fig. 1). induced by IL-1 in endothelial cells. The rapid increase
Mature lymphoid cells circulate in blood, from where in these surface proteins facilitates the adhesion of leu-
they populate peripheral lymphoid tissues, such as the kocytes to the endothelium, thus accelerating the initial
paracortical areas of the lymph nodes and the periarte- stages of the inflammatory reaction (Fig. 2).
riolar sheaths of the spleen, the skin, and the mucosal In addition to traffic to and from secondary lymphoid
linings of the digestive, respiratory, and genitourinary organs, a small number of lymphocytes travel through
tracts. The migration of circulating B and T lympho- most non-lymphoid tissues of the body. This traffic

Macrophage
IL-6
TGF-β

Plt
Monocyte
IL-1
PDGF PMN TNF-a

LOCAL IMMUNE SYSTEMIC


EFFECTS RESPONSE EFFECTS

Vascular Permeability Fever


IL – 1 IL – 6 TNF – a
Leukocyte Adherence Sleep
Anticoagulant T Lymphocyte Activation Acute Phase Reactants
IL-1/ Chemotaxis Cytokine Production Insulin
PGI Synthesis Antibody Sythesis Lipoprotein Lipase
Prolif. Endothelial Cells Steroid Synthesis
Shock
Neutrophilia
Proliferation GM – CSF
TGF – β
Proteolytic Enzymes Bone Marrow Stimulation
Collagen Synthesis Appetite
T Lymphocyte Proliferation
PGE Production Hypotension

Figure 2. Cytokine participation in inflammatory and immune responses. Early monouclear cell recruitment is induced by
platelet-derived transforming growth factor β (TGF-β). Activated macrophages produce additional TGF-β and other cytokines,
including interleukin 1 (IL-1), tumor necrosis factor α (TNF-α), and interleukin 6 (IL-6), that have local and systemic effects as
well as effects on the immune response. PDGF, platelet-derived growth factor; PMN, polymorphonuclear cell; Plt, platelets
88 Current concepts in immunology

pattern is designed to optimize the interaction of foreign processes by modulating the activity of endothelial cells
antigens with the appropriate receptor specificities in T and fibroblasts, thus linking the inflammatory and the
and B cells and to assure the fast development of a spe- immune response (Fig. 2) [124].
cific immune response. This process is also facilitated
by migration of antigen presenting cells (APC) from the
peripheral tissues to local lymph nodes [115]. Following
cognitive interaction (i.e., involving antigen and class II
T lymphocytes
histocompatibility complex molecules) between APC T cells, which comprise 70–80% of all circulating lym-
and T cells and T-B cell cooperation, lymphocyte prolif- phocytes, are central to the development of normal
eration and maturation take place, with the generation immune responses. They play a critical role in cellular
and subsequent recirculation of antigen specific effector immune reactions, including delayed-type hypersensi-
and memory cells (Fig. 3). During these events, lym- tivity (DTH), resistance against certain bacteria, viruses
phoid cells secrete a number of cytokines, such as trans- and tumor, and as effector cells mediating the rejection
forming growth factor β (TGF-β), IL-1, IL-6, tumor of organ transplants [2, 54]. T cells also have important
necrosis factor α (TNF-α), and platelet-derived growth regulatory function, due to their ability to produce
factor (PDGF), that affect the inflammatory and healing cytokines that act on other T cells, B cells and

Tr GM–CSF
M–CSF Mφ B
TGF–β IL–1
IL–6

Ag II
IL–1 IL–4; IFN–γ
TGF–β
IL–6 GM–CSF

Ag
Tact
IL–2; IL–4
IFN–γ
Tind IL–2; IL–3; IL–4
IL–5; IL–6; IFN–γ Bact

IL–2; IL–4
IFN–γ

Proliferation Proliferation Proliferation


Differentiatioin Differentiatioin Differentiation
LGL

IL–1; IL–2; CSF Pc Pc


IFN–γ ; TNF–β
Tumor cell killing

Tm Th
Ts Ts Tc Tc
Ab

Figure 3. Simplified scheme of the lymphokine cascade and its role in the modulation of immune response. Various cytokines
with unique and overlapping biological activities are produced during the immune response. These cytokines act in concert to
regulate the activation, proliferation and maturation of cells participating in the specific and nonspecific arms of the immune
system. Mo, macrophage; Tr, T activated lymphocyte; Tind, T inducer; Tm, T memory; Th, T helper; B, B cell; Ts, T suppressor; Tc,
T cytotoxic; Pc, plasma cell; LGL, large granular lymphocyte; IL-1, interleukin 1; IL-2, interleukin 2; IL-3, interleukin 3; IL-4,
interleukin 4; IL-5, interleukin 5; IL-6, interleukin 6; CSF, colony-stimulating factor; TNF-β, tumor necrosis factor β/lymphoto-
xin; IFN-γ, interferon γ TGF-β, transforming growth factor β; GM-CSF, granulocyte/monocyte colony-stimulating factor; M-CSF,
monocyte colony-stimulating factor; Ag, antigen; Ab, antibody; II, class II histocompatibility complex molecules
Robert K. Oldham 89

macrophages, and by directly interacting with other


lymphocytes [18, 67, 74, 112, 113, 116]. In general APC
terms, the functions of T cells can be divided into regu-
latory and effector categories. The positive and negative
MHC
regulatory functions are mediated by different lympho- Class
cyte subsets. The positive signals are associated with the I or II Processed
helper/inducer subpopulation of T cells (Th/I) and the Antigen
negative with the suppressor T-cell subpopulations (Ts)
T cell receptor
[2, 19, 95, 105, 125]. The effector category includes the CD4
cytotoxic (killer) T cells (Tc) reactive against virally or
infected cells and foreign histocompatibility antigens, CD8 CD3
and the sensitized Th(TDTH) cells, mediating delayed
Th/I or Tc/s
hypersensitivity reactions [24, 91].
T cell subpopulations can be identified by immuno-
fluorescence or flow cytometry with the use of specific
monoclonal antibodies (mAb), which react with spe- Figure 4. A current model for T-cell antigen recognition.
Antigen-presenting cells (APC) incorporate, process, and
cific surface markers. An International Nomenclature express the modified antigen in conjunction with MHC mol-
Subcommittee has examined a large number of mAb ecules for presentation to T cells. The interaction of T helper
directed to leukocyte antigens and clustered them into (Th/i) cells with APC is class II restricted; that is, it occurs
groups of antibodies with the same reactivities (clus- when the antigen is presented to the Th/i cell together with
ters of differentiation or CD), and a number was class II molecules that interact with the T-cell receptor and
CD4 molecules, respectively. Class I restriction occurs
assigned to each group [46]. Helper/inducer T lympho- between APC expressing class I and the T-cell receptor in
cytes bear the CD4 marker, while cytotoxic/suppressor CD8+ (Tc/s) cells. In both cases the delivery of the transduc-
T cells are recognized by the presence of the CD8 tion signal for cell activation appears mediated by the CD3
marker [41, 46, 95, 96]. Other mAb, which recognize molecular complex
the T3 portion of the T-cell antigen receptor complex
(CD3), and others that recognize the CD2, react with
virtually all T cells and are widely used for the deter- Both α/β and γ/δ receptors are non-covalently associ-
mination of total T-cell numbers [41, 46, 95, 96]. In ated with the CD3 molecular complex, which is com-
human peripheral blood, there are twice as many CD4 posed of at least four polypeptide chains, γ,δ,ε,and ζ one of
as CD8, a proportion that is altered in many diseases which (the γ chain) has a long intracytoplasmic portion
including immunodeficiencies and cancer. More with several phosphorylation sites [12]. It is believed that
recently, the phenotypes of the two main subsets of the CD3 complex is involved in mediating signal trans-
CD-4 lymphocytes have been defined as helper-inducer duction during the interaction of antigen with the specific
(CDw29+) and suppressor-inducer (Treg) (CD25 + binding site in the α/β receptor (Fig. 4) [12].
FOXp3 +, CD127neg) Th/I subpopulations, respectively Antigen recognition by T cells requires that the antigen
[60, 107]. be presented to the T cells in association with the appro-
The T-cell antigen receptor (TCR) has been isolated priate major histocompatibility complex (MHC) mole-
and characterized [1, 12]. It is a disulfide-linked het- cules [32] (Fig. 4) and is amplified by co-stimulatory
erodimer composed of an α and β polypeptide chain, molecules T-helper cells recognize antigen in the context
each containing a constant and a variable (antigen binding) of class II MHC molecules expressed by antigen-present-
region somewhat similar to the structure of immuno- ing cells and B cells (class II restriction), while
globulins (Fig. 4). In a small number of T cells present T-suppressor and most cytotoxic T cells recognize anti-
in peripheral blood (0.5–10% T cells), thymus, epidermis, gen in association with class I molecules (class I restric-
and gut epithelium, a different type of T-cell receptor tion). Recent work has demonstrated that CD4 molecules
consisting of two distinct polypeptide chains (γ and δ) present on T cells bind with low affinity to certain invari-
has been recognized [92]. The majority of these cells do able regions in the class II molecules. These findings sup-
not express CD4 or CD8 on the cell surface and their port the theory that a complex involving the TCR α/β-CD3
functions are not completely understood, but they are and CD4 or CD8 molecules is formed on the surface of
believed to represent a mature functional lineage of the T cells during antigen recognition (Fig. 4). In this situ-
lymphocytes that can be activated by triggering through ation, the specificity is determined by the variable region
the γ/δ receptor. of the α/β receptor, the reaction is stabilized by binding
90 Current concepts in immunology

of CD4 and CD8 to class II or Class I molecules, respec- Antigen-activated CD4 T-cells release a number of
tively, and the activation signal is transduced by the CD3 cytokines including IL-2, gamma-interferon (IFN-γ),
complex [64, 106]. colony-stimulating factors (CSFs), B-cell growth factors
(IL-4, IL-5, IL-6), etc. [38, 40, 49, 73, 79, 87, 88, 117,
132] (Fig. 3). These cytokines, in turn, induce activa-
tion, proliferation and differentiation of other antigen-
T-helper Cells specific T and B cells resulting in a specific immune
T-helper cells were first described as the T lymphocyte response and the production of memory T cells (Fig. 3).
subpopulation that “helps” B lymphocytes mount an There is some evidence that lymphokine release from
optimal antibody response and “induces” the generation TH cells can lead to tumor regression (see Chapter 8).
of cytotoxic T cells. These T cells recognize processed Although human counterparts are not fully character-
antigen presented by macrophages or other APC in the ized, there are two well-defined subpopulations of
context of Class II (DR) products (Fig. 4). The induc- murine Th/I lymphocytes, termed TH1 and TH2 [69].
tion of lymphoid cell proliferation upon stimulation by These subpopulations are characterized by the secretion
mitogens or antigens is characterized by two distinct of different sets of cytokines, leading to different func-
phases: competence and progression. The activating tional properties [69]. Although some cytokines such as
signals (competence signals) cause the resting T cells to GM-CSF, TNF-α, and IL-3 are produced by both cell
move into the early G1 phase of the cell cycle. During types, TH1 but not TH2 clones produce IL-2, γ-interferon,
this “competence stage,” binding of antigens to the and lymphotoxin (TNF-β) [69]. In contrast, TH2 but not
TCR/CD3 (TCR complex) induces the generation of TH1 cells synthesize IL-4, IL-5 and IL-6. TH2 cells
intracellular signals, such as increases in intracellular induce growth and immunoglobulin (Ig) secretion of B
free calcium, membrane depolarization, generation of cells in response to specific antigens and polyclonal
diacylglycerol (DG) and phosphatidyl inositol (PI) activators. These functions require not only IL-4 and
turnover, activation of protein kinase C (PKC), and IL-5 synthesis but also TH2-B cell-cell interactions. In
changes in the levels of cAMP, cGMP, protein phos- addition, IL-4 is essential for IgE production, and IL-5
phorylation, and express of c-fos, c-myc, and other appears to play a role in B-cell hyperactivation observed
proto-oncogenes [13, 93, 94, 108, 111, 126]. These in mice prone to develop a severe lupus-like auto-
events (some of which are triggered by IL-1) lead to the immune syndrome.
expression of specific genes [including interleukin 2 TH1 cells appear to be involved in proliferation (but
and IL-2 receptor (IL-2R)] critical for the progression no Ig synthesis) of B cells, induction of T-cell activity,
phase [6, 126]. During the progression stage, binding and generation of cells participating in the late phase of
of IL-2 to its high-affinity receptor is a critical event delayed hypersensitivity reactions. Immune responses
leading to passage of competent T cells from early G1 characterized by predominant activation of TH1 cells
through the other phases of the cell cycle, culminating may result in strong induction of macrophage-mediated
in cell proliferation [6, 7, 29]. Other changes noted dur- cytotoxic reactions induced by IFN-α and TNF-β and
ing activation are increased numbers of interleukin 1 increased expression of Fc receptors for IgG2a in mac-
receptors (IL-IR), de novo expression of Class II mol- rophages. Thus, these responses would result in effec-
ecules, and acquisition of transferrin receptors (TR) to tive killing of target cells with intracellular viral or
ensure incorporation of iron, since this element is parasite infections and strong DTH reactions. In con-
essential for cell division [75]. trast, activation of TH2 cells lelads to immune responses
The rate of T-cell proliferation is tightly regulated characterized by high levels of antibody production.
by the transient expression of IL-2R and the limited Normal immune responses probably involve the partici-
production of IL-2. The high-affinity IL-2R pation of both cell types [69].
(HA-IL-2R), a heterodimer consisting of an α chain
(p55) associated with a β chain (p70), is rapidly inter-
nalized in the presence of the ligand and is widely rec-
ognized as the IL-2R species that mediates the
T-suppressor Cells
biological responsiveness to IL-2 [20, 53, 97, 98, 118, T-suppressor (regulatory) cells suppress (down-regulate)
122]. Small numbers of α-chain (MW 55 kD) mole- T cell activity, antibody production by B cells, DTH,
cules are normally expressed by resting CD4 + T cells. contact sensitivity, cytolytic T-cell function, prolifera-
Normally, IL-2R expression lasts for about 1 week, tion of T cells, and immune responses against tumors
and IL-2 production for 2 or 3 days. [2, 19, 105]. The Ts cells are activated during normal
Robert K. Oldham 91

responses to a variety of antigens, thereby providing a granules. The presence of calcium is the limiting factor
safety mechanism that continuously controls the magni- in this phase, since polymerization of the perforin mol-
tude of the immune response. Although T-suppressor cells ecules on the membrane of the target cell with channel
are antigen-specific, they are also able to bind antigen in formation does not occur in the presence of calcium
the absence of accessory cells or specific products of the chelators [42, 66, 71]. The granules also contain other
MHC. T-suppressor cells appear to be selectively acti- factors known to mediate cytotoxic and cytostatic effects
vated when the antigen is presented in certain routes, that on tumor cells, such as esterases and proteoglycans [89].
is, intravenously or orally, or when administered in very The third phase includes the delivery of the lethal hit
low doses (low zone tolerance). T-cell induced suppres- and death of the target cell. Following interaction with
sion might be mediated directly by Ts cell or by antigen- the cytotoxic T cell, disturbances in the ion concentra-
specific and non-antigen-specific soluble factors released tions and DNA fragmentation and fluid extrusion occur,
by them (TSF). Ts binding consists of an antigen-binding culminating in target-cell disintegration. Interestingly,
portion and an I-J molecule (28 kD) that binds to the cytolytic cells are resistant to the cytolytic mechanisms
acceptor cell through the antigen molecule and an I-J that they generate, and in this way each effector Tc may
binding site [68]. Regulatory T cells inhibit tumor immu- kill more than one target cell.
nity in some experimental systems [105], and recent Studies of Tc cells infiltrating human tumors has
clinical trials [20] utilizing antibodies to Treg cells have helped define therapeutically active T cells leading to
demonstrated that these biological drugs have clear ther- tumor regression. These cells have proven exceedingly
apeutic activity (see Chapter 6). useful in defining relevant human tumor associated antigens
for vaccine formulation.

T-cytotoxic Cells
T-cytotoxic cells were originally described as the effector
B lymphocytes
cells of specific cell-mediated cytotoxicity against B cells are responsible for humoral immunity, and, like
allografts, virus-infected cells, bacteria, tumor cells, etc. T cells, are present in peripheral blood where they rep-
The Tc lymphocytes also interact with the antigen (most resent about 10–20% of the circulating lymphocyte
commonly a foreign cell, tumor cells, or virus-infected pool. After activation, B cells mature into specific anti-
cells) through the T-cell receptor [54, 71]. The interaction body-secreting cells or plasma cells [48, 128, 133].
of specific cytotoxic T cells with the target structure Mature B cells express surface immunoglobulins (Ig)
results in lysis of the latter. “Killing” of a target cell, with identical specificity to the antibodies (Ab) they
including tumor cells, consists of a number of mecha- secrete [128]. The majority of peripheral blood B cells
nisms that can be divided in three phases [35, 36, 131]. express both IgM and IgD, while most B lymphocytes
Initially, binding between target and effector cells must present in body tissues express IgG, IgA, and IgE. Most
take place. The majority of cytotoxic T cells are CD8+ B cells also express Class II molecules in the cell mem-
and therefore recognize the antigen in the context of brane. These molecules participate during the physical
MHC 1 molecules (class I restriction [32]). In addition, cell–cell interaction that takes place during T-B cell
they can also recognize foreign class I antigens alone, cooperation. In addition, receptors for the Fc portion of
indicating that they can be effective in the destruction of IgG, the third component of complement (C3b), IL-2
allogeneic transplanted tissues. A small percentage (10%) and a large number of other markers (CD19, CD20,
of cytotoxic T cells bearing the CD4+ phenotype recog- CD21, CD22, etc.) have been found on the surface of
nize antigens in association with class II molecules and mature B lymphocytes [48, 128, 133].
might play a role in lysis of virus-infected cells [36]. The process of activation and maturation of B lym-
The second phase (“programming for lysis”) involves phocytes into plasma cells involves the interaction of
the reorganization of cytoplasmic organelles, including antigens with the specific immunoglobulins on the sur-
polarization of the microtubule organizing center and face of B lymphocytes, which triggers cell activation
tubulin and actin. This leads to an increased area of con- and proliferation. The cooperation between B cells and
tact between the target and effector cells, leading to an Th/I, macrophages, and other accessory cells, a process
increase in the efficiency of the cytolytic process. This requiring identity of Class II products of the MHC
is followed by reorientation of the cytoplasmic granules among these cells, is necessary to induce optimal B
toward the binding region, fusion to the membrane, and lymphocyte responses. However, binding of antigen to
release of lytic molecules (perforins) contained in those B cells is necessary but not sufficient to initiate antibody
92 Current concepts in immunology

production. Nonspecific maturation and differentiation ing and killing the attacking microorganisms [57, 123].
factors (i.e., cytokines) mainly produced by Th/I cells are They have also been shown to be very effective in
also involved in the activation and progression of mature destroying neoplastic cells and removing dead or injured
B cells into antibody-forming cells [49, 128]. Among cells (scavenger function). In addition, monocyte/mac-
these factors are IL-1 (produced by macrophages and rophages are critical for the development of normal
other accessory cells), IL-2, IL-4, IL-5, IL-6, IFN-γ and immune responses since they process and present anti-
other factors secreted by activated T lymphocytes [17, gen (dendritic cells) to T lymphocytes and secrete
22, 40, 44, 49, 52, 78, 79, 88, 90, 128]. Several of these cytokines that play a major role in the initiation of spe-
cytokines, including IL-1 and IL-4 are also produced by cific immune responses, such as IL-1 and IL-6 [10, 11,
B cells [22, 49]. Most of the circulating B lymphocytes 17, 22, 44, 51, 57, 90, 104, 119] (Fig. 3).
are in the resting state (G0 phase of the cell cycle), but Monocytes/macrophages have been shown to secrete
they become activated after interaction with the specific more than a hundred different molecules that mediate
antigen or antigen-presenting cells in the presence of their functions. These products include (a) enzymes with
IL-4 and IL-1, progressing to the G1 phase of the cell bactericidal capacities, such as lysozyme and lysosomal
cycle. Activated B cells undergo cell division in the hydrolases, neutral proteases (e.g., collagenase and plas-
presence of T cells and secreted cytokines. Differentiation minogen activator); (b) arachidonic acid metabolites
of proliferating B cells into plasma cells is mediated by (e.g., prostacyclin, prostaglandin E2, and leukotrienes),
IFN-γ, IL-4, IL-5 and IL-6 [40, 78, 79]. Alternatively, which have profound effects in the regulation of the
proliferating cells may return to the resting state and immune response; (c) reactive oxygen metabolites (e.g.,
remain as memory B lymphocytes (Fig. 3). superoxide, O2 radical, and H2O2), which are important
During the maturation process, they not only increase mediators in macrophage-mediated cytotoxicity; (d)
their rate of Ig synthesis and actively secrete Ig, but they complement components; (e) coagulation factors; and (f)
also switch the class of heavy chains that carry the variable cytokines, which in turn exert a multiplicity of regula-
region of Ig involved in antigen recognition [49, 128]. It tory actions including playing a critical role in antigen
has been demonstrated that a single clone of proliferat- presentation to T cells [10, 11, 17, 22, 44, 51, 57, 90, 104,
ing B cells may switch at any division. Not all the pro- 119, 123] (Fig. 3).
liferating clones mature into secreting plasma cells. Antigen-presenting cells, which include dendritic
Some return to the resting state and remain as long-lived cells, Langerhans cells, veiled cells, interdigitating cells,
memory cells [128]. and others in addition to macrophages, are characterized
by their ability to process and present antigen to T cells
in conjunction with MHC class molecules, as well as
cytokine production, resulting in T-cell activation.
Monocytes and Macrophages Macrophages incorporate antigens nonspecifically by
Monocytes are large cells (15–30 μm in diameter) that phagocytosis or by binding of immune complexes to the
comprise about 10–25% of peripheral blood mononu- Fc receptors. In contrast, activated B lymphocytes bind
clear cells (PBMC). They originate in the bone marrow antigen via the specific antigen receptor, and dendritic
from monoblasts, circulate in peripheral blood as mono- cells are likely to process antigen directly in the cell
cytes, and then enter various tissues where they are membranes. These short peptides (8–24 amino acids)
termed resident tissue macrophages (Fig. 1). The large are then linked to the antigen cleft in the class II mole-
degree of heterogeneity that exists within the macrophage cules and returned to the cell membrane for interaction
population is believed to represent different stages of the with the Th/s cells [32] (Fig. 4).
maturational process and reflect environmental condi-
tions at the tissue level, rather than distinct macrophage
subpopulations [57]. Monocytes/macrophages can be
identified by a number of methods, including morphol-
Natural Killer Cells
ogy, ingestion of particles (such as latex), histochemical Natural killer cells were first discovered by Oldham and
staining of cytoplasmic enzymes (such as nonspecific co-workers [80, 81]. Later studies defined their ability to
esterase), and by flow cytometry with a number of mAb bind and lyse sensitive tumor and virus-infected normal
that recognize markers present on their membrane (i.e., cells without the need of previous sensitization [31, 59, 84,
CD40, CD54, CD80 and 86, etc.) [46]. 121]. These cells, which constitute approximately 15% of
Monocytes/macrophages play a critical role in the peripheral blood lymphocytes, are a relatively homoge-
defense against bacterial and other infections by ingest- neous cell type identified as large granular lymphocytes
Robert K. Oldham 93

(LGL). LGL are nonphagocytic, express receptors for the in long-term cultures stimulated with IL-2 and anti-CD3
Fc portion of IgG (FcR-positive), and have a high cyto- was observed in the CD3+ CD16−; CD3− CD16+;
plasm to nucleus ratio, indented nucleus, and a few dis- CD3+ CD4− CD8−; CD16− and CD3− CD56+ cell
crete azurophilic granules in the cytoplasm [84, 121]. populations. On the contrary, the CD3+ CD4+ or CD3+
Several monoclonal antibodies have helped define the CD8+ populations present in these cultures exhibited a
surface markers and the phenotype of natural killer significantly lower level of LAK activity [77].
cells. Some human NK cells express the following T-cell Interleukin-2 mediated induction of LAK cells can be
markers: CD2, CD8; and after activation they express enhanced by the addition of IL-1α or IL-1β, probably
T10 (a marker present in thymocytes and activated T by rendering LAK precursors more susceptible to the
cells) [46, 59, 84, 121]. activity of IL-2 [14, 21]. Interestingly, recent evidence
NK cell function is regulated by stimulatory and indicates that the generation of LAK cells is dependent
inhibitory mechanisms. The generation and function of upon the expression of the p70/75 (intermediate affin-
NK cells are regulated mainly by IFN-α, IFN-γ, IL-2 and ity) IL-2 binding protein in the cell membrane. The
IL-4 [59]. IL-2 and IFNs increase the lytic activity of expression of p70 occurs in the absence of the p55 (low
mature NK cells, promote the recruitment and activation affinity, Tac) IL-2 binding protein, which is critical for
of nonlytic cells, and induce proliferation of precursors the expression of high-affinity IL-2R in activated T lym-
and mature cells either in vivo or in vitro [121]. In addi- phocytes [86]. In addition to their remarkable tumori-
tion to their nonspecific cytotoxic activity against tumor cidal activity, LAK cells are able to secrete IL-1α,
cells, NK cells may exert a regulatory role in specific IL-1β, IFN-γ, TNF-α, and TNF-β (lymphotoxin) [56].
immune responses mediated by T and B cells because of Experiments in murine models have clearly established
their ability to produce a variety of cytokines including the in vivo antitumor activity of the cells mediating LAK
IFN-α and γ, IL-2, IL-1, CSF, and TNF-α [59, 84, 121]. activity (i.e., LAK cells) when administered in conjunc-
Natural killer cells play an important role in the resis- tion with high doses of IL-2 [55, 56, 70, 103]. Based on
tance to growth and metastasis of malignant tumors [81]. these observations, a number of clinical trials in advanced
Results obtained using several in vivo experimental cancer patients were done and these have demonstrated
models suggest that NK cells are of paramount impor- clear, but limited, antitumor activity [16, 101, 127].
tance during the early stages of tumor development. For
example, NK-sensitive clones of tumor cell lines take
longer to develop into palpable tumors when injected in
the footpads of syngeneic hosts than NK-insensitive
Clinical Assessment
clones with similar doubling times [4, 39, 47]. of Immune Competence
Furthermore, selective depletion of natural killer cells in
experimental animals has been correlated with increased In Vivo
frequency of spontaneous tumor metastasis.
Delayed Type Hypersensitivity (DTH)
DTH is a test of cell-mediated immunity based on the
response against a test antigen after it is injected intrader-
Lymphokine-activated mally, or applied topically to the skin (Table 1). Reddening
and induration of the test site occur in 8–12 h, reaches a
Killer Cells peak at 48–72 h, and thereafter slowly subsides. The DTH
The lymphokine-activated killer phenomenon (LAK) lesion is characterized by the accumulation of mononu-
was originally defined as the ability of PBMC incubated clear cells in the subcutaneous and deep and superficial
for several days in vitro in the presence of IL-2 to lyse dermis [8]. It should be stressed that DTH reactions are
freshly isolated tumor cells and NK-resistant targets, complex immunological phenomena requiring the par-
such as the HL-60 cell line [33, 34, 76, 100, 102, 130]. ticipation of effector T lymphocytes as well as mono-
The LAK phenomenon, which is not MHC restricted cytes/macrophages as accessory cells. Thus, a deficit of T
[85], can be mediated by cells expressing markers pres- cells or monocyte/macrophages, or the presence of cer-
ent in both T and NK cell populations. For example, the tain serum inhibitory factors that impair T cell or mono-
presence of cell populations with LAK activity express- cyte/macrophage activity, or active suppressor cells, leads
ing CD16 (Leull, an NK marker) and/or CD56 (Leu19, to impaired DTH reactivity.
a marker present in both Tc and NK cells) has been Antigens used in DTH testing can be divided into two
reported in short-term cultures. Most of the LAK activity classes: recall antigens and neoantigens (an antigen to
94 Current concepts in immunology

Table 1. Selected immunologic tests patients, has been used as an indication of the existence
Parameter Test of alterations of immunoregulatory processes or of
defects in the production of Ig. Quantitation of the
T Cells
Total and subpopulations Flow cytometry levels of Ig is usually measured by the single radial
Responses to antigens Cell proliferation elicited by immunodiffusion method, ELISA, or by nephelometry
PHA, Con A, MLR [62, 63]. Among the antigens used to elicit primary and
Delayed-type hypersensitivity secondary antibody responses are brucella endotoxin,
Cytokine production Bioassays/RIA/ELISA
salmonella extract, hemocyanin, etc. Antigens have
T-receptor gene Southern blot analysis
rearrangement been divided into T-cell dependent or T-cell-independent
Cytotoxicity Release of radiolabeled depending upon whether or not Th/I cells are required for
compounds from target cells maximal antibody production by B cells.
B Cells
Total number Flow cytometry
Surface Ig Flow cytometry
Ig gene rearrangements
Serum immunoglobulins
Southern blot analysis
Serum electrophoresis
Reticuloendothelial System
Serum Ig classes Nephelometry, RID The in vivo phagocytic cell function can be assessed by
Reticuloendothelial system: measuring the rate of clearance from the bloodstream
Monocytes, macrophages after intravenous injection of a variety of materials
Cytokine production Bioassays, RIA, ELISA
Phagocytosis including colloidal gold, bacterial proteases, lipid emulsions,
Chemotaxis or aggregated human albumin labeled with radioactive
Tumor cell killing Cytotoxicity in vitro iodine [72, 114]. Another technique that has been used
Activation Production of oxygen radicals to study reticuloendothelial function is the Rebuck skin
window. In this procedure, a microabrasion of the skin
surface is made, followed by application of a cover slip
which the subject has never been previously exposed). to the raw area, and assessment of the accumulation of
One of the most commonly used neoantigens is 2,4-dini- macrophages onto the coverslip [45].
trochlorobenzene (DNCB), which has been used exten-
sively in cancer patients. Among the recall antigens,
tuberculin (PPD), mumps, tricophytin, and candida have In Vitro
been the most commonly used [8]. In general, several
recall antigens are studied simultaneously to ensure that Immune Cell Quantitation
a patient will have been exposed to at least one or more The accuracy in the quantitation of the levels of the dif-
of them. The interpretation of skin tests can vary widely ferent cells and cell subsets involved in the immune
between studies, since antigen concentration, reader response has improved dramatically with the advent of
variability, patient’s prior exposure to the antigen, boosting the monoclonal antibody technology. It is possible to
effects of repeated antigen administration, time course accurately estimate the numbers of monocytes/mac-
of the reaction, and definition of a positive reaction are rophages, Th/I, Tc, and Ts (regulatory) lymphocytes, LGL,
all variables that can influence the final assessment of a B lymphocytes, granulocytes, etc. with the use of flow
positive or negative response. cytometry using mAb that recognize specific markers
Of the available agents, only DNCB has been a con- on the surfaces of individual cell types [46, 99].
sistently useful skin test agent. However, DNCB is an
awkward reagent to employ, as repeated testing will
clearly yield anamnestic responses. Thus, only the original Lymphoproliferative Responses
test on an individual can provide clear information on This has been one of the most widely applied tests for the
the responsiveness to a neoantigen. determination of lymphocyte function. It is based on the
property of lymphocytes to undergo blast transformation
and proliferate in response to mitogenic or antigenic stim-
Humoral Immunity ulation [61]. The most commonly used mitogens include
phytohemagglutinin (PHA) and concanavalin A (Con A),
The testing of the levels of total and different classes of which primarily stimulate T cells, and pokeweed mitogen
Ig, as well as the primary or secondary antibody (PWM), which stimulates both T and B cells. Whereas
responses to a variety of antigens in the sera of cancer mitogens nonspecifically activate broad subpopulations of
Robert K. Oldham 95

cells, antigens activate specifically sensitized antigen- been developed for their quantitation in the clinical set-
reactive clones. The assay can be performed with whole ting [28]. For example, the measurement of IL-2 is
blood but most laboratories usually first isolate PBMC based on (a) the fact that T lymphocytes produce IL-2 in
(from whole blood, lymph nodes, or tumor specimens) by response to mitogenic and antigenic stimulation and (b)
density-gradient fractionation on Ficoll-Hypaque fol- the ability of IL-2 to induce lymphocyte proliferation
lowed by incubation with the appropriate mitogen at vari- and maintain in vitro T cell lines (called IL-2 dependent
ous concentrations. Seventy-two hours later cultures are cell lines). The first step is the generation of cell culture
pulsed with radiolabeled [3H]thymidine, which is incorpo- supernatants containing IL-2 by incubating PBMC with
rated into the cellular DNA. Cells are then harvested and mitogens (usually PHA or Con A) for 24–48 h. The IL-2
the radioactivity quantitated in a liquid scintillation coun- content in these supernatants is then determined by their
ter and expressed as counts per minute (cpm). A similar ability to support the in vitro growth of an IL-2 dependent
technique has been extensively employed to measure pro- T-cell line, as assessed by [3H]thymidine incorporation
liferative responses to antigenic stimulation. In these studies, after 24–48 h of culture [30]. Currently, the concentrations
mitogen is replaced by the specific antigen under study of a variety of cytokines, including IL-1, IL-2, IL-4,
and longer incubation periods are usually required [61]. IL-6, GM-CSF, TNF-α, IFN-γ, etc., are measured using
Antigen responses are usually lower in magnitude that commercially available immunoassays [radioimmuno-
those observed with mitogens, since they represent the assay (RIA) and/or ELISA], which employ specific
activation of specific T-cell clones, rather than most lym- monoclonal antibodies (see Chapter 8).
phocytes as is the case with mitogens.
Another proliferative response assay is the mixed leu-
kocyte reaction (MLR). This test is based in the ability of Immunoglobulin Production
T cells to proliferate in response to alloantigenic stimula- Immunoglobulin production can be assessed in vitro by
tion (that is cells expressing different MHC antigens). exposing a suitable effector cell population (e.g., PBMC)
This is the in vitro correlate of in vivo graft rejection, and to either a polyclonal B-cell mitogen (e.g., PWM) or spe-
is usually performed by incubating PBMC obtained from cific antigen and following a period of incubation, detect-
the patient (responder cells) with irradiated PBMC ing the immunoglobulin produced. Secreted Ig can be
obtained form one or more histoincompatible individuals detected either in the culture supernatants by using
(stimulator cells) for 6 days. Response is measured by ELISA or radial immunodiffusion (RID) techniques.
[3H]thymidine incorporation as described above [61].

Phagocytic Cell Function


Cytotoxicity
Both mononuclear (e.g., monocytes/macrophages)
Cytotoxic reactions mediated by antigen-specific T and polymorphonuclear (PMN; e.g., granulocytes) leu-
cells, NK cells, antibody-dependent killer cells, lym- kocytes exhibit phagocytic activity. Well-established tech-
phokine-activated killer cells (LAK), and activated niques exist for the assessment of monocyte phagocytosis,
macrophages are measured most commonly in microcy- hemotaxis (motility), and response to lymphokines (e.g.,
totoxicity assays [9, 30]. In general, target cells are migration inhibition factors, MIF) [25, 26, 65, 70, 108].
labeled with a radioactive element able to bind to some Established techniques also exist for the assessment of
intracytoplasmic structure, which is released to the monocyte/macrophage cytotoxicity [50]. Various tests are
media when the cell dies. Fixed numbers of labeled tar- also available to measure PMN functions, including tests of
get cells are incubated in microtiter plates with different phagocytic cell activity, bactericidal capacity, and the ability
numbers of effector cells. The assays last for from 4 to to take up and reduce nitroblue tetrazolium dye (NBT) [3].
48 h, after which the supernatants are collected and the
radioactivity counted. The extent of radioactivity pres-
ent in the supernatants is in direct relationship with the Immunoregulatory Cell Functions
amount of killing. The results are expressed as percent
These assays are technically difficult to perform and are
cytotoxicity [9, 31].
not usually part of the routine assessment of immune
competence. Three broad classes of immunoregulatory
Lymphokine Production cell assays have been utilized:
As the roles of some of the soluble mediators in the 1. Coculture assays. In these assays effector cells (e.g.,
immune response have become clearer, assays have B cells, T cells) are exposed to a polyclonal activator
96 Current concepts in immunology

(e.g., PWM, MLR) in the absence or presence of test Baltimore, Maryland) contributed much of this chapter in
cells. The test-cell population might induce either the second edition, which was revised by the current
enhancement (e.g., increases in antibody production author in the third, fourth, and fifth editions.
or enhanced MLR responses) or reduction (suppres- I would especially like to acknowledge the assistance
sion) of the response of the effector cells [125]. of Dr. Richard S. Schulof, who co-authored this chapter
2. Mitogen-induced suppressor T-cell activity. In this in the second edition, but died in an accident during the
system suppressor cells are generated from resting T preparation of the third edition.
cells by exposing them to a polyclonal T-cell lectin
such as Con A. Con A induced suppressor cells are
then added in coculture to an effector-cell assay as
described above. References
3. Adherent suppressor cells. The presence of suppres- 1. Allison JP, Lanier LL. Structure, function and serology of the T-cell
sor monocytes can be assessed by either a positive or antigen receptor complex. Annu Rev Immunol 1987; 5:503–540.
2. Asherson GL, Colizzi V, Zembala M. An overview of T-suppressor
negative effect. In the former situation, removal of
cell circuits. Annu Rev Immunol 1986; 4:37–68.
adherent cells (e.g., monocytes) leads to augmenta- 3. Baehner RL, Boxer LA, Davis J. The biochemical basis of nitroblue
tion of effector cell function (e.g., lymphoproliferative tetrazolium reduction in normal human and chronic granulomatous
responses to mitogens), while in the latter case, read- disease polymorphonuclear leukocytes. Blood 1976; 48:309–313.
dition of adherent cells leads to suppression of the 4. Barlozzari T, Reynolds CW, Herberman RB. In vivo role of natural
killer cells: involvement of large granular lymphocytes in the
response [120].
clearance of tumor cells in anti-asialo GM1-treated rats. J Immunol
1983; 131:1024–1027.
5. Bevilacqua MP, Pober JS, Mendrick DL, Cotran RS, Jr.
Quality Control of in vitro Assays Identification of an inducible endothelial-leukocyte adhesion mol-
In practice, it has been difficult to standardize lymphop- ecule. Proc Natl Acad Sci USA 1987; 84:9238–9242.
6. Bich Thuy LT, Dukovich M, Peffer NJ, et al. Direct activation of
roliferative or cytolytic assays to insure the universal human resting T cells by IL 2: The role of an Il 2 receptor distinct
validity and reproducibility of results. A number of con- from the Tac protein. J Immunol 1987; 139:1550–1556.
ditions affect the comparability of in vitro assays as per- 7. Bierer BE, Mentzer SJ, Greenstein JL, Burakoff SJ. The role of
formed in different laboratories such as assay incubation functional cell surface antigens in T cell activation. Year Immunol
time, presence or absence of physiological buffers and/ 1986; 2:39–59.
8. Buckley CE III. Delayed hypersensitivity skin testing. In: Rose
or serum supplementation, choice of methodology to NR, Friedman H, Fahey JL, eds. Manual of clinical laboratory
prepare the responder cell population (i.e., purified immunology. Washington, DC: American Society for Microbiology,
lymphocytes or unfractionated mononuclear cells), and 1986; 259–273.
the purity and/or concentrations of mitogen/antigen, or 9. Carpenter CB. Lymphocyte-mediated cytotoxicity. In: Rose NR,
Friedman H, Fahey JL, eds. Manual of clinical laboratory immunol-
radioactive label.
ogy. Washington, DC: American Society of Microbiology, 304–307.
In addition, various in vivo confounding variables, 10. Ceuppens JL, Baroja ML, Lorre K, et al. Human T cell activation
such as the influence of concurrently administered drugs, with phytohemagglutinin. The function of IL-6 as an accessory
or seasonal and/or diurnal effects, provide further sources signal. J Immunol 1988; 141:3868–3874.
of assay variability. A number of procedures have been 11. Chiu CP, Moulds C, Coffman RL, et al. Multiple biological activi-
ties are expressed by a mouse interleukin 6 cDNA clone isolated
employed in an attempt to minimize interassay variabil-
from bone marrow stromal cells. Proc Natl Acad Sci USA 1988;
ity such as the use of cryopreserved reference lympho- 85:7099–7103.
cyte preparations [43, 82] or indices based on results 12. Clevers H, Alarcon B, Wileman T, Terhorst C. The T cell receptor/
from a panel of reference lymphocytes [15]. Nevertheless, CD3 complex: a dynamic protein ensemble. Annu Rev Immunol
because of the inherent variability of in vitro assays, only 1988; 6:629–662.
13. Crabtree GR. Contingent genetic regulatory events in T lympho-
limited successes have been achieved in clinical trials to cyte activation. Science 1989; 243:355–361.
date. Indeed, neither appropriate guidelines for interpret- 14. Crump WL 3d, Own Schaub LB, Grimm EA. Synergy of human
ing the results of clinical trials employing immunorestor- recombinant interleukin 1 with interleukin 2 in the generation of
ative agents nor identified statistically significant end lymphokine-activated killer cells. Cancer Res 1989; 49:149–153.
points upon which to select appropriate sample sizes for 15. Dean JH, Connor R, Herberman RB, et al. The relative prolifera-
tion index as a more sensitive parameter for evaluating lymphopro-
such clinical trials currently exist [83]. liferative responses of cancer patients to mitogens and alloantigens.
Int J Cancer 1977; 20:359–370.
Acknowledgment Dr. Susana A. Serrate-Sztein (National 16. Dillman RO, Oldham RK, Tauer KW, et al. Continuous interleukin-2
Institutes of Health, Bethesda, Maryland) and Dr. Marcelo and lymphokine activated killer cells for advanced cancer: an
NBSG trial. J Clin Onc 1991; 9:1233–1240.
B. Sztein (University of Maryland School of Medicine,
Robert K. Oldham 97

17. Dinarello CA. Interleukin-1 and its biologically related cytokines. 39. Hanna N, Fidler IJ. Role of natural killer cells in the destruction of
Adv Immunol 1989; 44:153–206. circulating tumor emboli. J Natl Cancer Inst 1980; 65:801–809.
18. Dinarello CA, Mier JW. Lymphokines. N Engl J Med 1987; 40. Harada N, Matsumoto M, Koyama N, et al. T cell replacing factor/
317:940–945. interleukin 5 induces not only B-cell growth and differentiation,
19. Dorf ME, Benacerraf B. Suppressor cells and immunoregulation. but also increased expression of interleukin 2 receptor on activated
Annu Rev Immunol 1984; 2:127–157. B-cells. Immunol Lett 1987; 15:205–215.
20. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer 41. Haynes BF. Human T lymphocyte antigens as defined by monoclonal
therapy following non-ablative but lymphodepleting chemotherapy antibodies. Immunol Rev 1981; 57:127–161.
for the treatment of patients with refractory metastatic melanoms. 42. Henkart PA. Mechanism of lymphocyte-mediated cytotoxicity.
J Clin Onc 2005; 23:2346–2357. Annu Rev Immunol 1985; 3:31–58.
21. Dukovich M, Wano Y, Le thi BT, et al. A second human interleu- 43. Herberman RB, Thurman GB. Approaches to the immunological
kin-2 binding protein that may be a component of high-affinity monitoring of cancer patients treated with natural or recombinant
interleukin-2 receptors. Nature 1987; 327:518–522. interferons. J Biol Response Modif 1983; 2:548–562.
22. Durum SK, Schmidt JA, Oppenheim JJ. Interleukin 1: an immuno- 44. Hirano T, Taga T, Yamasaki K, et al. A multifunctional cytokine
logical perspective. Annu Rev Immunol 1985; 3:263–287. (IL-6/BSF-2) and its receptor. Int Arch Allergy Appl Immunol
23. Dustin ML, Springer TA. Lymphocyte function-associated antigen-1 1989; 88:29–33.
(LFA-1) interaction with intercellular adhesion molecule- (ICAM-1) 45. Hong R. Immunodeficiency. In: Rose NR, Friedman H, Fahey JL,
is one of at least three mechanisms for lymphocyte adhesion to eds. Manual of clinical laboratory immunology. Washington, DC:
cultured endothelial cells. J Cell Biol 1988; 107:321–331. American Society for Microbiology, 1986; 702–722.
24. Dvorak HF, Galli SJ, Dvorak AM. Cellular and vascular manifes- 46. IUIS-WHO Nomenclature Subcommittee. Announcement. J
tations of cell-mediated immunity. Hum Pathol 1986; 17: Immunol 1985; 134:659–660.
122–137. 47. Kawase I, Urdal DL, Brooks CG, Henney CS. Selective depletion
25. Fauci AS, Pratt KR. Activation of human B lymphocytes. I. Direct of NK cell activity in vivo and its effect on the growth of
plaque-forming cell assay for the measurement of polyclonal acti- NK-sensitive and NK-resistant tumor cell variants. Int J Cancer
vation and antigenic stimulation of human B lymphocytes. J Exp 1982; 29:567–574.
Med 1976; 144:674–684. 48. Kehrl JH, Muraguchi A, Butler JL, et al. Human B cell activation,
26. Gallin JI, Quie PG. Leukocyte chemotaxis: methods, physiology proliferation and differentiation. Immunol Rev 1984; 78:75–96.
and clinical implications. New York: Raven Press, 1978. 49. Kishimoto T. Factors affecting B-cell growth and differentiation.
27. Ganser A, Ottmann OG, Erdmann H, et al. The effect of recombi- Annu Rev Immunol 1985; 3:133–157.
nant human granulocyte-macrophage colony-stimulating factor on 50. Kleinerman ES, Schroit AJ, Fogler WE, Fidler IJ. Tumoricidal
neutropenia and related morbidity in chronic severe neutropenia. activity of human monocytes activated in vitro by free and lipo-
Ann Intern Med 1989; 111:887–892. some-encapsulated human lymphokines. J Clin Invest 1983;
28. Gearing AJ, Johnstone AP, Thorpe R. Production and assay of the 72:304–315.
interleukins. J Immunol Methods 1985; 83:1–27. 51. Koj A. The role of interleukin-6 as the hepatocyte stimulating factor
29. Geller RL, Gromo G, Inverardi L, et al. Stepwise activation of T in the network of inflammatory cytokines. Ann NY Acad Sci 1989;
cells. Role of the calcium ionophore A23187. J Immunol 1987; 557:1–8.
139:3930–3934. 52. Kovacs EJ, Beckner SK, Longo DL, et al. Cytokine gene expres-
30. Gillis S. Interleukin 2: biology and biochemistry. J Clin Immunol sion during the generation of human lymphokine-activated killer
1983; 3:1–13. cells: Early induction of interleukin 1β by interleukin 2. Cancer
31. Goldfarb RH, Serrate SA. Natural killer cells. In: Yoshida T, ed. Res 1989; 49:940–944.
Investigation of cell-mediated immunity. New York: Churchill- 53. Kumar A, Moreau JL, Baran D, Theze J. Evidence for negative
Livingston, 1985; 65–80. regulation of T cell growth by low affinity interleukin 2 receptors.
32. Grey Hm, Sette A, Buus S. How T cells see antigen. Sci Am 1989; J Immunol 1987; 138:1485–1493.
261:56–64. 54. Kupfer A, Singer SJ. Cell biology of cytotoxic and helper T cell
33. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine- functions: immunofluorescence microscopic studies of single cells
activated killer cell phenomenon. Lysis of natural killer-resistant and cell couples. Annu Rev Immunol 1989; 7:309–337.
fresh solid tumor cells by interleukin 2-activated autologous human 55. Lafreniere R, Rosenberg SA. Adoptive immunotherapy of murine
peripheral blood lymphocytes. J Exp Med 1982; 155:1823–1841. hepatic metastases with lymphokine activated killer (LAK) cells
34. Grimm EA, Owen Schaub LB, Loudon WG, Yagita M. and recombinant interleukin 2 (RIL 2) can mediate the regression
Lymphokine-activated killer cells. Induction and function. Ann of both immunogenic and nonimmunogenic sarcomas and an ade-
NY Acad Sci 1988; 532:380–386. nocarcinoma. J Immunol 1985; 135:4273–4280.
35. Gromo G, Geller RL, Inverardi L, Bach FH. Signal requirements 56. Lafreniere R, Rosenberg SA. Successful immunotherapy of murine
in the step-wise functional maturation of cytotoxic T lymphocytes. experimental hepatic metastases with lymphokine-activated
Nature 1987; 327:424–426. killer cells and recombinant interleukin 2. Cancer Res 1985; 45:
36. Gromo G, Inverardi L, Geller RL, et al. The stepwise activation of 3735–3741.
cytotoxic T lymphocytes. Immunol Today 1987; 8:259–261. 57. Lasser A. The mononuclear phagocytic system: a review. Hum
37. Groopman JE, Molina JM, Scadden DT. Hematopoietic growth pathol 1983; 14:108–126.
factors. Biology and clinical applications. N Engl J Med 1989; 58. Lipton JN, Nathan DJ. Interactions between lymphocytes and mac-
321:1449–1459. rophages in hematopoiesis. In: Golde DW, Takaku F, eds. Hematopoietic
38. Han X, Itoh K, Balch CM, Pellis NR. Recombinant interleukin 2 stem cells. New York: Marcel Dekker, 1980; 145–202.
(RIL-4) inhibits interleukin 2-induced activation of peripheral 59. Lotzova E, Herberman RB. Immunobiology of natural killer cells.
blood lymphocytes. Lymphokine Res 1988; 7:227–235. Boca Raton, FL: CRC Press, 1986.
98 Current concepts in immunology

60. Liu W, et al. CD127 expression inversely correlates with FoxP3 80. Oldham RK. Natural killer cells: history and significance. J Biol
and suppressive function of human CD4(+) Treg cells. J Exp Med Response Modif 1982; 1:217–231.
2006; 203:1701–1711. 81. Oldham RK. NK cells: artifact to reality, an odyssey in biology.
61. Maluish AE, Strong DM. Lymphocyte proliferation. In: Rose NR, Can Metas Rev 1983; 2:232–336.
Friedman H, Fahey JL, eds. Manual of clinical laboratory immu- 82. Oldham RK, Gail MH, Baker MA, et al. Immunological studies in
nology. Washington, DC: American Society of Microbiology, a double blind randomized trial comparing intrapleural BCG
1986; 274–281. against placebo in patients with resected stage I non-small cell
62. Mancini G, Carbonara AO, Heremans JF. Immunochemical quantita- lung cancer. Cancer Immunol Immunother 1982; 13:164–173.
tion of antigens by single radial immunodiffusion. Immunochemistry 83. Oldham RK, Weese JL, Herberman RB, et al. Immunological
1965; 2:235–254. monitoring and immunotherapy in carcinoma of the lung. Int J
63. Meriney DK. Methodology of immunologic assays relating to Cancer 1976; 18:739–749.
humoral components. In: Grieco MH, Meriney DK, eds. 84. Ortaldo JR, Herberman RB. Heterogeneity of natural killer cells.
Immunodiagnosis for clinicians. Chicago, IL: Year Book Medical Annu Rev Immunol 1984; 2:359–394.
Publishers, 1983; 19–39. 85. Ortaldo JR, Mason A, Overton R. Lymphokine-activated killer
64. Meuer SC, Acuto O, Hercent T, Schlossman SF, Reinherz EL. The cells. Analysis of progenitors and effectors. J Exp med 1986;
human T-cell receptor. Annu Rev Immunol 1984; 2:23–50. 164:1193–1205.
65. Michel RH, Pancake SJ, Noseworthy J, Karnovsky ML. 86. Owen-Schaub L, Yagita M, Tsudo M, et al. Evidence for distinct
Measurement of rates of phagocytosis: the use of cellular mono- IL-2 receptors in induction versus maintenance of LAK function.
layers. J Cell Biol 1969; 40:216–224. Ann NY Acad Sci 1988; 532:480–481.
66. Millard PJ, Henkart MP, Reynolds CW, Henkart PA. Purification 87. Paetkau V, Bleackley RC, Riendeau D, et al. Toward the molecu-
and properties of cytoplasmic granules from cytotoxic rat LGL lar biology of IL-2. Contemp Top Mol Immunol 1985; 10:35–61.
tumors. J Immunol 1984; 132:3197–3204. 88. Palacios R, Henson G, Steinmetz M, McKearn JP. Interleukin-3
67. Miyajima A, Miyatake S, Schreurs J, et al. Coordinate regulation supports growth of mouse pre-B-cell clones in vitro. Nature
of immune and inflammatory responses by T cell-derived lym- 1984; 309:126–131.
phokines. FASEB J 1988; 2:2462–2473. 89. Pasternack MS, Verret CR, Liu MA, Eisen HN. Serine esterase in
68. Moller G. Concanavalin-A-activated lymphocytes suppress cytolytic T lymphocytes. Nature 1986; 322:740–743.
immune responses in vitro but are helper cells in vivo. Scand J 90. Perlmutter DH. IFNβ2/IL-6 is one of several cytokines that mod-
Immunol 1985; 21:31–34. ulate acute phase gene expression in human hepatocytes and
69. Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns human macrophages. Ann NY Acad Sci 1989; 557:332–342.
of lymphokine secretion lead to different functional properties. 91. Poulter LW, Seymour GJ, Duke O, et al. Immunohistological
Annu Rev Immunol 1989; 7:145–173. analysis of delayed-type hypersensitivity in man. Cell Immunol
70. Mule JJ, Shu S, Rosenberg SA. The antitumor efficacy of lym- 1982; 74:358–369.
phokine-activated killer cells and recombinant interleukin 2 in 92. Raulet DH. The structure, function, and molecular genetics of the
vivo. J Immunol 1985; 135:646–652. gamma/delta T cell receptor. Annu Rev Immunol 1989; 7: 175–207.
71. Muller Eberhard HJ. The molecular basis of target cell killing by 93. Reed JC, Alpers JD, Nowell PC, Hoover RG. Sequential expres-
human lymphocytes and of killer cell self-protection. Immunol rev sion of proto-oncogenes during lectin-stimulated mitogenesis of
1988; 103:87–98. normal human lymphocytes. Proc Natl Acad Sci USA 1986;
72. Munthe-Kaas AC, Kaplan G. Endocytosis by macrophages. In: 83:3982–3986.
Carr I, Daems WT, eds. The reticuloendothelial system: a compre- 94. Reed JC, Prystowsky MB, Kern JA, et al. Regulation of proto-
hensive treatise. New York: Plenum, 1980; 19–55. oncogene expression during lymphocyte activation and prolifera-
73. Nagler A, Lanier LL, Phillips JH. The effects of IL-4 on human tion. In: Gupta S, Paul WE, Fauci AS, eds. Advances in experimental
natural killer cells. A potent regulator of IL-2 activation and prolif- medicine and biology. New York: Plenum, 1986; 249–262.
eration. J Immunol 1988; 141:2349–2351. 95. Reinherz EL, Schlossman SF. Current concepts in immunology:
74. Nathan CF. Secretory products of macrophages. J Clin Invest regulation of the immune response – inducer and suppressor
1987; 79:319–326. T-lymphocyte subsets in human beings. N Engl J Med 1980;
75. Neckers LM, Cossman J. Transferrin receptor induction in mito- 303:370–373.
gen-stimulated human T lymphocytes is required for DNA synthe- 96. Reinherz EL, Schlossman SF. The characterization and function
sis and cell division and is regulated by interleukin 2. Proc Natl of human immunoregulatory T lymphocyte subsets. Immunol
Acad Sci USA 1983; 80:3494–3498. Today 1981; 2:69–73.
76. Ochoa AC, Gromo G, Alter BJ, et al. Long-term growth of lym- 97. Robb RJ, Greene WC. Internalization of interleukin 2 is mediated
phokine-activated killer (LAK) cells: role of anti-CD3, beta-IL 1, by the beta chain of the high-affinity interleukin 2 receptor. J Exp
interferon-gamma and -beta. J Immunol 1987; 138:2728–2733. Med 1987; 165:1201–1206.
77. Ochoa AC, Hasz DE, Rezonzew R, et al. Lymphokine-activated 98. Robb RJ, Rusk CM, Yodoi J, Greene WC. Interleukin 2 binding
killer activity in long-term cultures with anti-CD3 plus interleukin molecule distinct from the Tac protein: analysis of its role in for-
2: Identification and isolation of effector subsets. Cancer Res mation of high-affinity receptors. Proc Natl Acad Sci USA 1987;
1989; 49:963–968. 84:2002–2006.
78. OGarra A, Umland S, DeFrance T, Christiansen J. ‘B-cell factors’ 99. Rocklin RE, Meyers OL, David JR. An in vitro assay for cellular
are pleiotropic. Immunol Today 1988; 9:45–54. hypersensitivity in man. J Immunol 1970; 104:95–102.
79. OGarra A, Warren DJ, Sanderson CJ, et al. Interleukin-4 (B cell 100. Rosenberg SA, Eberlein TJ, Grimm EA, et al. Development of
growth factor-II/eosinophil differentiation factor) is a mitogen and long-term cell lines and lymphoid clones reactive against murine
differentiation factor for preactivated murine B lymphocytes. Curr and human tumors: a new approach to the adoptive immunother-
Top Microbiol Immunol 1986; 132:133–141. apy of cancer. Surgery 1982; 92:328–336.
Robert K. Oldham 99

101. Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the monocytes by interleukin-2 (IL-4). Agents Actions 1989;
systemic administration of autologous lymphokine-activated 26:199–200.
killer cells and recombinant interleukin-2 to patients with meta- 118. Teshigawara K, Wang HM, Kato K, Smith KA. Interleukin 2
static cancer. N Engl J Med 1985; 313:1485–1492. high-affinity receptor expression requires two distinct binding
102. Rosenberg SA, Mule JJ. Immunotherapy of cancer with lym- proteins. J Exp Med 1987; 165:223–238.
phokine-activated killer cells and recombinant interleukin-2. 119. Tosato G, Pike SE. Interferon-beta 2/interleukin 6 is a co-stim-
Surgery 1985; 98:437–444. ulant for human T lymphocytes. J Immunol 1988; 141:
103. Rosenberg SA, Mule JJ, Spiess PJ, et al. Regression of estab- 1556–1562.
lished pulmonary metastases and subcutaneous tumor mediated 120. Tracey DE. Macrophage mediated injury. In: Rose NR, Siegel
by the systemic administration of high-dose recombinant inter- BV, eds. The reticuloendothelial system: a comprehensive treatise.
leukin 2. J Exp Med 1985; 161:1169–1188. New York: Plenum, 1983; 77–101.
104. Rosenblum MG, Donato NJ. Tumor necrosis factor α: A multifac- 121. Trinchieri G, Perussa B. Human natural killer cells: biologic and
eted peptide hormone. CRC Crit Rev Immunol 1989; 9:21–44. pathologic aspects. Lab Invest 1984; 50:489–513.
105. Schatten S, Granstein RD, Drebin JA, Greene MI. Suppressor T 122. Tsudo M, Kozak RW, Goldman CK, Waldmann TA. Demonstration
cells and the immune response to tumors. CRC Crit Rev Immunol of a non-Tac peptide that binds interleukin 2: a potential partici-
1984; 4:335–379. pant in a multichain interleukin 2 receptor complex. Proc Natl
106. Schwartz RH. T-lymphocyte recognition of antigen in association Acad Sci USA 1986; 83:9694–9698.
with gene products of the major histocompatibility complex. 123. Unanue ER. Antigen-presenting function of the macrophage.
Annu Rev Immunol 1985; 3:237–261. Annu Rev Immunol 1984; 2:395–428.
107. Seddiki N, et al. Expression of interleukin (IL)-2 and IL-7 recep- 124. Wahl SM, McCartney-Francis N, Mergenhagen SE. Inflammatory
tors discriminates between human regulatory and activated T cells. and immunomodulatory roles of TGF-β. Immunol Today 1989;
J Exp Med 2006; 203:1693–1700 10:258–261.
108. Shipp MA, Reinherz EL. Differential expression of nuclear 125. Waldmann TA, Broder S. Suppressor cells in the regulation of the
proto-oncogenes in T cells triggered with mitogenic and non- immune response. Prog Clin Immunol 1977; 3:155–199.
mitogenic T3 and T11 activation signals. J Immunol 1987; 126. Weiss A, Imboden JB. Cell surface molecules and early events involved
139:2143–2148. in human T lymphocyte activation. Adv Immunol 1987; 41:1–38.
109. Sieff CA. Hematopoietic growth factors. J Clin Invest 1987; 127. West WH, Tauer KW, Yannelli JR, et al. Constant-infusion
79:1549–1557. recombinant interleukin-2 in adoptive immunotherapy of
110. Smith CW, Rothlein R, Hughes BJ, et al. Recognition of an advanced cancer. N Engl J Med 1987; 316:898–905.
endothelial determinant for CD 18-dependent human neutrophil 128. Whitlock C, Denis K, Robertson D, Witte O. In vitro analysis of
adherence and transendothelial migration. J Clin Invest 1988; murine-B-cell development. Annu Rev Immunol 1985; 3:213–235.
82:1746–1756. 129. Woodruff JJ, Clarke LM, Chin YH. Specific cell-adhesion mech-
111. Smith KA. Dissection of the molecular events occurring during T anisms determining migration pathways of recirculating lympho-
cell cycle progression. In: Gupta S, Paul WE, and Fauci AS, eds. cytes. Annu Rev Immunol 1987; 5:201–222.
Advances in experimental medicine and biology. New York: 130. Yang SC, Owen Schaub L, Grimm EA, Roth JA. Induction of
Plenum, 1986; 125–128. lymphokine-activated killer cytotoxicity with interleukin-2 and
112. Smith KA. The interleukin 2 receptor. Adv Immunol 1988; tumor necrosis factor-alpha against primary lung cancer targets.
42:165–179. Cancer Immunol Immunother 1989; 29:193–198.
113. Smith KA. Interleukin-2: inception, impact, and implications. 131. Young JD, Liu C. Multiple mechanisms of lymphocyte mediated
Science 1988; 240:1169–1176. killing. Immunol Today 1988; 9:140–144.
114. Spencer RP, Pearson HH. Radionuclide studies of the spleen. 132. Zlotnik A, Fischer M, Roehm N, Zipori D. Evidence for effects of
Cleveland: CRC Press, 1975. interleukin 4 (B cell stimulatory factor 1) on macrophages:
115. Springer TA, Dustin ML, Kishimoto TK, Marlin SD. The lym- enhancement of antigen presenting ability of bone marrow-derived
phocyte function-associated LFA-1, CD2, and LFA-3 molecules: macrophages. J Immunol 1987; 138:4275–4279.
cell adhesion receptors of the immune system. Annu Rev 133. Zola H. The surface antigens of human B lymphocytes. Immunol
Immunol 1987; 5:223–252. Today 1987; 8:308–310.
116. Taniguchi T. Regulation of cytokine gene expression. Annu Rev 134. Zucali JR, Broxmeyer HE, Gross MA, Dinarello CA. Recombinant
Immunol 1988; 6:439–464. human tumor necrosis factors alpha and beta stimulate fibroblasts
117. Te Velde AA, yard BA, Klomp JP, et al. Modulation of pheno- to produce hemopoietic growth factors in vitro. J Immunol 1988;
typic and functional properties of human peripheral blood 140:840–844.
6 Therapeutic approaches to cancer-associated
immune suppression
ROBERT K. OLDHAM

It is now well established that many cancer patients both chemical and biological, that were administered
exhibit in vivo and in vitro evidence of immune suppres- either to restore depressed immunity to normal, or to
sion, which often correlates with tumor-cell burden, stage prevent the deterioration of immune competence due to
of disease, and prognosis. Except in transplantation surgery, radiation therapy, or chemotherapy. BRMs that
associated cancers, cancer-associated immune suppres- have been employed as adjuvants along with tumor cell
sion appears to be a direct result of the presence of or tumor antigen vaccines to boost specific anti-tumor
disease, or follows treatment for it, rather than being an immune responses, or whose primary mechanisms of
antecedent or predisposing condition. However, the pre- action are by activating effector cells directly, such as
cise role that nonspecific and/or specific antitumor interferons and/or interferon inducers, muramyl dipep-
immunity plays in the control of human cancer remains tide and cogeners, or interleukin-2 and other interleu-
controversial. Indeed, there is some evidence that sug- kins (e.g., tumor necrosis factor), will not be covered,
gests that the development of certain immune responses nor will more “traditional” whole organisms from the
may lead to augmented tumor cell growth rather than older literature [e.d., Bacillus Calmette-Guerin (BCG),
tumor regression [178, 287, 406, 407, 582]. Corynebacterium parvum, or mixed bacterial vaccine].
The clinical relevance of the relative state of general
immunocompetence in determining whether or not a
patient can be cured of cancer also remains controver-
sial. A case in point is the dissociation between immu-
Immunosuppression and Cancer
nodeficiency and curability of patients with Hodgkin’s It is clear that preexisting immunodeficiency plays a
disease. Despite the well-recognized immune suppression permissive role in the development of certain cancers,
associated with this malignancy, and the immunosup- such as malignant lymphoma or Kaposi’s sarcoma [393,
pressive therapies used to treat patients (e.g., lymphoid 448]. Patients with primary (e.g., Wiscott-Aldrich syn-
irradiation and steroid-containing combination chemo- drome, at ataxia-telangiectasia) or secondary (e.g.,
therapy), it is one of the most curable of all cancers. Such acquired immune deficiency syndrome, AIDS) immu-
a discrepancy suggests that unique biological properties nodeficiency syndromes in which defects in cell-medi-
of malignant cells, rather than the general immune com- ated immunity predominate and patients receiving
petence of the patient, are the more critical factors in immunosuppressive drugs following organ transplanta-
determining the curability of cancer. tion all exhibit an increased incidence of Burkitt’s and
Even though the precise relationship between the non-Burkitt’s lymphoma, Kaposi’s sarcoma, and a vari-
general state of immune competence and cancer curabil- ety of other tumors not otherwise commonly seen. In
ity has not been established, investigators have adminis- several instances, following the cessation of immuno-
tered a variety of agents to cancer patients including suppressive therapies, cancer regressions have been
biologicals, vitamins, hormones, and drugs, hoping that noted, suggesting that when immunocompetence is
the reversal or prevention of general immune suppres- restored, control of tumor growth may occur. Patients
sion might translate into prolonged disease-free remis- with primary (e.g., Bruton’s-type agammaglobuline-
sions and improved patient survival. This chapter will mia) and secondary (e.g., chronic lymphocytic leuke-
review the multifactorial basis of cancer-associated mia) immunodeficiency syndromes in which defects in
immune suppression and the therapeutic strategies that humoral immunity predominate also exhibit an
have been utilized. The sections on immune suppression increased incidence of malignancies including skin can-
will focus exclusively on the general assessment of cer, primary brain neoplasms, sarcomas, carcinomas,
immunity, and not specific antitumor immunity. The and leukemias [167, 240, 394, 436].
sections on therapy of immune suppression will be lim- Since the malignancies associated with underlying
ited to those biological response modifiers (BRMs), immunodeficiency states are not those common in the

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 101
© Springer Science + Business Media B.V. 2009
102 Therapeutic approaches to cancer-associated immune suppression

general population, it would appear that most adult times impossible) to directly compare the results of in
malignancies do not reflect underlying immunodeficiency. vitro and in vivo immunologic assays from different
It is more likely that a combination of genetic factors, studies. Nevertheless, a number of general conclusions
chronic immune system stimulation (possibly as a result have been reached concerning cancer-associated immune
of recurrent infections), the presence of infectious suppression. No single explanation, or generally agreed-
carcinogenic agents (e.g., viruses), chronic chemical upon concept, has emerged to explain the immunodefi-
carcinogenesis, and other undefined factors leads to the ciency. Rather, there is a complex set of interactions
high incidence of certain cancers in patients with pri- involving a number of different mechanisms. This multi-
mary or secondary immunodeficiency states. There is factorial basis of immunodeficiency is outlined concep-
evidence to indicate that certain carcinogens – for example, tually in simplified form in Fig. 1. The scheme is equally
asbestos – can suppress immune functions such as NK applicable to T−, B−, NK−, or phagocytic effector-cell
activity [430]. However, for the majority of common immune mechanisms.
cancers, the overwhelming evidence suggests that Five major factors each play a role in determining
immunodeficiency arises secondarily as a consequence the immune responsiveness of cancer patients: (a) the
of cancer and the therapies used to treat it; that is, proportions and absolute numbers of circulating, tis-
cancer itself is an immunosuppressive disease. Cancer- sue-derived, or intratumoral effector cells; (b) the
associated immunodeficiency is further influenced intrinsic functional capabilities of the effector cells on
by other factors, including age and genetic background, a per-cell basis; (c) the influence of immunoregulatory
as well as environmental factors, such as nutritional helper and suppressor cells; (d) the influence of local
status, stress, and infections. For example, nutritional systemic-circulating and immunomodulatory soluble
status is frequently impaired in patients with head and factors; and (e) the influence of systemic treatment. In
neck cancer [87], which accounts for many of the immu- Fig. 1, it is shown schematically how the relative con-
nodeficiencies reported in these patient populations such tributions of these five factors modulate immune
as decreased T cell numbers. Thus, it is the balance of responsiveness of cancer patients as assessed with in
many different endogenous and exogenous factors that vitro and in vivo assays.
ultimately contributes to the overall immune deficiency
state of cancer patients.
Effector Cell Numbers and Function
Multifactorial Basis of Immunodeficiency Any basic immune response reflects both the number
in Cancer Patients (or relative proportion) of effector cells present and the
intrinsic functional capability of the effector cells. Thus,
Because of subtle variations in methodologies employed impaired immunity can result purely from a deficiency
by different investigators, it is often difficult (and some- in absolute numbers (or proportions) of effector cells

Local
Immunomodulatory Immune Boosters
Helper Cells Factors (adjuvants)

Tumor-derived
Ontogenesis Lymphokines
Factors In Vivo
Assays

EFFECTOR CELLS
Precurser (Stem) Cells EFFECTOR CELLS IMMUNITY
FUNCTION

In Vivo
Assays

Suppressor Cells Serum Inhibitory Factors Cancer


Therapy
(i.e. RT, Chemo)

Figure 1. Multifunctional basis of immunodeficiency in cancer patients


Robert K. Oldham 103

that otherwise exhibit normal function on a per-cell 136, 172, 203, 487]. NK-cell activity is also frequently
basis, from effector cells that exhibit intrinsic functional depressed in cancer patients [158, 409, 542]. At the local
defects despite normal cell numbers, or from a combi- level, NK activity may be more impaired than other
nation of both decreased cell numbers and impaired lymphocyte functions such as LPRs [17].
function of the individual cells. Thus, many studies now suggest that intrinsic func-
The primary immunodeficiency syndromes, such as tional defects of effector cells contribute, at least in part,
the DiGeorge syndrome, ataxia-telangiectasia, and to the immunodeficiency of cancer.
severe combined immunodeficiency syndrome (SCID),
are examples of immune deficiency resulting from
defective ontogenesis of the immune system. However,
Immunoregulatory Cells
since the development of the immune system occurs The intrinsic functional capabilities of T, B, NK, and
during fetal and neonatal life, defects in ontogeny are phagocytic cells are modulated by interactions with a
not a consideration in the immune deficiency of cancer variety of immunoregulatory cells. The T- and B-cell
patients. Cancer patients do, however, often exhibit functions can be influenced either positively or nega-
lymphocytopenia and decreased T-cell numbers [75, tively, depending upon the relative balance between Tind
101, 320, 352, 376, 384, 416, 481, 547], which contrib- and Tc/s cells, as well as by number and function of
ute to their overall state of immunodeficiency. In con- T-regulatory cells (Treg)[178, 540, 582]. Once again,
trast to primary immunodeficiency states, the effector both absolute numbers (or proportions) and immuno-
cell abnormalities detected in cancer patients arise sec- regulatory function per cell are considerations in deter-
ondarily, probably as a result of the suppressive effects mining the overall influence of immunoregulatory cells
of cancer-derived factors on effector cell production on effector cell function.
and/or survival. Another important immunoregulatory cell is the mono-
When PBMC is used as the source of effector cells in cyte/macrophage. Most normal immune responses require
a lymphocyte or monocyte function assays, it cannot be the presence of monocytes/macrophages as accessory
established whether a depressed functional immune cells for optimal processing of antigen and appropriate
response results from a decreased proportion of effector activation of effector lymphocytes. In many disease
cells within the PBMC mixture, from intrinsic functional states, including cancer, monocytes/macrophages exhibit
defects of the effector cells themselves, from the pres- suppressor-cell activity rather than helper-cell activity
ence of excessive suppressor cell activity, or from a com- [540]. It is likely that such “suppressor” cells may develop
bination of abnormalities in cell proportions, functions, in cancer patients to release factors capable of suppress-
and immunoregulation. There is considerable evidence ing tumor growth as their primary action and thus that the
indicating that cancer patients exhibit depressions in suppression of immune reactivity is an “innocent
mitogen and antigen-induced proliferative assays [75, bystander” effect. The influence of monocytes/mac-
94, 97, 101, 102, 141, 171, 174, 197, 239, 284, 310, 320, rophages is dependent upon cell numbers (or proportions)
333, 352, 376, 408, 420, 429, 481, 547, 574] and in and their state of activation [540].
cytolytic activity [29, 287, 409, 496, 542] using PBMC There is considerable evidence for immune suppres-
as effector cells. In some cancers, for example, head and sion mediated by activated monocytes/macrophages in
neck cancer [399] and Hodgkin’s disease [466], it has cancer patients [5, 28, 30, 50, 80, 193, 194, 195, 250, 257,
been possible using purified T cells to document intrinsic 330, 461, 523, 532, 540]. For example, monocyte-mediated
functional T-cell defects. Studies in cancer patients have suppression of lymphocyte-proliferative functions have
shown an impaired ability of T cells to produce the been demonstrated in Hodgkin’s disease [154, 175, 189,
lymphokine IL-2 [190, 321, 353] following activation by 336, 454, 535] as well as in patients with lung cancer [5,
mitogens; but that spontaneous IL-2 production (reflecting 93, 250], breast cancer [194, 253], malignant melanoma
possible stimulation due to circulating tumor antigens) is [363], colorectal cancer [28, 194, 524], head and neck
increased [235]. However, in Hodgkin’s disease, although cancer [30, 50, 550], bladder cancer [195], and a variety
IL-2 production and/or IL-2R expression of PBMC has of other malignancies [86, 540]. Monocyte-mediated
been reported to be low [48, 359, 490, 580], the abnor- suppression of cytolytic effector-cell activity has also
mality in T-cell lymphoproliferation does not appear to been demonstrated in cancer patients [14, 18, 123, 151,
be related to defects in the IL-2 system [48]. 228, 229, 540], including NK-cell activity [246], and
Peripheral blood monocytes and polymorphonuclear autologous T-cell-mediated anti-tumor cytotoxic
leukocytes isolated from patients with a variety of cancers responses [151, 461]. In comparative studies, it has been
also exhibit depressed functional activity [31, 73, 103, demonstrated that such suppressor-cell activity is of
104 Therapeutic approaches to cancer-associated immune suppression

greater magnitude at the local level (i.e., intratumoral, ety of disease states including cancer, there may be a
effecting TIL cells and in draining lymph nodes) than in deficiency in the production of cytokines by cells that
the systemic circulation [14, 18, 123, 228, 229]. Of normally produce them [159, 190, 206, 279, 353, 360,
interest is the finding that monocyte/macrophages acti- 548, 572, 579].
vated to kill cancer cells can suppress T-cell cytotoxicity The release in cancer patients of a variety of immuno-
toward the very same cells [532]. modulatory factors produced by monocytes/mac-
The contribution of suppressor lymphocytes to the rophages and/or the tumor cells themselves has been
generation of cancer-associated immunodeficiency is described. Immunosuppressive factors, for example, the
less certain and still somewhat controversial. Several E series of prostaglandins, are released in excessive
studies that identified helper and suppressor T-cells on quantity from both monocytes/macrophages [5, 28, 30,
the basis of the presence of surface Fc receptors for IgM 50, 80, 154, 189, 195, 363, 386, 453, 523] and tumor
(Tμ cells) or IgG (Tγ cells), respectively, have suggested cells [72, 139, 140, 221, 261, 358, 523], which could
that the Tind/Tc/s ratio is depressed in cancer patients [204, suppress cytolytic activities [358] and proliferative
267, 531]. However, more recent studies using monoclo- functions [123] of TIL cells as well as of circulating
nal antibodies to detect surface antigens indicate that the lymphocytes. However, prostaglandins alone do not
relative proportions of Tind and Tc/s cells are preserved in mediate the complete suppressor cell activity of mono-
most untreated cancer patients, unless they have cytes [28, 30, 154, 453, 523]. Monocyte-derived toxic
extremely widespread disease [130, 200, 266, 310, 462]. oxygen metabolites, for example, hydrogen peroxide
Few reports have focused on functional aspects of sup- [336], and other as yet undefined mediators also appear
pressor lymphocyte activity in cancer patients. Definitive to play a role. Increased monocyte production of prosta-
conclusions are lacking, although such activity has been glandins has been shown to directly impair lymphocyte
reported in patients with Hodgkin’s disease [217, 480] proliferative and cytolytic functions [139, 140] as well
and various solid tumors [151, 236, 237, 283, 537]. as the phagocytic function of monocytes themselves
Inducible suppressor T-cell activity has generally [104, [150]. A variety of other factors have been identified
183, 236, 464] but not always [521] been found to be that are shed by tumor cells and modulate both local as
depressed in cancer patients. well as systemic immune responses. These include
Over the last 10 years, studies of immunoregulation tumor-associated glycoproteins [536] and lipids [196].
have focused increasingly on a subset of T-cells called For example, various melanoma-associated ganglio-
T-regulatory cells (Treg). Tregs have been well defined as a sides have been shown to both up-regulate and down-
distinct population of CD4 T-lymphocytes, which con- regulate lymphocyte responses to IL-2 [230]. Thus, it
stitutively express CD-25 and are further characterized has been suggested that the unique tumor-antigen-asso-
by the molecules CTLA-4, FOXP-3, and a TNF receptor. ciated phenotype of each individual tumor (based on the
Treg cells also produce immunosuppressive cytokines, proportion of various immune-suppressing and aug-
such as IL-10 and TGF beta [307, 444, 467]. Treg cells menting factors released by it) determines whether the
with the phenotype CD-25+, FOXP-3+, CD-127− have individual tumor will exhibit immune stimulation or
been found in tumor specimens and are suspected as suppression. In this regard, it has been demonstrated
cells which suppress the activation of CD-4− and CD-8+ that whereas primary melanomas stimulate autologous
tumor-infiltrating lymphocytes active in tumor destruction. lymphocyte responses, metastatic melanomas suppress
The presence of these cells correlate with poor clinical immune responsiveness [510].
outcome in cancer patients [218, 450, 467]. Clinical It is also apparent that a number of different substances
studies are now underway, using antibodies to CTLA-4, with immunosuppressive properties can be detected in
to attempt to lessen the immunosuppressive effects of the blood of patients with cancer [469]. Many investiga-
Treg and, thereby, enhance the effectiveness of the CD-4 tors have shown that serum from cancer patients is
and CD-8 T-cells and immune effectors [573]. immunosuppressive in that it suppresses mitogen and
antigen responsiveness of lymphocytes from normal
donors [11, 55, 98, 164, 182, 188, 226, 416, 421, 475,
Immunomodulatory Factors 482, 502]. Cancer serum inhibitory factors include
It is now well established that, in normal immune reac- acute-phase reactants, such as α1-acid glycoprotein [39,
tions, the influence of immunoregulatory cells is medi- 509], α-globulins [234], C-reactive protein [383], and
ated at least in part by the local release of cytokines such immune complexes [35, 259]. A serum factor in young
as IL-1 by monocytes, and IL-2, 6 and 10, as well as cancer patients has been reported to inhibit serum thy-
γ-interferon by helper T cells (see Chapter 8). In a vari- mic-hormone bioactivity [121]. Circulating immune
Robert K. Oldham 105

complexes have been shown to produce immunosup- abnormalities of immune competence, more advanced
pressive effects by a variety of mechanisms including disease, particularly after treatment, leads to abnormali-
(a) blocking of B-cell differentiation and antibody pro- ties in all measurable immune parameters [131].
duction [435, 516]; (b) stimulating the production of
anti-idiotype antibody which then interferes with the
immune response to the original antigen [435, 516]; (c) Solid Tumors
inducing suppressor T-cells [105]; (d) reducing IL-2
levels [418]; and (e) blocking Fc receptors on effector
Delayed-Type Hypersensitivity (DTH)
cells [168]. Theoretically, the quantitative removal of Many reports have confirmed that as the extent of dis-
tumor antigens, antitumor antibodies, and/or immune ease increases, the incidence of positive DTHS reactions
complexes could lead to a specific or nonspecific stimu- decreases [457]. For example, in one large study of 234
lation of the immune system, leading to an increase in patients with various types of cancer, patients without
general immune competence as well as in specific anti- metastases exhibited positive reactions to DNCB (83%)
tumor immune responsiveness. This has formed the more often than those with regional metastases (67%) or
basis for therapeutic trials with extracorporeal treatment with distant metastases (41%) [401]. Similar findings
of cancer with immobilized staphylococcal protein A using both DNCB as well as recall antigens have been
[335, 362] and for attempts at plasma ultrafiltration. The noted in a wide variety of solid tumor-bearing patients,
mechanism of antitumor activity of such approaches has including those with breast cancer [2, 34, 36, 75, 76, 146,
still not been defined. However, plasmapheresis has 296, 496], gastrointestinal cancer [76], head and neck
been associated with increased LPRs and antitumor cancer [495], lung cancer [8, 227, 389, 350, 561], renal
immune responses [468, 470], probably due to removal cancer [86, 355], gynecologic cancer [561], urologic
of immunosuppressive serum factors. cancer [39, 85, 456], malignant melanoma [145, 408],
In conclusion, it is clear that the state of immunocom- sarcomas [145], and primary brain cancers [84, 317].
petence of an individual cancer patient is dependent The incidence of positive DTH responses varies
upon a number of complex interactions among effector according to type of cancer. For example, patients with
cells, immunoregulatory cells, and local and systemic localized head and neck cancer had a much lower reac-
immunomodulatory factors. A simplified explanation tivity (42%) than patients with sarcomas (73%) and
for the immunosuppression of cancer is that products lung cancer (80%) [76]. This result has been explained,
released from the malignant cells themselves lead to (a) at least in part, by the alcohol intake and general state of
activation of suppressor cells; (b) impaired effector cell malnutrition associated with head and neck cancer
production and survival; and (c) direct inhibition of patients [54, 311]. In another study, it was found that the
effector cell function. In assessing the state of immune correlation between DNCB reactivity and clinical status
competence with in vitro and in vivo assays, the under- was more pronounced in patients with squamous-cell
lying basis of immunodeficiency may or may not be carcinomas of the head and neck than in those with
identified. sarcomas or melanomas [562]. These findings suggest
that although DNCB reactivity generally correlates with
Immunosuppression and Tumor Cell extent of disease, the relationships are modified depend-
Burden ing upon the particular type of cancer.
It has been easy to demonstrate the marked degree of
In newly diagnosed, untreated cancer patients, the degree impairment in DTH reactions found in patients with
of immunodeficiency generally parallels the extent of dis- metastatic cancer; however, it has been much more dif-
ease [564]. The most reasonable explanation for such an ficult to discern relative differences in reactivity of
association is that the release of tumor-derived immuno- patients with similar stages of disease whose tumors dif-
suppressive factors relates directly to the tumor cell bur- fer in size, regional lymph-node involvement, or local
den. Immune parameters of which impairment correlates invasiveness. Therefore, DTH responses are insensitive
with extent of disease include DTH to recall antigens, immunological tests [101].
blood effector cell levels (e.g., T-lymphocyte levels),
lymphocyte functions, including proliferative responses
to mitogens and antigens, cytotoxic activity, phagocytic Lymphoproliferative Responses (LPR)
cell activity, serum immunoglobulin levels, and primary An exhaustive literature has accumulated in which vari-
antibody responses to a variety of immunizing antigens. ous in vitro lymphoproliferative assays were employed
Whereas quite early cancer is associated with only subtle to assess the state of immune competence of cancer
106 Therapeutic approaches to cancer-associated immune suppression

patients. In general, PBMC were the responder cells, advanced malignancies [578]. Other studies found an
and T-cell mitogens (e.g., PHA, Con A) were used to inverse correlation between total lymphocyte count and
induce blastogenic transformation. Many reports have stage of disease in breast [416] and lung cancer [547].
documented an inverse correlation between PHA The observation that human T lymphocytes could be
response and stage of disease for patients with a variety easily identified by a binding reaction to sheep red blood
of solid tumors. For example, among 179 patients with cells to form E rosettes (E-RFC), coupled with the
gastric carcinoma, 87% with stage I-II, 49% with stage emerging recognition of the importance of T cells in
II, and only 24% with stage IV disease could exhibit immune responsiveness, led to numerous studies on
morphological evidence for blastogenic transformation cancer patients. Using E-rosette techniques, the percent-
of PBMC following exposure to PHA [376]. A study of age of circulating T cells has been determined for
154 patients with carcinoma of the lung revealed that patients with a wide variety of solid tumors and corre-
lymphoproliferative responses were significantly lated with clinical stage of disease [320, 481, 547, 567].
decreased in patients with stage III disease, but not in Other studies have not found such a correlation [75,
those with stages I and II [547]; other studies have 352, 376]. In general, however, as with the in vitro
reported similar results [102, 141, 197, 420]. Depressed assays of T-cell functions, peripheral blood T-cell num-
LPR to alloantigens in mixed leukocyte culture (MLR) bers tend to decrease in association with more advanced
were observed in 46% of patients with small (TINOMO) stages of cancer.
stage I lung cancers [97]. Depressed mitogenic respon- Although assessments of B cells, monocytes, and Tind
siveness has been correlated with advancing stage of and Tc/s ratios have been performed less frequently in
disease in breast cancer [49, 239, 320, 333, 464] and, in cancer patients, in general, B-cell numbers have paral-
one study, lymphocyte responses to PHA were impaired leled T-cell numbers [266, 310, 352, 440], whereas
in earlier stages of disease than was DTH reactivity absolute monocyte counts tend to increase with advanc-
[496]. Similar inverse correlations between stage of dis- ing disease [310]. T-cell subset abnormalities have been
ease and LPRs have been noted in colorectal cancer found in some patients with malignant melanoma [266],
[171], malignant melanoma [174, 284, 481], and head head and neck cancer [130, 204], and lung cancer [13,
and neck cancer [101]. 527, 565]. T-cell subset abnormalities were more pro-
Although many reports have documented that LPR nounced in lung lavage cells than in PBMC from patients
generally decrease with advancing stage in solid tumor with lung cancer [160], suggesting that such perturba-
patients, this finding has not been universal. For exam- tions occur first at a local level before systemic abnor-
ple, lack of correlation between blastogenic responses malities become detectable. In general, abnormalities of
and disease stage has been reported in patients with the Tind and T c/s ratios are found in patients with advanced
breast cancer [71, 310, 429], malignant melanoma [94, or progressive disease. The ratio between Treg and CO8+
174, 408], and colorectal cancer [352]. These reported cytotoxic T cells correlates with immunosuppression
inconsistencies have probably resulted from differences and clinical outcome [218, 450].
in techniques and quality control procedures for assays
with a great deal of inherent variability. Nevertheless,
the same general conclusion holds for in vitro LPR and
Cytolytic Functions
DTH skin testing, namely, that there tends to be an The measurement of nonspecific lymphocyte-mediated
inverse correlation between immune reactivity and cytotoxicity has been employed with increasing fre-
tumor burden and/or stage of disease. quency as part of the general assessment of immune
competence in patients with solid cancers. In general,
cytotoxic activity (most often NK activity) was
Immune Cell Quantitation depressed in cancer patients, particularly those with
There are many monoclonal antibodies available to metastatic disease, but clear correlations have not been
quantitate blood, organ, and intratumoral levels of vari- identified between impaired function and clinical stage
ous immune effector cells; however, most of the early of disease [262, 307, 505, 542]. Studies in patients with
observations concerning lymphocyte quantitation and small-cell lung cancer and stage I and II malignant mel-
cancer were made using more primitive assay methods. anoma have found an inverse correlation between NK
The quantitation of lymphocytes in the peripheral blood activity and amount of clinically detected tumor [158].
of cancer patients has been studied as a measure of Similarly, NK-cell activity was more likely to be
immunocompetence since the early 1920s, when lym- depressed in stage II breast cancer than in stage I dis-
phocytopenia was found to be common in patients with ease. A large study of 247 cancer patients showed that
Robert K. Oldham 107

circulating NK cell numbers, assessed by monoclonal Phagocytic Cell Function


antibody methods, were significantly reduced in patients
with colon, lung, and breast cancer, but not in those with Other impairments of immune responsiveness in solid-
melanoma or sarcomas [29]. Thus, a depression of NK tumor patients have been documented. For example, the
cell numbers could explain the depressed NK function inflammatory response of patients with advanced cancer
reported in some, but not all, cancer patients [3]. is associated with a reduced capacity to mobilize mono-
The relationship between NK activity and other mea- cytes [31, 137, 172, 546]. Reticuloendothelial function
sures of immunocompetence has been explored. A lack in patients with breast and colorectal cancer is depressed
of association was found between DTH reactivity to [138]. Depressed monocyte chemotaxis has been corre-
PPD and NK activity [409]. A comparison between TIL lated with disease stage for a variety of tumor types [73,
activity and PBMC revealed that TIL expressed dimin- 203, 487]. Detailed evaluations of monocyte function in
ished NK activity compared with PBMC [225]. In one solid-tumor patients reveal abnormalities in patients
study, although PBMC exhibited depressed NK activ- with malignant melanoma, breast cancer, colorectal
ity, proliferative responses to PHA and in MLR were cancer, and head and neck cancer, but no consistent cor-
maintained [409]. This suggested that cytolytic and relations have been identified between tumor type,
proliferative effector-cell mechanisms represent dis- monocyte defect, and clinical stage of disease.
tinct functional entities.
Correlations Among Immune Cell
Numbers and Function
Antibody Formation A number of studies have attempted to find correlations
A variety of humoral immune abnormalities have been between in vitro and in vivo abnormalities of immune
found to occur in association with solid tumors. A large cell numbers and function. For example, both in vitro
study of 984 patients with nonhematopoietic cancers PHA responses and in vivo DNCB reactivity were normal
found no general trends, but did find increases in serum in patients with early bladder cancer, but significantly
IgG and IgA in males with skin and lung tumors, impaired in patients with advanced disease. Similar cor-
increases in IgM in males with sarcomas and females relations have been seen in patients with lung cancer
with melanoma, decreases in IgM in patients with ovar- [424] and breast cancer. In contrast, in a study of 48
ian cancer, and increases in IgA in patients with oral, patients with lung cancer, LPRs were more often sup-
gastrointestinal, and uterine cancers [238]. Increased pressed than were skin test responses [8], and no corre-
IgA has also been demonstrated in head and neck cancer lations between DNCB reactivity and in vitro immune
[319, 552] and prostate cancer [6], but to date, no cor- functions were found in patients with head and neck
relations have been observed between serum immuno- cancer. It has also not been possible to identify any con-
globulin level and clinical stage of disease [125]. sistent association between alterations in effector-cell
The serologic response to several different B-cell numbers and function [103, 251].
immunogens has also been studied in solid-tumor
patients. Patients with nonlymphomatous malignancies
were found to exhibit decreased specific antibody
Hematopoietic Malignancies
responses to Salmonella extract [297]. Patients with Reed’s report in 1902 that patients with Hodgkin’s dis-
stage III squamous-cell lung cancer exhibited deficits in ease (HD) failed to react to tuberculin skin tests even if
IgG and IgA production following immunization with they were known to have had active tuberculosis [410]
Helix pomatia hemocyanin, a T-cell-dependent antigen led to a vast number of immunologic studies for this
[248]. A significant impairment in the ability of certain malignancy. Many of the immune abnormalities first
patients with solid tumors to produce both IgM and IgG reported – for example, alterations of absolute lympho-
antibody in response to primary challenge with mono- cyte counts, impairments of T-cell LPR, and the presence
meric S. Adelaide flagellin has been reported [297]. Both of suppressor monocytes/macrophages – were subse-
complete and incomplete primary antibody responses to quently also found in patients with a variety of other can-
heat-killed Brucella were reduced in patients with breast cers. Even though the primary immune defects described
and lung cancer [544]. However, precise kinetic data for in untreated patients with HD involve T-cell immunity,
humoral antibody production in most patients with solid other defects have also been documented [58, 264, 433]
tumors are lacking, as are correlations between humoral including impaired in vitro B-cell production of antibod-
immunity and extent of disease [143]. ies, phagocytic cell function, and NK-cell activity [161].
108 Therapeutic approaches to cancer-associated immune suppression

The clinical relevance of the T-cell immunodeficiency disease are the most important prognostic indicators for
in patients with HD has been recognized for many years any particular cancer, it is logical that the general state of
[419]. Patients with HD have an increased susceptibility immunocompetence and prognosis should be closely
to infections associated with defective T-cell immunity, related. The potential importance for correlating immu-
including Pneumocystis carinii pneumonia and viral nocompetence and prognosis at the time of diagnosis or
infections such as herpes simplex, herpes zoster, and at the onset of therapy is obvious [155]. Assessment of
cytomegalovirus [22, 433]. In most of these studies, immune status could, in theory, help to select patients
however, it has been difficult to assess the role of the with a poor prognosis who might require more aggressive
treatment for HD (radiation therapy, chemotherapy) in therapy than usual, or adjuvant therapy even if all disease
exaggerating the T-cell immune deficiencies, since many appeared to be surgically excised. In a now classic study,
of the infectious complications occurred only during or it was observed that patients who could be sensitized to
after the completion of therapy. DNCB and freed of disease by surgery had a good prog-
A number of studies have now attempted to correlate nosis, whereas those who were apparently freed of dis-
defects in T-cell functions with depression in T-cell ease by surgery but could not react to DNCB had a poor
numbers in untreated patients with HD [71, 224, 460]. prognosis and relapsed within 6 months to 1 year [562].
In general, no such relationships could be identified, The DNCB responses of patients in the poor prognosis
although lymphocytopenia patients were more likely to group were identical to those of patients who were found
exhibit impaired LPR. There is an easily identifiable to be inoperable at surgery. The two groups of patients
correlation between defects in immune responsiveness were otherwise comparable with regard to type of tumor,
and clinical stage of disease, in that most immune abnor- extent of disease, and all other usual clinical prognostic
malities are more readily apparent in patients with factors. This report formed the basis for a number of sub-
advanced stages of disease than in those with localized sequent studies attempting to correlate immunodeficiency
disease [460]. with prognosis for previously untreated patients with
Immune deficiencies have also been shown to corre- solid tumors and hematopoietic malignancies.
late with lymphoma subtype and extent of disease in the
non-Hodgkin’s lymphomas. These include impaired in Solid Tumors
vitro LPR to mitogens [12, 312] and impaired in vivo
DTH responses [12]. In general, patients with high- Since 1970, there have been numerous attempts to cor-
grade lymphomas exhibit more profound abnormalities relate impaired DTH skin reactions with prognosis.
than those with more favorable histologies [258]. Associations between impaired DNCB reactivity and
It has been very difficult to relate the extent of immu- disease recurrence have been observed in lung cancer
nodeficiency to tumor burden in patients with cancers [299], head and neck cancer [318, 441], gastrointestinal
involving the bone marrow (e.g., leukemia, multiple cancer [77], and breast cancer [146]. A number of reports
myeloma) since, with the exception of multiple myeloma, have suggested that patients with or without metastases
staging systems based on tumor cell burdens do not exist. but without reactivity to DNCB have a poor prognosis
However, a wide variety of in vitro as well as in vivo [74, 85, 147, 265, 298, 318, 402]. Lack of reactivity to
abnormalities have been documented in patients with common skin test recall antigens has also been corre-
lymphoid and nonlymphoid leukemias [199, 209]. lated with poor prognosis in breast [160, 349], lung [15,
Furthermore, a spectrum of intrinsic functional abnor- 244, 256, 299], gastrointestinal [382], urologic [334],
malities has been identified in B cells and also purified and head and neck cancer [494], although this has not
T-cell populations of patients with chronic lymphocytic been a universal finding [54, 296, 350, 361, 397, 406,
leukemia and multiple myeloma [236, 247]. Both of these 452]. Several studies have subcharacterized patients
diseases are often associated with profound depressions with identical clinical stages of disease on the basis of
of normal serum immunoglobulin levels and impaired impaired skin test reactivity to define further the rela-
ability to mount primary humoral immune responses. tionship between anergy and prognosis. Such correla-
tions have been found, for example, in patients with
stage I [19] or stage III and IV [295] malignant mela-
Prognostic Implications of noma, and in limited-disease small-cell lung cancer
Immunosuppression [256]. However, other studies of accurately staged
patients with breast cancer, although DNCB-negative
Since immunodeficiency generally correlates with the patients had a worse overall survival, when survival dis-
stage and extent of disease, and since stage and extent of tributions of DNCB-positive and –negative patients
Robert K. Oldham 109

with either primary operable or advanced breast cancer both cell-mediated and humoral immune functions have
were compared separately, significant differences were been found to correlate with prognosis [59, 120]. While
not seen. Thus, studies to date have yielded conflicting DTH was only moderately decreased, diminution in anti-
conclusions concerning the correlation between impaired body responses and PHA responses correlated with the
in vivo immunity and prognosis. duration of disease, status of therapy, degree of lympho-
Associations between poor prognosis and impaired cytosis, and immunoglobulin levels [120]. A correlation
in vitro LPR have also been reported [97, 392]. Other studies has been reported between depressed absolute circulating
have not produced such correlations [94, 174, 429, 446]. NK-cell levels and poor prognosis in patients with large-
Studies correlating absolute lymphocyte counts or cell lymphoma [38]. Patients with high-grade lymphomas
absolute T-cell levels with prognosis have provided who have high levels of serum IL-2 receptor have been
conflicting results [376, 479]. Furthermore, the use of shown to have more advanced disease [410] and a worse
multiparameter immunological assessments has not prognosis [543]. However, in this instance, the IL-2
generally improved the ability to assess prognosis for receptor is synthesized by the tumor cells so that serum
previously untreated solid tumor patients [128, 174, levels parallel tumor cell bulk. Nevertheless, the predic-
305, 320, 352]. However, some investigators have tive value of soluble IL-2 receptor was superior to that of
developed predictive indices based on multiple immu- other markers that reflected tumor cell bulk such as lactic
nological parameters. For example, in one study, an dehydrogenase level (LDH) or clinical stage [410].
integrated score of immunocompetence based on vari-
ous in vivo and in vitro assays showed that the recur-
rence of breast cancer was significantly higher in
Perioperative Immunosuppression
suboptimal (61%) as opposed to optimal (28%) respond- Although it is perhaps not universally recognized, the
ers [3]. Similarly, an immunological staging system operative procedure and its associated general anesthesia
based on absolute lymphocyte counts and serum immu- result in a variety of transient immunological defects that
noglobulin level was found capable of predicting the can persist for several weeks after surgery [559]. During
outcome off stage III head and neck cancer patients in operative procedures under general anesthesia for a vari-
86% of cases [268]. However, in a report using logistic ety of benign and malignant conditions, patients exhibit
regression methodology, it was demonstrated that only inhibition of skin reactivity to DNCB [511] and DTH
the level of complement binding activity, which may recall antigens [527], suppression of circulating T-cell
reflect levels of circulating immune complexes, corre- levels [273, 528, 529], diminished LPR to PHA and
lated with the likelihood of responding to induction che- other mitogens [260, 437, 483], and depressed NK activ-
motherapy [452]. No associations could be determined ity [456, 513]. Patients with cancer appear more likely to
between response to chemotherapy and abnormalities of experience these periods of postsurgical immunosup-
a variety of other immune parameters, including lym- pression than do patients who undergo surgery for benign
phoproliferative responses, NK activity, lymphocyte conditions. Among the conditions associated with the
subset numbers and percentages, and serum immuno- greatest degree of postoperative immunosuppression are
globulin levels. intraabdominal and intrathoracic procedures, blood trans-
fusions, and longer operating times [439, 471].
Hematopoietic Malignancies
Among the hematopoietic malignancies, most attention
Perioperative Blood Transfusion
has focused on correlating defects in T-cell immunity Several studies have suggested that perioperative blood
with prognosis in HD [488, 577]. More recent reports transfusion, possibly by inducing a greater degree of
suggest that there is a correlation between extent of immunosuppression, results in an adverse effect on
immune dysfunction and prognosis [57, 153, 533]. prognosis for postoperative patients with colorectal cancer
Relationships between immunocompetence and prog- [222], breast cancer [514], non-small-cell lung cancer
nosis have also been reported for a variety of other [512], prostate cancer [205] and soft-tissue sarcoma [434].
hematopoietic tumors. For example, in acute leukemia, However, three recent studies in breast cancer [541],
patients with showed DTH skin reactivity and, to a lesser colorectal cancer [550], and lung cancer [271] have not
degree, in vitro blastogenic responses to mitogens exhib- confirmed initial reports. Thus, it has not been conclu-
ited the best overall survival, outranking such prognostic sively proven that perioperative blood transfusions worsen
variables as age, type of leukemia, and absolute blast cell the prognosis of patients with cancer. It is possible that
count [177, 303, 304]. In chronic lymphocytic leukemia, the requirement for transfusion is a marker for other risk
110 Therapeutic approaches to cancer-associated immune suppression

factors such as advanced stage of disease, need for more following radiation therapy. Mediastinal irradiation for
extensive operation, or greater blood loss during surgery, treatment of localized lung cancer has been associated
thus accounting for the worse prognosis following surgery. with a decrease in proliferative responses of purified
The precise mechanisms for postoperative immune peripheral blood T cells, suggesting that radiation
suppression have not been fully defined. Among the directly impairs their functional capabilities [462].
considerations are the immunosuppressive effects of Several studies have assessed the effects of radiotherapy
surgical stress [124], or of the anesthetic agents them- on T-cell subset proportions. Although there was a drop
selves [511], a relative decrease in circulating T-cell lev- in the absolute numbers of both helper and cytotoxic/
els compared with other cell types [180, 181], and/or the suppressor T-cells following mediastinal irradiation, the
generation of suppressor cells [360]. In nonsurgical drop in cytotoxic/suppressor T-cells was greater, so that
patients, chronic blood transfusion is associated with treatment resulted in a significant increase in the Tind/Tc/s
depressed Tind/Tc/s ratios and impaired NK activity. ratio [462]. These observations are consistent with
Theoretically, the associations between perioperative in vitro functional data indicating that suppressor T-cells
blood transfusion and earlier cancer recurrence, and are more radiation-sensitive than helper T cells [184,
between prolongation of renal allograft survival and 398]. In contrast, radiotherapy for patients with breast
transfusion, may be attributed to a transfusion-induced cancer [452, 397], head and neck cancer [249], and
immune suppression resulting from a graft-versus-host Hodgkin’s disease [404, 432] has been associated with a
reaction mediated by transfused T-lymphocytes. Although relative increase in the proportion of T cells bearing sur-
the precise mechanisms to explain perioperative immune face antigens and/or Fc receptors (IgG) of cytotoxic/
suppression have not yet been defined, this well-docu- suppressor cells, leading to a decrease in the Tind/Tc/s
mented abnormality has nevertheless formed the basis ratio. Indeed, in postradiotherapy patients with breast
for a number of therapeutic trials in which various puta- cancer [397] and Hodgkin’s disease [185], helper T-cell
tive immunorestorative agents have been administered levels remained low for years after irradiation.
as surgical adjuvants. Several studies have assessed the influence of thera-
peutic irradiation on suppressor cell function. They
Radiation Therapy-induced indicate that radiation therapy can activate suppressor
Immunosuppression monocytes [63, 68, 313] as well as increase the sensitivity
of lymphocytes to the suppressive effects of prostaglan-
The immunological effects of external-beam radiation dins [313].
therapy have been delineated in detail. Radiation ther- External-beam radiotherapy, such as pelvic [375, 413,
apy to a variety of portals, including mediastinal [146, 498, 499] or chest wall [64, 513, 498, 499, 522, 556]
273, 306, 371, 498], pelvic [65, 306, 375, 413, 498], radiotherapy, has also been associated with a decrease in
head and neck [306, 371, 483, 494, 551], lymphoid B-cell and NK-cell numbers and functions in many, but
[165, 185, 432], and breast [33, 191, 306, 413, 522, 556] not all, reports. Mediastinal irradiation more severely
portals, results in similar acute and chronic changes depressed NK activity than treatment to nonmediastinal
characterized by a generalized lymphocytopenia involv- sites [332], possibly reflecting the relatively large volume
ing primarily a depletion of circulating T cells as well as of blood and/or lymph node volume included within the
marked depressions in various T-cell functions such as mediastinal radiation portal, leading to damage of NK
in vitro LPRs to mitogens and antigens and in vivo cells, or their more radiation-sensitive precursor cells.
DTHS reactions [265, 484]. However, within 3–4 months following the completion
The depression of T-cell numbers and function occurs of radiotherapy, NK-cell activity returned to pretreat-
progressively with radiotherapy. The magnitude of ment levels, which then persisted for at least 2 years
immune depression reflects both the dose of radiation [67]. There was a partial recovery in B-cell function,
received and the volume of tissue, blood, lymph, or bone which remained below pretreatment levels for 12–18
marrow included within the radiation portals. This acute months after completion of therapy [438, 500]. Reports
immune suppression is short-lived, and substantial recov- concerning the effects of radiotherapy on absolute blood
ery is apparent within 3 weeks of cessation of therapy. monocyte levels have been conflicting [522, 549]. Other
However, most patients show a modest chronic depression immunological parameters have not been significantly
in both numbers and functions of T cells, which may last affected. For example, radiotherapy does not produce
for years following the completion of radiotherapy [252]. changes in serum immunoglobulin levels or neutrophil
A number of other observations have been made con- function. However, the ability of monocytes to differen-
cerning the mechanism of functional immune impairment tiate into macrophages was impaired in breast cancer
Robert K. Oldham 111

patients treated by radiotherapy [515], and absolute temporary enhancement of immune responsiveness due
monocyte levels increased after treatment [522], whereas to preferential inhibitory effects on suppressor cells.
pelvic radiotherapy produced a decrease in both mono- Traditionally, it has been felt that intermittent chemo-
cyte numbers and functions in patients with colorectal therapy schedules of administration using single or mul-
cancer [549]. tiple drugs tend to be associated with relatively little
In the earlier literature it was not possible to correlate effect on DTH responses and with transient depressions
the extent of radiotherapy-induced immunosuppression of lymphocyte numbers, in vitro LPR, antibody responses,
with relapse or survival [133, 462, 555]. In general, how- and inflammatory responses, with at least some recovery
ever, patients who exhibited skin reactivity to DNCB several weeks after discontinuation of treatment [210,
prior to radiotherapy had a better prognosis than those 417]. In contrast, continuous therapy, if given in adequate
who did not [79]. In two more recent studies of patients doses, has been shown to lead to a progressive decline in
who received primary radiation therapy for breast cancer all phases of immune reactivity [166, 207, 210, 331, 417],
[499, 555] and/or cervical cancer [499], those who which is reversible after the cessation of administration
exhibited the greatest postradiotherapy depressions of of drug(s). In several more recent studies, however, it has
LPRs to mitogens or antigens had the shortest survival been demonstrated that intermittent combination chemo-
[555]. Several studies on postradiotherapy patients with therapy regimens lead to cumulative depressions of lym-
lung cancer have serially assessed the immune status of phocyte numbers and functions over months to years
those who responded transiently to treatment but relapsed [315, 501], which may or may not be reversible. These
at a later date [133]. Lung cancer patients who responded changes include depressions of absolute T- and B-cell
clinically to radiotherapy showed some improvement of levels, depression in the Tind/Tc/s ratio, and depressions of
absolute T-cell levels in the months following cessation LPR to mitogens and antigens.
of treatment, whereas those who progressed did not show Another phenomenon that has been described is the
such partial recovery [133]. In addition, for postradio- recovery or transient, rebound overshoot of immune
therapy patients with locally advanced non-small-cell responsiveness after a single cycle of chemotherapy
lung cancer, there was a gradual and progressive decrease [81, 108, 201]. Rebound is found in patients who are
in T-cell and helper T-cell percentages, and in the Tind/Tc/s responding clinically to treatment. Patients whose
ratio that preceeded relapse [462]. immune functions remain suppressed either have not
responded clinically to treatment or are at high risk for
Chemotherapy-induced relapse compared with those who exhibit recovery to
Immunosuppression levels above those pretherapy. It has been suggested that
the rebound overshoot in immune reactivity following
It has been much more difficult to assess the effects of chemotherapy may be due to a reduction in monocyte
cancer chemotherapy than of radiation therapy on suppressor cell function [81]. The effect can result from
immune responsiveness. In part, this is due to the fact a transient, chemotherapy-induced decrease in relative
that a wide variety of different cancer chemotherapy numbers of monocytes/macrophages, from a reduction
drugs are available for clinical use, many of which can in suppressor cell function on a per-cell basis, or from a
be administered by a variety of routes and doses, either combination of both. Recovery of suppressor monocyte/
alone or in combination with other drugs. The majority macrophage function is responsible for the eventual
of anticancer drugs produce a dose-dependent pancy- decline in immune function following drug-induced
topenia, which itself is immunosuppressive because of rebound overshoot.
the associated granulocytopenia. In addition, most, if
not all, cancer chemotherapy drugs exhibit immunosup-
pressive properties as assessed with conventional in vivo Antimetabolites
and in vitro assays. This includes the alkylating agents, All of the antimetabolites commonly used for treating
antimetabolites, and antitumor antibiotics. cancer patients, including thiopurines such as 6-mercap-
Cancer chemotherapy may lead to profound depres- topurine (6-MP) and 6-thioguanine (6-TG), antifolates
sions of cellular as well as humoral immunity [202, 207, such as MTX, fluorinated pyrimidine analogs such as
213]. Newly acquired DTH and primary humoral immune 5-fluorouracil (5-FU), cytosine arabinoside (Ara-C), and
responses appear to be more sensitive to drug-induced DTIC (5-[3-3-dimethyl-1-triazeno]-imidazole-4-carbox-
immunosuppression than are secondary responses. Less amide) are immunosuppressive in humans. 6-MP has
often, the administration of selective drugs, for example, been one of the most extensively studied agents [207,
cyclophosphamide (CTX), at low doses can lead to a 213, 300]. Azathioprine, a nitroimidazole derivative of
112 Therapeutic approaches to cancer-associated immune suppression

6-MP, is a potent immunosuppressive drug, and has found erentially decreased circulating B-cell numbers. Doses
more use in clinical transplantation than as a cancer che- from 200 to 600 mg/m2 selectively depleted cytotoxic/
motherapy drug [343]. 6-MP, 6-TG, MTX, and 5-FU suppressor T cells, leading to a transient increase in the
have all been shown to preferentially depress primary Tind/Tc/s ratio, whereas higher doses affected all T-cell
antibody responses and the development of new DTH subsets equally. Intravenous administration of conven-
reactivity [213, 343, 345, 346]. However, 5-FU treatment tional doses of CTX for 5 days has been shown to
restored DTH to recall antigens such as PPD, mumps, depress established DTH by 7–10 days after the cessa-
and trichophytin [62, 345], but caused a transient decrease tion of therapy, at a time when the peripheral white
of in vitro LPRs to PHA and PPD [370]. Intravenous blood cell count was at its nadir.
5-FU produced a rapid (within 1–2 days) decrease in Perhaps the most intriguing aspect of CTX-induced
absolute T- and B-lymphocyte levels, which returned to immune changes has been the elucidation of the selective
baseline over the ensuing 1–3 weeks [152]. Ara-C has immunopotentiating effects of CTX [144, 325]. For
been shown to partially suppress primary and secondary example, in a study of 22 patients with metastatic cancer,
antibody responses [117]. Although both MTX and CTX pretreatment significantly augmented the develop-
Ara-C abolished established DTHS to recall antigens, ment of DNCB reactivity as well as DTHS to new anti-
Ara-C was much more potent in inhibiting DNCB reac- gens [44], even though absolute lymphocyte counts fell
tivity [346]. In vitro MTX did not affect the phagocytic or within 1–2 days and did not recover for 21 days [46]. The
cytolytic activities of human neutrophils [255]. DTIC has T-cell, B-cell, and T-cell subset numbers were all affected
been considered only weakly immunosuppressive in equally. Lymphoproliferative responses to mitogens and
humans, based on studies in patients with malignant mel- alloantigens also fell significantly within a day, but recov-
anoma in which only a minority of treated patients exhib- ered to pretreatment levels by day 3; some cases exhibited
ited depressions in antibody production to typhoid vaccine rebound overshoot by day 7. Inducible T-cell suppressor
or in DTHS reactions to primary sensitization [89]. More cell activity was also diminished within 1 day after CTX
recent studies with DTIC have indicated that treatment is administration; however, in contrast to LPRs, suppressor-
associated with a cumulative decrease of T-cell numbers, cell activity remained significantly impaired on day 3 and
but no change in B-cell numbers [52]. A normalization of only partially recovered by day 7. Thus, between 3 and 7
Tind cell numbers occurred after cessation of treatment. days after intravenous administration of CTX, there
appears to be a preferential impairment of nonspecific
T-cell-mediated suppressor cell activity, which could
Alkylating Agents account for the augmented DTHS noted in CTX-treated
The alkylating agents comprise a variety of drugs includ- patients. Most recently, a single intravenous low dose
ing cyclophosphamide (CTX), nitrogen mustard (HN2), (300 mg/m2) of CTX was shown to inhibit the generation
chlorambucil (CLB), thiotepa (TT), and busulfan (BS). of inducible suppressor cell activity in cancer patients for
These compounds have found widespread clinical use, par- up to 19 days without affecting lymphoproliferative
ticularly for the treatment of leukemias and lymphomas. responses to T-cell mitogens or the Tind/Tc/s ratio [45].
Their antiproliferative effects also extend to normal host This CTX dose (300 mg/m2) 3 days prior to immunother-
hematopoietic precursor cells of all lineages, including apy has been employed clinically in a variety of circum-
lymphoid precursors. In animals, these agents are potent stances in an attempt to abrogate suppressor cell activity,
immunosuppressants of antibody formation [447]. for example, as an adjunct to active specific immunother-
Recent clinical interest has focused on the novelty of apy with a melanoma tumor cell vaccine [43], and in
CTX as an immunomodulatory agent [37, 144, 202, combination with low-dose intravenous IL-2 [344].
207, 213, 216, 325, 380]. Administration of oral daily Although the mechanism by which CTX augments the
CTX maintenance therapy to patients with lymphomas immune response has not yet been elucidated, the leading
did not result in interference with the development of hypothesis, at present, is an abrogation of suppressor-cell
normal humoral and cell-mediated immune response to function, with blockage of the subset of helper T-cells
keyhole limpet hemocyanin. Other studies have revealed that is the inducer of suppressor cells [144].
that prolonged oral therapy with CTX can produce lym-
phopenia and suppress in vitro LPRs to PHA. CTX
administered intravenously at conventional doses for 7
Antitumor Antibiotics
days inhibited the production of primary antibody Surprisingly, clinical information concerning the immu-
responses, but did not significantly interfere with DTHS. nosuppressive effects of this class of anticancer drugs
Low intravenous doses of CTX (100–600 mg/m2) pref- that includes such widely employed drugs as adriamy-
Robert K. Oldham 113

cin (ADR), daunomycin (DNR), mitomycin (MTC), exhibit varying degrees of immunomodulatory activity
and bleomycin (BLEO) is limited. However, in animal when incubated in vitro with human PBMC [492], but
models, these agents affect a variety of immune cells, in clinical data are sparse. Single intravenous doses of cis-
particular, macrophages and immunoregulatory cells platin have been associated with a rapid depression of
[144, 340], suggesting that they should exert a spectrum LPR, with recovery in 1–2 days, whereas a single dose of
of immunomodulatory effects in humans. On the other CCNU leads to depressed PHA responsiveness for up to
hand, ADR has been shown to augment both monocyte- 6 weeks, in the absence of changes in T- and B-cell pro-
and lymphocyte-mediated cytotoxicity of human PBMC portions. Long-term therapy with CCNU has been asso-
[20, 21, 282]. Following a single intravenous dose ciated with cumulative depressions of absolute levels of
(25 mg/m2), PBL from cancer patients showed an both and T cells [52]. The reduction of T-lymphocyte
increase in lymphocyte cytotoxicity as well as an levels was due mainly to a depletion of Tind cells. Taxanes
increase in Tc/s levels [20]. In cancer patients, intrave- have moderate immunosuppressive effects.
nously administered ADR has been shown to produce a
rapid but reversible lymphocytopenia of both T and B
cells [152]. In vitro, ADR has been shown to impair
Hormones
phagocytic function of human polymorphonuclear leu- Hormonal agents such as tamoxifen (TAM) and
kocytes [536], whereas similar effects were not seen medroxyprogesterone acetate (MPA) are used widely in
with a variety of other drugs, including MT, VCR, 5-FU, the treatment of breast cancer. Both estrogen and pro-
and cisplatin. In addition, ADR has also been shown to gesterone receptors have been identified in human lym-
inhibit granulocyte degranulation and release [95]. phocytes [117, 126]. There is considerable experimental
Some evidence has been provided that low doses of and clinical evidence suggesting that hormones can
ADR can also augment both T- and B-cell mediated modulate immune mechanisms [114, 371, 381]. Long-
immune responses [144]. Thus, it now appears as if term treatment of patients with TAM has not produced
ADR, like CTX, can exhibit a variety of immunomodu- significant immunosuppression [254, 451], whereas
latory activities. MPA depressed LRP as well as the Tind Tc/s ratio of treated
patients (451).
Vinca Alkaloids and Podophyllotoxins
The vinca alkaloids, vincristine (VCR) and vinblastine
Combination Chemotherapy
(VLB), possess a unique mechanism of antitumor activ- When multiple cancer chemotherapy drugs are adminis-
ity involving drug-induced microtubular dysfunction tered together, it is impossible to predict what effects
leading to mitotic arrest. VCR has been shown to depress their interactions will have on their individual immuno-
granulocyte aggregation, lysozyme release, and chemot- suppressive properties. Nevertheless, at the present
axis [95, 492]. However, there remains a substantial time, most cancer chemotherapy regimens include a
lack of studies concerning the immunomodulatory combination of drugs. A number of recent studies have
effects of these agents in humans. They are often admin- begun to define the immunological consequences of
istered in conjunction with steroids as treatment for combination chemotherapy as currently used for the
hematopoietic cancers, making it impossible to define treatment of patients with advanced disease, as well as
their selective immunosuppressive effects. Not unex- for patients treated in the adjuvant setting.
pectedly, they inhibit human LPR in vitro, and they A comparison of intravenous doses of single drugs,
should exert similar effects in vivo. Both VCR and VLB namely ADR and 5-FU, with combinations such as
have not been particularly immunosuppressive in COBAM (CTX, VCR, BLEO, ADR, MTX) or DOMF
humans [208]. Vindesine, a new semisynthetic vinca (DTIC, VCR, methyl-CCNU, 5-FU × 5) revealed that
alkaloid, was devoid of immunosuppressive activity in both the single agent and multiagent regimens produced a
preliminary clinical trials [425]. Podophyllotoxins such rapid drop (within several days) in the percentage and
as VP-16 have mild immunosuppressive effect, similar absolute numbers of circulating B and T cells, which was
to the vincas. more pronounced for the combined drug regimens [152].
Such decreases were transient, and either a partial or
complete recovery (with or without rebound) to baseline
Other Drugs levels was then observed over the 1 or 2 weeks after drug
Other frequently employed anticancer drugs, such as administration [152]. Similar acute effects of multiagent
cisplatin, nitrosoureas (BCNU, CCNU) and taxol also chemotherapy regimens have been noted with respect to
114 Therapeutic approaches to cancer-associated immune suppression

functional immune parameters such as in vitro LPRs [87], improvement and/or normalization of immune cell
B-cell activation [232], reticuloendothelial cell function numbers and function for patients who achieve clinical
[151], and cytotoxic cell (NK) activity [83, 442]. In gen- remission following chemotherapy or radiation therapy.
eral, there has not been a good correlation between the Nevertheless, some immunologic impairment persist
depressions in immune cell numbers and functions. for years after completion of successful treatment. Thus,
Cyclic combination chemotherapy can result in cumu- in many cases, it may be extremely difficult to dissoci-
lative immunosuppressive sequelae [315, 396, 501]. ate the chronic immunologic consequences of therapy
Three similar studies have all involved patients receiving from the presence of persistent immune defects while in
cyclic adjuvant chemotherapy for breast cancer who did clinical remission.
not exhibit evidence of immune suppression before treat-
ment. Serial immune assessments revealed that cyclic
Solid Tumors
chemotherapy was associated with a marked initial
(within 1 month) decrease in T- and B-lymphocyte num- Patients surgically cured of their cancers generally have
bers, and then gradual progressive further decreases a restoration of immune cell numbers and function after
[501], the same being observed in Tind Tc/s ratios [396]. the perioperative periods [260, 437, 483]. Although
There was also a rapid initial decrease in functional patients in clinical remission following potentially cura-
immune parameters, such as in vitro LPRs to antigens, tive radiation therapy may have some normalization of
with stabilization or some improvement over the ensuing immunity, it does not usually become apparent until
year of treatment. In general, the functional immunologi- years after the completion of treatment [375]. Finally, as
cal parameters tended to normalize during or, more often, discussed previously, it has also become apparent that
within several months of stopping chemotherapy, whereas adjuvant chemotherapy for breast cancer results in
the abnormalities of immune cell numbers were still cumulative depressions of both lymphocyte numbers
apparent 1 year after cessation of treatment [315]. Thus, and functions [315, 396, 501].
it appears that prolonged administration of cyclic combi-
nation chemotherapy results in cumulative impairments Hematopoietic Malignancies
of immune cell numbers and functions, some of which
are long-lasting [439, 501]. In none of these studies were Much important information concerning the long-term
investigators able to predicate relapses in individual effects of chemotherapy and radiation therapy in cured
patients based on immune parameters. patients has resulted from studies of children who
Recent attempts to manipulate the immune system received maintenance chemotherapy for acute lympho-
have involved the combination of total body irradiation blastic leukemia and of adult patients with HD. In acute
with aggressive, but non-ablative, chemotherapy. Such lymphoblastic leukemia patients who remained in clini-
therapeutic studies are being done to deplete regulatory cal remission for 1–3 years after completion of chemo-
cells that negatively influence T-cell function followed therapy, immune competence was found to recover.
by the reinfusion of T-lymphocytes derived from the Those patients destined to relapse showed a subsequent
patient’s own tumor, so-called tumor-infiltrating lym- deterioration in immune status, which was detectable
phocytes (TIL). These studies, which will be described some months before clinical relapse [178, 210, 211,
in more detail elsewhere in this book, are producing 212, 214, 472]. The immune status of patients with HD
clinical responses in 50% or more of patients with in long-term clinical remission after treatment with che-
advanced melanoma [573]. Such aggressive approaches motherapy or radiation therapy has also been evaluated
indicate that the immune system can be manipulated in detail. At the completion of radiotherapy, DNCB
with chemotherapy and radiation therapy, but the future reactivity was lost in almost all patients who were ini-
will probably in the area of more selective immune tially sensitive. However, many patients regained their
depletion with monocloncal antibody of the targets DTHS responses during the first year after discontinua-
agents imparting less general toxicity. tion of chemotherapy [7, 427] or radiotherapy [165,
428]. NK-cell activity also improved following success-
Immune Status of Patients in Clinical ful therapy for HD. [161]. Several studies have shown
that, following the completion of successful MOPP che-
Remission
motherapy, patients in remission from HD exhibited
A number of studies have evaluated the immune status gradual improvements of in vitro T-cell functions [7,
of patients in remission following the successful treat- 427]. In contrast, other studies have revealed a persistent
ment of their cancer. In general, there tends to be depression of LPRs at 1–10 years after the completion
Robert K. Oldham 115

of treatment, with no evidence for recovery [56, 57, 100, Table 1. Biological response modifiers with immunorestor-
154, 189, 428], along with a preferential chronic deple- ative properties
tion of Tind cells [29, 185, 404, 427, 432]. In some of Chemicals Biologicals
these studies, patients had received radiation therapy, Azimexone Bestatin
which was felt possibly to contribute to the prolonged Cimetidine CSFs
Copovithane FK-565
immune deficiency [165, 185, 404, 432]. Several authors Coumarin IMreg-1
have argued, however, that patients with HD in pro- DTC ImuVert
longed clinical remission continue to manifest immune Ibuprofen Interleukin 1-–32
abnormalities as a reflection of their underlying disease Indomethacin Interferon gamma
Interferon inducers Lentinan
[56, 57, 100, 154, 291]. For example, persistent defects
Isoprinosine OK-432
in T-cell functions were more severe in irradiated Levamisole Retinoids
patients with HD than in similarly treated patients with NPT 15392 T-Cell reconsti-
testicular cancer [47, 432]. Although some improve- Oxyphenbutazone tuting factor
ments were noted on serial immune assessments follow- Piroxicam (SR 270258)
Ranitidine Thymic factors
ing discontinuation of treatment, there have been no Transfer factor
correlations between the status of immunity during clin- Tuftsin
ical remission and likelihood of relapse [427, 428]. It
has been suggested that an increased sensitivity of
T-cells to the inhibitory effects of suppressor cells may
account for the persistent depressions of T-cell func- the protective effect of a Brucella vaccine [423] led to
tions in cured HD patients [535]. widespread clinical investigation of the immunomodu-
latory activity of both tetramisole and levamisole. In
various animal models, and also following in vitro incu-
Treatment of Cancer-associated bation with effector cells from human donors, levami-
sole has been found to increase both T-cell numbers and
Immunodeficiency functions (e.d., LPR), if initially depressed [424, 574],
The term immunotherapy was introduced to clinical as well as phagocytic and chemotactic activities of poly-
oncology 2 decades ago following the independent morphonuclear leukocytes and monocytes [111]. Its
observations that Bacillus Calmette-Guerin (BCG) immunorestorative mechanism of action is currently
administration could prolong the survival of patients unknown [220]. It has been shown to induce thymic
with acute lymphoblastic leukemia [326] and induce factor-like activity, which has been attributed to the
regressions of injected as well as noninjected malignant presence of a sulfur atom in its structure [186].
melanoma lesions [357]. Since these reports, a wide Levamisole, which contains an imidazole ring, may
variety of chemicals and biologicals have been adminis- function like imidazole in affecting enzymes that con-
tered to immunosuppressed cancer patients in the hope trol cyclic nucleotide levels in lymphoid precursors of
of reconstituting or boosting host immune mechanisms lymphocytes. Both imidazole and levamisole, which
(Table 1). A broader term, biotherapy, is now being used themselves are not mitogenic, elevate cyclic GMP levels
since there are many biological substances that stimu- in lymphocytes in vitro and enhance their proliferative
late cells outside of those from the immune system. responses to mitogens or foreign antigens [111].
These stimulations with colony stimulating factors, growth Only a limited number of dose- and schedule-seeking
and maturation factors may have secondary effects on trials for cancer patients were performed with levami-
cancer and immune function. sole. In general, it has been administered intermittently,
using two or three daily doses every 1–2 weeks [9]. The
drug is well absorbed, and a single oral dose of 150 mg
Immunorestorative Agents produces a peak plasma level in 2 h (0.49 + 0.05 μg/ml),
which is the concentration required for in vitro activity.
Chemicals
The plasma half-life of levamisole is 4 h. It is widely
Levamisole, an orally active synthetic phenylimidazole, distributed and can be detected in all tissues and fluids,
2,3,5,6-tetrahydro-6-phenyl-imidazol[2, 1-6]thiazole, is with the highest levels in liver and kidneys. It is excreted
the levo isomer of tetramisole, a potent, broad-spectrum primarily in the urine, most of it by 24 h, although much
antihelminthic agent introduced in 1966 [517]. The of the excreted product has already undergone extensive
demonstration in mice that tetramisole could augment metabolic changes.
116 Therapeutic approaches to cancer-associated immune suppression

A number of side effects of levamisole have been 517]. Extensive tolerance and safety studies have been
reported [411, 414, 415, 431, 503, 568]. In studies on conducted in which IPS was administered orally for peri-
3,900 patients with a variety of diseases (including ods of 1 week to 2 years at doses of 1–8 g/day. Minimal
rheumatic and inflammatory diseases and cancer), reac- side effects have been noted, including transient rises in
tions included idiosyncratic or allergic ones, such as a serum and urine uric acid levels and, occasionally, tran-
rash or febrile influenza-like illness, sensorineural reac- sient nausea associated with higher daily dosages.
tions such as alterations of taste and smell, and gastroin- Following oral or intravenous administration to rhesus
testinal symptoms. Rashes and fever resulted in the monkeys, the inosine moiety of IPS is rapidly metabo-
cessation of levamisole treatment in 7% and 1.5% of lized, with a half-life of less than 4 h.
cases, respectively, but were quickly reversible. The Azimexone (BM 12.531) is an orally active aziridine
major serious side effects have been agranulocytosis dye, 2-[2-cyanaziridinyl-(1)-2-[2-carbamoylaziridinyl-
and/or neutropenia and, less commonly, thrombocy- (1)]-propane, which has been found to increase the
topenia, which have been observed in between 0.2% and number of cytotoxic autoreactive cells. It exhibits anti-
2% of all treated patients. Agranulocytosis could not be tumor activity in animal models [53, 111, 186] and
related to dose or schedule of administration, and was exerts a variety of immunomodulatory effects on T cells,
always spontaneously reversible following discontinua- monocytes, phagocytic cells, and NK cells. Azimexone
tion of treatment [9]. The incidence of side effects in has exhibited immunorestorative activity in various ani-
various clinical trials has varied from insignificant to mal models of infectious diseases. In vitro studies using
major, requiring interruption of therapy in up to 21% of PBL from advanced cancer patients revealed that
cases [385]. azimexone at various concentrations (0.2–10 μg/ml)
Levamisole was approved for the adjuvant treatment enhanced the LPR to PHA with a maximal effect at
of stage III colon cancer as an immunomodulator to be 0.2 μg/ml. Other studies have indicated that azimexone
used with adjuvant chemotherapy. It enjoyed some use also enhances the percentage of activated T lympho-
for several years in this setting, but is no longer used cytes in vitro.
because of improved regimens for the adjuvant treat- In a limited number of clinical studies, the only sig-
ment of stage III colon cancer. nificant side effect observed with intravenous adminis-
Isoprinosine (IPS), a synthetic antiviral agent, is a tration has been a dose-dependent self-limiting hemolysis
complex of inosine and the p-acetamidobenzoate [358]. Oral absorption of azimexone is almost complete,
(PacBA) salt of N,N-dimethylamino-1-propanol (DIP) and the serum half-life is 6 h.
is a 1:3 molar ratio [111, 186]. In early clinical trials
with rhinovirus-infected humans, IPS increased the Histamine Receptor Antagonists
titers of circulating antiviral antibody, suggesting it had Cimetidine is a histamine type II receptor antagonist
B-cell immunomodulatory activity [489]. Subsequent in widely used for the treatment of gastrointestinal ulcers.
vivo studies in animal models and in vitro studies with A growing body of evidence has suggested that suppres-
human PBMC have indicated that IPS enhances T-cell sor T cells that possess histamine receptors (H2 type)
functions such as LPR to mitogens and alloantigens may play an important immunoregulatory role in nor-
[111, 186, 356]. Helper T cells appear to be the main mal immunologic responses [88, 149, 377]. The ratio-
target for the drug in humans. IPS has also been shown nale for administering H2 blockers to cancer patients is
to induce the appearance of T-cell surface markers in based on the observation that cimetidine could abrogate
mouse prothymocytes, similar to that of thymic factors, in vitro histamine-induced suppressor T-cell activity
as well as increase the proportions of various T-cell sub- using human PBL. Results of in vitro preclinical testing
populations following incubation with human PBMC. with cimetidine have been summarized [329]. It has
In vitro, IPS at a concentration of 100 μg/ml restored been shown to augment LPRs and IL-2 production of
LPRs, NK activity, and monocyte chemotaxis of PBMC PBMC from cancer patients to mitogens and alloanti-
isolated from cancer patients [526]. Because the immu- gens [148, 289, 513]. In vitro cimetidine also enhances
nomodulatory activity of IPS is similar to that of a variety NK-cell activity of PBMC from cancer patients, proba-
of thymic factors, it has been classified as a thymomi- bly through its inhibitory effects on suppressor cells
metic drug. [156]. No effects have been seen on transformation of
Although IPS has been investigated clinically in several peripheral blood monocytes to macrophages [169].
different viral diseases (including human immunodefi- Cimetidine was employed in a phase I/II study [323]
ciency virus [HIV] infections), only a limited number of and in combination with coumarin [322, 518, 519].
studies have been performed with cancer patients [111, Varying degrees of antitumor activity have been noted
Robert K. Oldham 117

in patients with malignant melanoma [518, 519] and compound [186, 422]. DTC is a chelating agent in use
renal cancer [322] treated with the combination of for the treatment of heavy-metal poisoning. In animal
cimetidine plus coumarin. The mechanism of antitumor models, it exhibits a variety of immune-augmenting
activity has not been established but the doses used are effects on T-cell dependent immune responses as well as
approximately 2x that for treatment of ulcers. on the induction of T-cell differentiation. No significant
Histamine has been shown to suppress human LPR to toxicity has been noted following long-term administra-
mitogens, and cimetidine might block the inhibition. tion. Clinical trials in cancer patients and in patients
More recently, histamine was used in clinical trials as a with HIV disease have not been promising.
drug to assist IL-2 induced T cell function with some Coumarin (1,2 benzopyrone) has been reported to
evidence of useful clinical effects in melanoma metastases exhibit immunomodulatory activity [49, 581]. Unlike
to the liver. The clinical effects were marginal. warfarin, coumarin is devoid of anticoagulant activity. In
cancer patients, coumarin administration was reported to
Nonsteroidal Anti-inflammatory and Antipyretic enhance LPRs to PHA but did not effect T-cell numbers
Agents (NSAID) [49]. In vitro coumarin has augmented HLA DR expres-
Indomethacin and ibuprofen are prototype inhibitors of sion and NK activity [581]. It has been administered in
prostaglandin synthesis. A large number of in vitro combination with cimetidine to patients with malignant
studies using PBMC from patients with solid tumors melanoma [518, 519] and renal cancer [322] with varying
and HD have suggested that (a) the depressed LPRs of degrees of antitumor activity reported. The mechanism of
cancer patients result, at least in part, from the immu- antitumor activity of the combination has not been
nosuppressive effects of prostaglandins released by established.
activated monocytes/macrophages, and (b) indomethacin Copovithane (BAYi7433) is a copolymer of 1,3-bis
is capable of abrogating the suppressor cell influence. (methyl amino carboxy)-2 methylene propane carbamate,
In addition, it has been shown in vitro that indometha- has exhibited antitumor activity in a variety of pre-clinical
cin also acts directly on T cells of patients with malig- models [458]. A phase I trial in advanced cancer patients
nant melanoma to augment mitogen responsiveness using weekly intravenous dosing revealed minimal
[520]. It has been postulated that this direct immuno- fatigue, and occasional nausea and proteinuria, as the
modulatory action results from specific pharmacologic only side effects, some antitumor effects, and some
effects such as alteration of intracellular cyclic AMP improvements in Tind/Tc/s and in vitro toxicity responses
levels. Finally, a correlation has been observed between and LPRs [233].
tumor spread and content of prostaglandin E2 of the
tumor [42]. These observations have formed the basis Biologicals
for administering prostaglandin inhibitors to cancer
patients [96, 405, 563]. Thymic Factors
A functioning thymus gland is an essential requirement for
NPT 15392 the normal development and maintenance of cell-mediated
Studies with IPS suggested that the inosine moiety immunity [73]. The thymus is responsible for the nor-
might be responsible for its immunomodulatory activity mal maturation of all the various subclasses of
on LPRs. Subsequently, a number of purines with inos- T-lymphocytes, including various effector cells, as well
ine-like structures were synthesized, one of which is as immunoregulatory cells. The thymus exerts its influ-
NPT 15392 (9-erythro-2-hydroxy-3-nonylhypoxan- ence during the ontogenesis of the immune system of
thine) [157, 186, 578]. In animals, NPT 15392 has releasing, in situ from its epithelial stroma, a variety of
exhibited effects on T-cell and monocyte/macrophage differentiating factors that induce the maturation of resi-
functions and T-cell differentiation similar to those of dent pre-T stem cells (thymocytes) into mature T cells,
the parent compound [157]. Toxicological studies have which ultimately circulate in the peripheral blood and
shown that it is nontoxic at oral doses up to 35 mg/day. lymph. It is now well established that the thymus gland
This drug has been tested in clinical trials in cancer is an endocrine organ and that at least several of its
patients [575]. locally released differentiating factors are also secreted
DTC, sodium diethyldithiocarbamate (Immunothiol), into the bloodstream. Thymic hormone-like bioactivity
was developed as a result of preliminary studies with has been demonstrated in the blood of animals and
levamisole in an attempt to synthesize a chemically humans. This activity decreases following thymectomy,
defined sulfur-containing compound that would be la as well as with age, in parallel to the physiological age-
more potent immunorestorative agent than the parent dependent involution of the thymus. The presence of the
118 Therapeutic approaches to cancer-associated immune suppression

thymus is important well into adulthood in order to solid-phase, and recombinant DNA techniques, but only
maintain immune T-cell mechanisms. the chemically synthesized material has been employed
A number of factors with thymic hormone-like activity in clinical trials. Thymopoietin, THF, and TFX are purified
have been prepared from thymus tissue and blood, and thymic peptides with molecular weights ranging from
these are in varying stages of characterization [173, 372, 3,220 to 5,562. TP-5 is a biologically active synthetic
463, 478, 485]. The best studied are thymosin fraction 5 pentapeptide that represents amino acids 32–36 of thy-
(TF5), thymosin α1 (Tα1), prothymosin α (Pro Tα), thy- mopoietin. In contrast to all other thymic preparations,
mostimulin (TP-1), thymulin (FTS-Zn), thymopoietin thymulin (FTS-Zn) has usually been isolated from pig
(TP), thymic humoral factor (THF), and thymic factor x blood rather than thymus tissue. Although thymulin,
(TFX). These agents all exhibit a broad spectrum of Tα1, and thymopoietin are all detectable in the blood,
immunorestorative effects on T-cell numbers and func- only thymulin levels drop significantly following
tions in animal models and humans. In some bioassays, thymectomy, and are restored by thymic grafts.
many of the well-characterized thymic preparations have None of the well-characterized thymic peptides exhibit
identical, or even opposite, effects. An increasing num- any significant homology with the other characterized pep-
ber of thymic factors have been employed therapeuti- tides. However, a 50% homology has recently been identi-
cally in treating a variety of diseases – predominantly fied between a 35 amino acid region of Tα1 and that of the
cancer and HIV disease [134]. p17 core protein of the AIDS retrovirus (HIV) [449].
The well-characterized thymic preparations are listed Thymic factors, which have been administered to
in Table 2 [173, 192, 463]. Among the thymic factors more than 1,000 cancer patients, have shown minimal
that have been administered to cancer patients are TF5, toxicity, but some have been approved for general clinical
Tα1, TP-1, THF, and TFX. Both TF5 and TP-1 are par- use. The purified preparations that have been adminis-
tially purified extracts of calf thymus glands. They tered by intramuscular or subcutaneous injection have
include a mixture of different biologically active as well not produced any significant side effects [173, 463].
as inactive polypeptides. The purification procedures Partially purified bovine preparations, such as TF5, have
for TF5 and TP-1 are similar but not identical. TF5 con- produced rare (less than 1%) allergic reactions and ery-
sists of ten major – and at least 30 minor – polypeptides thema and/or pain at the sites of injection in about one
on analytical isoelectric gel focusing with molecular third of treated patients. However, potentially immuniz-
weights ranging from 1,000 to 15,000. The first biologi- ing treatment schedules, such as daily injections for 1
cally active polypeptide isolated from TF5 is Tα1. week followed by 3 weeks of rest, and reinstitution of
Biologically active Tα1 has a molecular mass of 3,108 treatment, have been associated with a high (10%) inci-
daltons. It has been synthesized by classical chemical, dence of anaphylactoid-like reactions. As a single agent,
TF5 has exhibited minimal antitumor activity in patients
with renal cancer in one study [465] and none in another
Table 2. Chemical properties of thymic hormones [135]. More recently, thymic factors have been used less
due to the availability of specific lymphokines with
Name Chemical properties more powerful activities.
Thymosin fraction 5 (TF5) Heat-stable, acidic Peptides MW
1,000–15,000 T-cell Reconstituting Factor
Thymosin α1 (Tα1) Peptide of 28 residues, MW 3108
Prothymosin α (Proα) 113 Amino acids, MW 13,500 This is a highly purified protein fraction isolated from
Thymosin α9 Acidid peptide, MW 2000, pl 3.5 human serum that has been shown to exhibit immuno-
Thymosin α11 (T α11) Peptide of 35 residues, 28 residues modulatory effects on T-cell numbers and functions
identical [364]. In preliminary phase I/II trials, no unexpected tox-
To Tα1 icities have been observed following SQ administration.
Thymosin β3 (T β3) Peptide of 49 residues, MW 5700,
43 residues TsIF is a thymic isolate, distinct from other thymic
Identical to Tβ4 factors and cytokines, that induces immature bone mar-
Thymosin β4 (Tβ4) Peptide of 43 residues, MW 4963 row cells to differentiate into competent suppressor T
Thymic factor X (TFX) Mixture of peptides; active cells. It has a molecular mass of 75,000 daltons as deter-
compound is a peptide, MW 4200
mined by gel filtration and high-performance liquid
Thymostimulin (TS/TP-1) Mixture of peptides
Thymulin (FTS-Zn) Nonapeptide, MW 857, chromatography. Recent studies in mice indicated that
Thymic humoral factor Peptide MW 3200 TsIF administration suppresses the development of
(THF) autoimmune disease in lupus-rheumatoid arthritis-prone
Thymopoietin (TP) Peptide of 49 residues, MW 5562 animals [341].
Robert K. Oldham 119

Transfer Factor have been shown to restore PHA responsiveness, to


In 1955 it was demonstrated in humans that the transfer induce augmentation of cytotoxic and helper T-cell
of DTHS to streptoccal M substance and tuberculin could numbers and functions, and to inhibit prostaglandin
be accomplished by administration of a suspension of synthesis by host tumor-activated macrophages.
leukocytes disrupted by either distilled water or repeated However, the role that altered immune mechanisms play
freeze-thaw cycles [294]. The substance (or substances) in the chemopreventative action of retinoids, such as
responsible for this transfer was resistant to RNAase and 13-cis-retinoic acid, remains unclear (see Chapter 16).
DNAase and was termed transfer factor. This initial work FK-565 [heptanoyl-8-D-Glu-(L)meso-α1-A2pm(L)
was extended using dialyzable human leukocyte extracts AlaOH] is a heptanoyl tripeptide analogue of FK-156, a
from patients sensitized to various antigens [25], includ- biologically active acylpeptide isolated from fermenta-
ing skin allograft antigens [493]. The crude dialyzable tion products of Streptomyces. Preclinical studies in mice
preparation has subsequently been shown to enhance have shown that FK-565 augments NK-cell activity and
LPRs in vitro [24, 142]. The further chemical character- macrophage functions as well as T-cell functions both in
ization of transfer factor has been hampered by the lack vitro and in vivo [507]. However, in animals, mitogen-
of a unique biological assay to monitor final purification. induced IL-2 production is decreased [4]. Clinical trials
Preliminary fractionation studies of human leukocyte in cancer patients have not been promising.
extracts capable of transferring DTHS responses have Bestatin [(2S,3R)-3-amino-2-hydroxy-4-phenylbutanoyl-
indicated that transfer factor is probably a low-molecular- L-leucine] is a low-molecular-weight immunomodifier
weight material (approximately 1,000) with the electro- found in supernatants of Streptomyces olivoreticuli
phoretic mobility of slow γ-globulin but with no reactivity [327]. It is a competitive inhibitor for the enzymes
to anti-immunoglobulin antisera. Its possible composi- aminopeptidase B and leucine aminopeptidase. These
tion, that is, a short polypeptide chain joined with a three- enzymes are associated with the outer membranes of
or four-base segment of RNA, has been difficult to verify. most mammalian cells, including lymphocytes. In ani-
Unlike other nonspecific immunorestorative BRMs, mal models, bestatin augmented both humoral as well
transfer factor appears to exert antigen-specific immune- as cell-mediated immune responses [60, 69], particu-
restorative effects. larly in immunosuppressed mice. Macrophage activa-
Despite the lack of readily reproducible in vitro or tion, but not NK activity, was also observed both in vitro
in vivo assays, the effects of transfer factor have been and in vivo [506]. It has also been shown to enhance
studied following subcutaneous administration to patients T-cell numbers and cytotoxic functions following in
with a variety of immunodeficiency diseases, including vitro incubation with human lymphocytes, but it has not
cancer [354]. significantly influenced LPR [69]. It has been shown to
augment phagocytic cell function in vitro.
Interleukins Bestatin has now been chemically synthesized and
Many of the BRMs currently being synthesized by recom- is in clinical trials. The drug has been well tolerated in
binant DNA techniques are products of lymphocytes cancer patients at daily doses of 30 mg for up to 2
(lymphokines), monocytes (monokines), or other cells years [60, 69, 369, 576], but has shown little clinical
(cytokines). Various interferons and interleukins have activity.
been purified to homogeneity produced by genetic engi- Tuftsin is a naturally occurring tetrapeptide (Thr-Lys-
neering to make large quantities available for clinical tri- Pro-Arg), found normally in human and animal plasma,
als. Various other cytokines are undergoing active clinical and represents residues 289–292 of the heavy chain of
investigation [187]. The characteristics and results of clini- γ-globulin [121, 157, 365, 395]. It is released enzymati-
cal trials with these materials are discussed in Chapter 8. cally from a protein carrier (Leukokinin) and is capable
of activating various functions (particularly phagocyto-
Retinoids sis) of polymorphonuclear leukocytes and monocytes/
Considerable interest has recently been focused on the macrophages, T-cell cytolytic activity, and IL-2 produc-
influence of vitamin A (retinol) and its natural and syn- tion at physiologic concentrations [125, 327]. It has also
thetic derivatives (Aretinoids) on the growth and differ- been shown to enhance antibody production to thymic-
entiation of neoplastic cells. Although retinoids have dependent as well as thymic-independent antigens in
been administered to cancer patients primarily as animals and Ia-suppression. It has also been able to
chemopreventive agents [74, 308], accumulated evi- reduce tumor necrosis factor in animals [571].
dence also indicates that they have beneficial effects on Lentinan is a purified polysaccharide obtained from
the host immune system. For example, in animals they the extracts of the edible mushroom Lentinus edodes
120 Therapeutic approaches to cancer-associated immune suppression

[66, 110, 243, 373]. Chemically, lentinan is a β-(1,3)- II study to assess the tolerability and immunomodula-
glucan with some β-(1,6)-glucoside side chains. It has a tory effects of the agent in advanced cancer patients has
molecular weight of about 500,000. Evidence has been been followed by phase III trials with random experi-
presented that lentinan is a T-cell adjuvant, but a variety mental designs in which survival was the end point and
of immunomodulatory activities have been observed immunological monitoring was minimal, or even omitted.
[109, 327]. In animal models, it has been shown to aug- Thus, it has been difficult to make any firm conclusions
ment antibody production only in the presence of T concerning the immunorestorative properties of these
cells. It has also been shown to augment a variety of cyto- agents in cancer patients.
toxic effector-cell mechanisms following administration A number of preliminary small-scale phase I and II
to animals and to trigger production of various kinds of studies have indicated that levamisole [111, 302, 349,
serum factors including IL-1, CSF, and IL-3 [109]. In 366, 558], isoprinosine [111, 356, 403], azimexone [53,
vitro, it activates monocytes/macrophages and NK-cell 388], bestatin [69, 288, 328, 369, 560, 576], OK-432
activity of PBMC from cancer patients but does not [530, 545], retinoids [337], NPT 15392 [575], DTC
enhance LPR or LAK-like cytotoxicity [263]. [422], coumarin [49], various thymic factors [173, 463],
OK-432 is a heat-killed substrain of Streptococcus lentinan [245], cimetidine [289, 329, 426, 488], and
pyogenes that has been studied extensively in Japan. transfer factor [493] could improve T-cell, NK-cell, or
OK-432 predominantly acts by augmenting NK-cell B-cell numbers and/or functions in advanced cancer
activity [378, 539, 545], as well as augmenting mac- patients with pretreatment abnormalities. In general,
rophage [269] and T-cell [223] cytotoxicity. It has also however, the effects have not been striking. When
been shown to induce various cytokines such as inter- administered to patients without immunodeficiencies,
feron, IL-1, and IL-2 [242, 368, 443]. A variety of however, therapy was often followed by a deterioration
Japanese phase I, II, and III studies have been performed of immune competence. In several reports, no immuno-
in cancer patients in which it has been administered intra- modulatory effects of levamisole were observed on
dermally [106, 115, 339, 545], intramuscularly or intral- T-cell numbers or functions [200, 558]. A study of
esionally [539]. The major toxicities have been fevers patients with colorectal cancer suggests that the major
and local inflammatory reactions at injection sites. in vivo effect of levamisole is augmentation of mono-
ImuVert is prepared from the bacterium Serratia cyte chemotaxis [179]. Of the thymic factors, THF and
marcesseus. Its primary components are vescicles TFX have been employed predominantly in patients
derived from the bacterial membrane and ribosomes with infectious complications, and reports have been
[554]. It augments natural killer-cell activity and has mostly anecdotal. Cimetidine has increased an in vivo
antitumor cell activity in animals. It has demonstrated local graft-versus-host reaction of advanced cancer
poor activity in clinical trials in patients with malignant patients [508]. However, in other studies, it had no
gliomas and other cancers. effects on immune cell numbers or functions, or on DTH
Imreg-1 is a natural, leukocyte-derived immunomod- [316]. Similarly indomethacin has been of only limited
ulator containing two low-molecular-weight peptides: a utility in restoring immunity in advanced cancer patients
dipeptide (tyrosine-glycine) and a tripeptide (tyrosine- [219]. The administration of cimetidine or ranitidine to
glycine-glycine). In vitro Imreg-1 enhances the production advanced cancer patients has been associated with
of various cytokines, including IL-2 [176]. Clinical trials improvements in performance status [92]. In one study,
to date have been limited to patients with HIV disease treatment with the combination of coumarin plus cime-
and have not shown much promise. tidine increased the percentage of monocytes and of
DR+ monocytes of treated patients [323].
Treatment of Cancer-associated
Immunosuppression: Phase I and II Studies Chemicals
Most of the agents described in the preceding section Hodgkin’s Disease
have already been employed therapeutically in patients HD was chosen because of the well-documented abnor-
with cancer. Of the chemical BRMs, levamisole has malities of T-cell immunity that are present prior to treat-
been by far the most exhaustively studied, whereas ment and that persist in patients during clinical remission.
among the biologicals, the interleukins have received Levamisole has been shown in vitro to enhance T-cell
the most attention. No organized approach has been proportions [50, 451] among PBMC of HD patients.
employed for the clinical evaluation of the immu- Treatment of patients with HD in remission for less than
norestorative BRMs. In general, a small phase I or phase 2 years after the completion of radiotherapy resulted in
Robert K. Oldham 121

improvement in T-cell percentages and functions [301]. negative prior to treatment. In a more recent report from
DTH reactions and PHA responses also increased [50]. the same group, 19 patients in remission for at least 6
Continuation of treatment beyond 3 months led to a months were randomized to receive TP-1 at 50 mg IM
slight but consistent decline of these parameters. daily, every other day for 35 days, or no treatment
[304]. Patients who received TP-1 were then main-
Solid Tumors tained on TP-1 twice weekly. Before treatment, patients
A pilot study was performed with levamisole in surgi- exhibited depressed percentages and absolute numbers
cally resected patients with malignant melanoma and of circulating T cells and Tind cells. Following 5 weeks
squamous-cell carcinoma of the head and neck [57]. of daily TP-1, the proportions and numbers of all T-cell
Patients treated with levamisole (150 mg orally twice a subpopulations increased significantly, while alternate-
week) for 6 months exhibited improvements in absolute day treatment was not as effective. Maintenance TP-1
circulating T-cell levels, and only one of eight mela- therapy did not produce any further improvements in
noma patients treated with levamisole relapsed. Based T-cell or B-cell numbers or proportions. PBMC from
on these results, large-scale phase III were designed, the some patients also exhibited improvements in mitogen-
results of which have been reported as negative in one induced lymphokine production (e.g., IL-2, IFN-γ), but
and marginally positive in the others [412, 491]. the improvements were not statistically significant
Based on phase III data in resected colon cancer, overall. No significant changes were observed in a vari-
levamisole was approved for use with 5-fluoruracil as ety of serum markers including neopterin and
adjuvant treatment of resected stage III colon cancer. β2-microglublin. It was concluded that TP-1 has the
However, more active regimens have recently been potential to expand the T-cell pool, and, in particular,
available, making levamisole little used in the current Tind cells, in patients with HD in remission, but that
treatment of colon cancer. intensive (daily) induction therapy is required. In this
small sample, no conclusions could be established and
large-scale clinical trials have not been done.
Biologicals
Hodgkin’s Disease Malignant Melanoma
In one study, 19 consecutive untreated patients with TP-1 has been studied in a novel random-design trial
HD received TP-1 at a dose of 1 mg/kg daily for 7 days, involving 32 patients with localized stage I malignant
and immunological monitoring was performed prior to melanoma who were rendered disease-free following
treatment and again on day 8 [324]. A majority of surgery [51]. Subjects were selected from a cohort of
untreated patients exhibited depressed T-cell percent- 211 postsurgical patients who were monitored at
ages as well as depressed DTHS and LPRs to PHA. The 3-month intervals in the first 2 years after surgery. In
mean T-cell percentage increased significantly, from prior work, it was demonstrated that patients who devel-
47% to 55.7% (normal is 58.9%) following treatment oped low circulating T-cell levels were at high risk for
with TP-1, whereas the mean PHA response improved relapse. Patients who either exhibited presurgical
but did not totally normalize. Similarly, DTH responses depressions of total T-cell numbers or on serial monitor-
to recall antigens were positive in 53% of patients ing developed a depression of absolute circulating
before therapy, and in 95% after therapy. The patients T cells to less than 1,000/mm3 were considered eligible
most likely to respond to TP-1 were the most lym- for random assignment to one of three treatment arms:
phopenic, for whom an in vitro enhancing effect of TP-1 alone (25 mg IM once a week, eight patients); che-
TP-1 was observed on T-cell percentages and LPRs of motherapy alone (DTIC, 200 mg/m2 intravenously for
PBMC prior to therapy. In a similar study, 15 patients 5 days, repeated monthly, eight patients); or no further
with HD (in remission and off therapy for at least 1 therapy (16 patients). In the eight patients who received
year) were treated with 50 mg TP-1 by daily intramus- TP-1, T-cell numbers began increasing within 3 days
cular injections for 60 consecutive days [303]. For after the first injection, and weekly immunological
patients with initially depressed T-cell numbers, includ- monitoring revealed that levels returned to normal and
ing depressed helper (Tind) cell numbers, reconstitution were maintained at that level. Only inconsistent effects
to normal occurred within 30 days after the onset of on T-cell functions were seen in the group that received
therapy, but returned to pretreatment levels after the TP-1, and no changes were observed in any parameter
discontinuation of therapy. There was also a significant in patients who were untreated or who received chemo-
increase in NK activity, but in vivo DNCB reactivity therapy. Overall survival was better for the TP-1 group,
did not become positive in any patient who had been but the improvement was not statistically significant.
122 Therapeutic approaches to cancer-associated immune suppression

Chronic Lymphocytic Leukemia (CLL) patients, but did not affect DNCB reactivity [477]. A
Patients with Cll have also been treated with TP-1 [290]. Belgian colorectal trial used levamisole (150 mg for 3
TP-1 significantly increased Tind-cell proportions, lead- days every 2 weeks) in a matched group of patients oper-
ing to an improvement in the Tind/Tc/s ratio accompanied ated on by the same surgeon. In both the overall group
by an increase in LPRs to PHA and helper-cell function. and the 40 patients with Dukes B2 and C cancers, survival
In this study, the in vitro incubation of purified T-cells was prolonged with levamisole [538]. Improved survival
with TP-1 did not produce modifications of subset pro- of resected patients was reported in a broad trial of 177
portions or of immune functions. In contrast, in vitro patients with various gastrointestinal cancers treated with
incubation of T-cells with Tα1 from patients with stable- levamisole [347]. There was also improvement in DTHS
phase CLL led to an increase in T-cell proportions and reactions and in vivo LPRs. Similar results from the
an improvement in T-cell functions [41]. Japanese literature have been reported for patients with
advanced gastric cancer treated with levamisole [348].
An interesting recent report of a randomized trial
Phase II/III Surgical Adjuvant Studies involving 181 resected patients with gastric cancer
Chemicals showed an increase in median survival for patients who
received cimetidine [524].
Lung Cancer
The vast majority of large-scale phase III surgical adju- Phase III Levamisole Trials in Colon Cancer
vant studies have been performed with levamisole. Two Within the past decade, results of a series of phase III
randomized, double-blind trials in patients with non- trial results have been reported, in which levamisole
small-cell lung cancer have been reported [10, 16]. In was administered alone, or in combination with adju-
the first trial, a fixed dose of levamisole was adminis- vant chemotherapy in patients with resected colorectal
tered preoperatively for 3 days, and then for 3 days cancer [23, 32, 113, 244, 292, 293, 351, 474, 569].
every 2 weeks [10]. Treatment was continued for 2 A large-scale trial that was designed to compare treat-
years or until relapse. Among 211 patients, although ment with levamisole to surgery only reported by the
there was no difference in overall survival, levamisole European Organization for Research and Treatment of
treatment was associated with a reduction in distant Cancer (EORTC) Gastrointestinal Tract Cancer
metastases and in cancer deaths. When analyzed on the Cooperative Group [23]. This double-blind placebo-
basis of adequacy of drug dosage, it was found that the controlled trial involved 297 patients with Dukes C
greatest benefit of levamisole was seen with patients colon cancer. Levamisole dosage was based on body
who received a daily dose of 2.1 mg/kg or more. The weight and ranged from 100 to 250 mg/day for 2 con-
second study was designed as a confirmatory trial, and secutive days each week for a period of 1 year. Treatment
a weight-related dose of levamisole was employed, was started as soon as possible after surgery, usually
with a schedule identical to that in the original study within 2 weeks. With a median follow-up time of 3
[16]. Nevertheless, among the 217 evaluable patients, years, no benefit was seen in disease-free survival for
overall survival was decreased in those who received patients who received levamisole. Although the proportion
levamisole, because of non-cancer-related deaths in the of patients alive at 5 years was 51% in the levamisole
perioperative period. group versus 39% in the placebo group, this difference
was not statistically significant.
Gastrointestinal Cancer Other large-scale trials convincingly demonstrated
In the early 1980s, results of several small-scale trials that postoperative adjuvant therapy with levamisole plus
were reported that suggested that levamisole could pre- 5-FU impacts significantly on survival of patients with
vent perioperative immune suppression. A British report colon cancer [244, 292, 293, 351]. Thus, levamisole plus
in 1979 first indicated that the administration of levami- 5-FU was considered standard adjuvant treatment for
sole on 3 postoperative days could accelerate the recov- resected Dukes C colon cancer until folinic acid plus
ery of antitumor immunity (in an LMI assay) but not 5-FU proved as effective, less expensive and easier to
LPRs to PHA [570]. In a Japanese trial of 50 patients with administer. Levamisole is currently not used in the treat-
colon cancer, 15 were treated with levamisole at a dose of ment of colon cancer or any other human malignancy.
150 mg daily for 2 consecutive days, every other week,
beginning 3 days before surgery. Treatment with levami- Gynecologic and Urologic Cancer
sole appeared to prevent the postoperative depression of Positive surgical adjuvant studies have been reported
LPRs to PHA, as compared with a control group of with levamisole in postoperative patients with cervical
Robert K. Oldham 123

cancer [447] and bladder cancer [486] but is no longer Head and Neck Cancer
being used. The impact of preoperative perilesional therapy with
OK-432 was studied in 13 patients [539]. Treatment
Malignant Melanoma was associated with less pronounced decreases in NK
Only one study has focused on assessing in detail the activity of PBMC, and higher NK activity in draining
immunorestorative effects of levamisole in patients with lymph nodes, than in patients treated by surgery alone.
locally advanced malignant melanoma following surgi-
cal resection [253]. Levamisole was administered at Malignant Melanoma
150 mg/twice weekly. No improvements were noted in TP-1, transfer factor [90], isoprinosine [403], and TF5
T-cell numbers, or in LPRs to mitogens or antigens. [463] have been administered to postoperative patients
A large-scale phase III trial with levamisole involved with malignant melanoma. Each of these trials involved
203 postsurgical patients with malignant melanoma. No random treatment assignments and varying degrees of
improvement in either relapse rates or overall survival immune monitoring. No adjuvant effects could be dem-
was noted in patients who received levamisole [491]. In onstrated in these small studies. The trial with TF5 was
stage I patients, there was a trend in favor of levamisole complicated by the fact that only 45 patients were allo-
regarding time-to-recurrence and survival. More cated to treatment with either a low dose (4 mg/m2) or
recently, a large study involving 548 patients with com- high dose (40 mg/m2) administered subcutaneously,
pletely resected malignant melanoma having a poor concurrently with BCG, with or without DTIC chemo-
prognosis (Clark levels 3, 4, or 5, greater than 0.75 mm, therapy. A large-scale, randomized, double-blind phase
satellite lesions, in-transit metastases, or regional lymph III trial that involved 168 resected patients with high-
node metastases) revealed that patients treated postop- risk stage I and stage II malignant melanoma has been
eratively with levamisole (2.5 mg/kg orally on 2 con- reported [342]. Therapy (normal donor transfer factor or
secutive days weekly for 3 years) survived significantly placebo) was initiated within 90 days of resection and
longer than those who received either no postoperative continued for 2 years. With a median follow-up period
therapy or weekly BCG, or a combination of BCG alter- of 25 months, patients who received placebo exhibited a
nating with levamisole [412]. Median follow-up time trend for improved disease-free survival and overall sur-
for this trial was 5.1 years. In a randomized trial involv- vival. Thus, this large-scale trial has indicated that normal
ing 156 stage I Clark level 3, 4, and 5 patients, there was donor transfer factor is not an effective postsurgical
a trend for a delay in appearance of distant metastases in adjuvant therapy for malignant melanoma.
patients receiving levamisole (30 months versus 9
months for patients receiving placebo), but overall sur- Treatment of Radiotherapy-induced
vival was not significantly improved [309].
Immunosuppression
Although a number of clinical trials using immunorestor-
Biologicals
ative BRMs in irradiated patients have been reported,
Lung Cancer little information is available concerning whether or not
Several studies have focused on the effects of transfer treatment can prevent and/or reverse radiotherapy-induced
factor administered as an adjuvant to surgery. A tumor immunosuppression.
antigen-specific preparation, prepared from household
contact family members with positive reactivity to lung Chemicals
cancer antigen, was administered to 28 resected patients
with non-small-cell lung cancer twice by subcutaneous Levamisole
injection. Treatment was associated with a significant Of the putative chemical immunorestorative agents,
improvement in a variety of specific as well as nonspe- levamisole has been most extensively evaluated as an
cific immune parameters, including DTH reactions and adjunct to radiation therapy. Since the larger trials have
LPR to PHA [163]. In a randomized study of 63 postop- focused almost exclusively on survival as an end point,
erative patients with non-small-cell lung cancer (some very little information has been obtained concerning its
of whom received mediastinal radiotherapy), those who immunorestorative properties.
received transfer factor from normal donors beginning 1 Several of the smaller trials have included serial
month after surgery and continuing every 3 months immune monitoring. In a study involving 57 patients
showed a significant improvement in survival at 2 years with colorectal cancer treated with surgery and radia-
compared with nontreated patients [276]. tion therapy, levamisole at a dose of 150 mg/day for
124 Therapeutic approaches to cancer-associated immune suppression

2 consecutive days every other week, beginning concur- domized to postoperative radiotherapy, chemotherapy,
rently with postoperative radiation therapy, enhanced or both with or without levamisole, patients receiving
LPRs to PHA as compared with an untreated control radiotherapy plus levamisole exhibited improved disease-
group [293]; no effects were observed on DNCB reac- free and overall survival [278, 280]. These results are in
tivities. In a randomized double-blind study involving 71 contrast to two other similar studies. In a randomized but
postoperative patients with stage II breast cancer, 38% of not double-blind study, levamisole treatment was associ-
those who received levamisole (2.5 mg/kg/day on 2 con- ated with an increased recurrence rate [127]. In a trial
secutive days, twice weekly, beginning with the initia- involving 198 patients with resectable axillary node-
tion of radiation therapy) exhibited improvement in positive disease, levamisole, when begun following
DTHS to at least three recall antigens, as compared with completion of postoperative radiotherapy, had no effect
only 19% of those given a placebo [277]. Similar results on either disease-free or overall survival [525]. Nearly a
were noted in an earlier trial using DNCB reactivity third of patients treated with levamisole had to terminate
[473]. Thus, such studies suggested that levamisole may therapy prematurely because of toxicity (primarily leu-
improve some aspects of T-cell functions in irradiated kopenia, skin rash, nausea, fever, and mucosal infection).
patients. It has been argued that differences in patient characteristics
A number of large-scale cooperative group trials have led to these discordant results.
now demonstrated that levamisole administration does In head and neck cancer, three reports from the same
not improve the survival of patients treated with radio- Italian group have indicated that radiotherapy led to a
therapy. Four separate negative trials have been reported decrease in both T-cell numbers and functions (LPRs),
involving patients with non-small-cell lung cancer and that treatment with levamisole accelerated the resto-
(NSCLC). The Southeastern Cancer Study Group ration of T-cell counts, compared to patients who
reported that levamisole was without significant clinical received placebo [26, 27, 382]. With a median follow-up
benefit in a large randomized, placebo-controlled trial of 30 months, there was some improvement in disease-
of 251 patients undergoing radiotherapy for inoperable free interval for patients who received levamisole.
non-small-cell lung cancer [285]. In this study, levami- Another similar trial failed to demonstrate an improve-
sole was administered at a dose of 2.4 mg/kg twice ment in survival for patients with head and neck cancer
weekly beginning at the initiation of radiotherapy. The treated with levamisole [374].
median survival of patients treated with levamisole was
shorter than that of those who received placebo. Negative Isoprinosine
results have also been reported in a SWOG similar trial In a double-blind, placebo-controlled trial designed to
[566]. The Radiation Therapy Oncology Group (RTOG) evaluate the immunorestorative effects of isoprinosine
reported results of two separate randomized, placebo- following radiotherapy, one half of 106 irradiated
controlled trials, one involving 74 patients with resected patients with breast, head and neck, or uterine cancers
NSCLC and positive lymph nodes [215], and a second were randomly assigned to receive isoprinosine or pla-
involving 285 patients with unresectable tumors [395]. cebo in discontinuous courses for 5 months. After 3
Levamisole (2.5 mg/kg on days 1 and 2, weekly) or pla- months of treatment, 64% of the isoprinosine-treated
cebo was initiated at the beginning of radiation therapy patients exhibited evidence of improvement of DTHS
and continued for up to 2 years. Accrual to the first trial and in vitro functional tests, whereas only 23% of pla-
was terminated prematurely when survival data became cebo-treated patients did [356]. No correlations have yet
available from the second study indicating a worse survival emerged between immune reconstitution and clinical
(9 months versus 12 months). Thus, it appears to be con- status.
clusively established that levamisole combined with
radiation therapy has no benefit in the treatment of Prostaglandin Antagonists
NSCLC [400]. The nonsteroidal anti-inflammatory drug oxyphenbuta-
In the randomized trial involving 71 stage II breast zone was found to improve survival of patients with
cancer patients treated with radiotherapy postoperatively, stage III cervical cancer when administered during radi-
those who received levamisole exhibited a slight prolon- ation therapy [563]. This trial involved 160 patients.
gation of disease-free survival [277]. Among postmeno- Thirty of 73 patients with stage III disease were treated
pausal patients, levamisole significantly increased both with 100 mg oxyphenbutazone three times daily.
disease-free and overall survival, and the levamisole Treatment was associated with an improved survival,
group showed fewer distant metastases as the first sign but no studies were performed to assess the drug’s effect
of recurrence. In another trial involving 150 patients ran- on immune responsiveness.
Robert K. Oldham 125

Biologicals Transfer Factor


In a randomized, double-blind trial involving 100
Thymic Factors patients with nasopharyngeal carcinoma, transfer factor
Several clinical trials with TF5 and Tα1 have indicated (derived from young adults with a proven history of
that thymic factors may accelerate the reconstitution of infectious mononucleosis and from normal blood donors
T-cell functions following radiation therapy to head and with high anti-Epstein-Barr virus capsid antigen anti-
neck or mediastinal portals. The first clinical trial body levels) was administered for 18 months in con-
involved 75 patients with localized but unresectable head junction with radiotherapy [170]. This trial was based
and neck cancer [553]. Patients were randomly assigned on the association of Epstein-Barr virus with nasopha-
to receive TF5 (60 mg/m2) subcutaneously in a loading ryngeal cancers. No significant effects of transfer factor
dose (daily for 10 days, then twice weekly) schedule or were noted on disease-free and overall survival. Immune
no further therapy. Treatment with TF5 began concur- competence studies were not performed.
rently with the initiation of radiotherapy and continued A study was reported involving 111 patients with
for 1 year or until relapse. TF5 administration did not NSCLC who had surgery followed by radiotherapy [93].
prevent, nor could it restore, the marked T-cell lympho- Twenty-six patients received transfer factor (normal donor)
cytopenia that followed radiotherapy. Patients treated bimonthly beginning after RT was completed. Increased
with TF5 exhibited a prolongation of disease-free sur- DTHS and a lower relapse rate were observed in patients
vival, but of only borderline statistical significance. who received transfer factor, but the small sample size of
Similar findings have been reported with Tα1 in this trial precludes any definitive interpretations.
postradiotherapy patients with non-small-cell lung In a small randomized, placebo-controlled trial of 47
cancer [459]. This study was a double-blind, random- patients receiving radiotherapy with or without chemo-
design trial involving 42 patients with localized, unre- therapy for Hodgkin’s disease, DTHS skin responses
sectable disease who had just completed a course of were markedly enhanced in 22 patients who received
radiotherapy to the primary tumor and mediastinum. transfer factor (prepared from normal donor buffy coat
Patients who received mediastinal radiation were cho- cells), but no improvements were noted in a variety of
sen specifically as the study population, based on an in other immunologic parameters, nor in the prevention of
vitro study, indicating that TF5 could increase T-cell infection, including varicella/zoster [198].
proportions of PBMC from patients who had received
mediastinal radiation to other portals [272]. Patients T-cell Reconstituting Factor
whose disease regressed or remained stable at the com- A pilot clinical trial has been performed in which this
pletion of radiotherapy were randomized to receive factor was administered by SQ injection (2 mg/m2 TIW)
Tα1 (900 μg/m2 subcutaneously) either by a loading for 1 month to 11 patients who had just completed radio-
dose (daily for 14 days, then twice a week) or on a therapy for a variety of locally advanced solid tumors. A
twice-weekly schedule; treatment began within a week variety of immune parameters were followed serially
after completion of radiotherapy. All patients exhib- and compared to those of five patients randomized not
ited a marked depletion in absolute circulating T-cell to receive treatment. Treatment produced a generalized
levels and in T-cell LPRs at the completion of radiation lymphocytosis involving both Tind and Tc/s cell numbers,
therapy. Serial immunological monitoring revealed but no effects were noted on DTHS or LPRs at the dose
that the patients who received Tα1 had a complete nor- and schedule employed [112].
malization of T-cell function in MLR, which became
apparent following 7 weeks of treatment. However, Retinoids
only patients treated on the twice-weekly schedule Therapeutic and immune restorative effects of vitamin
maintained normal Tind/Tc/s ratios throughout the study A have been evaluated in a randomized trial of 42
period. The Tα1 administration, however, did not influ- patients undergoing radiation therapy for inoperable
ence absolute circulating T-cell numbers or T-cell sub- cervical carcinoma [338]. Vitamin A palmitate was
set numbers, and all treated patients remained administered orally at a daily dose of 1.5 × 106 IU on
lymphocytopenia over the 15-week follow-up period. days 1–5, 8–12, 16–20, and 23–27, and radiotherapy
These results were interpreted to indicate that more began on day 22 for 8 weeks. Vitamin A was well toler-
intensive schedules of Tα1 administration are optimal ated, although most of the 21 treated patients developed
for inducing restoration of T-cell functions, whereas scaling of the skin. Serial immunological assessments
less intensive schedules are optimal for maintaining were performed on some of the patients. No effects were
immune balance of T-cell subsets. found on DTHS reactivity, in that only about 50% of
126 Therapeutic approaches to cancer-associated immune suppression

patients reacted in both the treatment and control groups. (a) the changing and varied combination chemotherapy
Vitamin A treatment increased in vitro LPRs (albeit to a regimens available for clinical use; (b) the lack of well-
low degree) from pretreatment values, whereas the con- defined doses and schedules of administration for the
trol patients showed no change or decrease in LPRs dur- immunorestorative agents; and (c) the almost universal
ing radiation treatment. Thus, it was concluded that omission of detailed serial immunologic assessments
vitamin A administered concurrently with radiation of patients receiving treatment. For the most part, these
could prevent the radiation-induced depression of at studies have emphasized conventional chemotherapy
least one T-cell function. parameters as their end point, that is, tumor regression
rates and overall patient survival, on the assumption
Bestatin that if improvements were noted in patients who
Several studies have explored the in vivo effects of received an immunorestorative agent, they would result
bestatin in irradiated cancer patients [61, 66, 70, 373]. from undefined immunomodulatory effects on the
In a prospective randomized trial, the clinical efficacy immune response.
of bestatin was evaluated in 151 evaluable patients who
had completed a course of local radiotherapy for blad-
Advanced Disease
der cancer. Patients were randomly assigned to receive
10 mg bestatin orally, three times daily, for at least 1 Three broad approaches are feasible for the treatment of
year, or no further treatment. The recovery of LPRs to advanced cancer patients with combined chemotherapy-
PHA and PPD proceeded at an accelerated rate for 9 immunorestorative therapy. The putative immunorestor-
months in the bestatin-treated patients [70]. Patients ative agent could be administered concurrently with
treated with bestatin exhibited a greater percentage of chemotherapy, following the completion of chemotherapy
circulating T cells after 1 month of treatment, but levels when clinical remission has been achieved, or during
declined to control values within 3 months. Similar maintenance chemotherapy if it is continued. All three
transient changes were noted in NK-like activity. It was approaches have been applied, and most studies have
concluded that the schedule or dose of bestatin was not employed levamisole and/or thymic factors.
optimal, and that either higher doses or intermittent
schedules require evaluation. An update of the trial has
Chemicals
shown that the bestatin-treated patients exhibited an
improvement in disease-free survival compared to the Levamisole
radiotherapy-only group, but no improvement in overall This compound has been evaluated in several large-
survival [65]. The disease-free survival benefit was scale clinical trials as an adjunct to conventional che-
more apparent in patients with earlier stages of disease. motherapy, with mixed results. In general, levamisole
has been administered as either 150–200 mg or 2.5 mg/
OK-432 kg on 2 consecutive days each week between chemo-
A large multicenter trial in Japan randomized 382 therapy courses. Serial immunological studies have
patients with cervical cancer, stratified by presence or usually not been performed. In 82 patients with meta-
absence of surgery and clinical stage, to radiotherapy static colorectal cancer, survival of those receiving
with or without intradermal OK-432 starting concur- 5-FU and levamisole was significantly greater than
rently at 2-day intervals with the initiation of radiation those receiving 5-FU alone [78]. Improvements in
treatment [106], and continuing biweekly for 2 years. response rates and/or survival have also been seen in
Patients who received OK-432 exhibited a decrease in patients receiving levamisole in addition to combina-
3-year recurrence rate, a more rapid restoration in DTHS tion chemotherapy for advanced breast cancer [278,
to PHA, a-polysaccharide antigen, and peripheral blood 281, 497]. In one study, an improvement in DNCB
lymphocyte counts than the control patients. reactivity was noted with levamisole treatment; how-
ever, negative reports have also appeared concerning
Combined-modality Studies with breast cancer [99, 387], colorectal cancer [91], non-
small-cell lung cancer [107, 129], and malignant mela-
Chemotherapy
noma [122]. A report in 669 patients with non-small-cell
Three major problems in combined-modality studies lung cancer indicated that patients who received com-
(Table 3) have made it difficult to draw conclusions bination chemotherapy along with levamisole plus
concerning the influence of putative immunorestorative warfarin plus tranexamic acid survived longer than
agents on chemotherapy-induced immunosuppression: those receiving chemotherapy alone [473].
Robert K. Oldham 127

Table 3. Summary of combined modality studies designed to treat chemotherapy-induced immunosuppression


Cancer types Outcome References
Advanced disease
Chemicals
Levamisole Colon + [78]
− [91]
Breast + [278, 281, 497]
− [99, 387]
Non-small-cell lung + [475]
− [107, 129]
Melanoma − [122]
Multiple myeloma + [445]
Acute lymphoblastic leukemia + [390, 391]
Acute myeloblastic leukemia − [441]
Isoprinosine Colon − [119]
Azimexone Breast + [111, 286]
Piroxicam Non-small-cell lung + [82]
Cimetidine Ovarian + [295]
Biologicals
Thymostimulin Gastrointestinal + [478]
Melanoma − [51]
Small-cell lung + [314]
Non-small-cell lung − [132, 312]
Thymosin Fr. 5 Small-cell lung + [118]
− [455, 476]
Non-small-cell lung − [40]
OK-432 Non-small-cell lung + [557]
Lentinan Gastric + [231, 504]
Bestatin Acute nonlymphoblastic leukemia + [379]
Adjuvant treatment
Chemicals
Levamisole Colon + [113, 292, 293]
− [244, 351]
Breast + [279, 280]
− [270]
Gastric + [367]
Ovarian − [274]
Biologicals
Thymostimulin Breast + [241]
Transfer factor Non-small-cell lung + [162]
OK-432 Non-small-cell lung + [557]

Levamisole has also been evaluated as an adjunct to Azimexone has been administered to ten patients
maintenance chemotherapy for a variety of hematologic with breast cancer after remission was induced by
malignancies. Improvements in survival from the start of chemotherapy (CTX, MTX, 5-FU, VCR, predni-
maintenance chemotherapy have been noted in patients sone) to assess whether it could ameliorate the
receiving levamisole in cases of multiple myeloma [445] immunosuppressive effects of treatment. Detailed
and acute lymphoblastic leukemia [390, 391]. Thus, weekly serial immunological assessments, per-
levamisole did appear to have potential as an adjunct to formed while patients were receiving 100-mg weekly
maintenance chemotherapy for patients with hematopoi- intravenous injections, revealed that chemotherapy-
etic cancers; however, no effects were observed when induced immunosuppression was markedly reversed
levamisole was administered concurrently with intensive during azimexone administration. Significant
induction chemotherapy for ANLL [534]. increases in peripheral blood lymphocyte counts and
Isoprinosine has been administered concurrently with in vitro LPR were noted without change in T- or
intravenous 5-FU at various doses without any observ- B-cell percentages [111, 286].
able antitumor effects [119].
128 Therapeutic approaches to cancer-associated immune suppression

Prostaglandin Inhibitors VP-16, cisplatin). An analysis based on pretreatment


It has been demonstrated that the concurrent administra- immune function, total white blood cell count, and abso-
tion of nonsteroidal anti-inflammatory drugs can prevent lute lymphocyte count revealed no difference in survival
some aspects of chemotherapy-induced immune sup- distributions. These results could not confirm the prior
pression [82]. reported study [118]. However, this latter trial differed
from the original study in that it used different chemo-
Cimetidine therapy regimens and included prophylactic chest and
A study of the immunomodulatory effects of cimetidine whole-brain radiotherapy for patients who responded to
was performed in patients with advanced ovarian cancer chemotherapy. Thus, no firm conclusions can be reached
who received concurrent chemotherapy (cisplatin, ADR, regarding the role of TF5 as an adjunct to chemotherapy
CTX) [275]. Treatment with chemotherapy produced a for small-cell lung cancer.
decrease in CD4 cell counts and in IL-2 production of Several different trials in patients with advanced
PBL, which was significantly improved in patients who non-small-cell lung cancer treated with combination che-
received concurrent cimetidine. motherapy with or without TF5 [41] or TP-1 [132, 312]
and in metastatic melanoma patients treated with TP-1
[51] failed to find improvements in survival for patients
Biologicals treated with thymic factors. In one of the lung cancer tri-
Thymic Factors als, only immunosuppressed patients were considered
Two studies performed in the late 1970s with TP-1 and eligible for study; however, no significant immunorestor-
TF5 suggested both a rationale and role for the use of ative effects were noted in the TP-1-treated patients except
thymic factors in conjunction with combination chemo- transient mild improvement in absolute T-cell levels.
therapy [478]. Although overall response rates were not In vitro LPR to mitogens remained suppressed following
altered by TF5, its administration at a high dose (60 mg/ treatment, and no improvements in DTHS were observed.
m2) led to a significant improvement in overall median Thus, follow-up combined modality studies with TF5
survival [116]. Improved survival was limited to patients and TP-1 have not provided evidence that thymic factors
who had exhibited pretreatment depressions of total could either prevent chemotherapy-induced immuno-
T-cell levels below 775/mm3 and serum HS glycopro- suppression or improve survival.
tein levels below 60.5 mg/dl. Although serial immuno-
logical assessments were not performed, it was Lentinan
concluded that, whereas TF5 had no detectable direct In small studies, administration of lentinan in combina-
antitumor effects, its administration to immunosup- tion with tegafur [504] or 5-FU and mitomycin [231] to
pressed patients may have improved survival by ame- patients with advanced gastric cancer has been reported
liorating the immune defects due to the presence of to improve patient survival.
tumor, and exacerbated by the chemotherapy.
These two reports culminated in the performance of a Bestatin
number of confirmatory trials, as well as a variety of In a randomized controlled trial of 101 patients with
other studies in which TF5 and TP-1 were administered ANLL, patients over 50 years of age who received
concurrently with combination chemotherapy for bestatin along with induction chemotherapy exhibited a
patients with solid tumors, generally small-cell or NSCLC better remission duration and survival than the chemo-
[40, 51, 132, 312, 314, 455, 476]. In only one of these therapy-only group [379].
studies did the administration of thymic factors improve
the overall response rate to the chemotherapy and sur- Adjuvant Chemotherapy
vival [314]. Although this trial involved very small patient
numbers, the favorable effects of TP-1 on both myelotox- Adjuvant chemotherapy has become an accepted treat-
icity and survival indicate that further studies should be ment modality for patients with breast and colon cancer.
performed adding TP-1 to conventional chemotherapy. Evidence is also accumulating that postsurgical adju-
In a randomized trial involving 91 of patients with vant chemotherapy for other solid tumors may also
small-cell lung cancer [455, 476], TF5 (60 mg/m2 sub- improve patient survival. The acute and chronic immu-
cutaneously, twice weekly) had no effect on survival nosuppressive effects of contemporary adjuvant chemo-
when combined with chemotherapy, compared to che- therapy regimens have recently been evaluated [315,
motherapy alone (CTX, ADR, VCR alternating with 396, 501].
Robert K. Oldham 129

Chemicals Thymic Factors


In a randomized trial 51 patients received adjuvant CMF
Levamisole chemotherapy with or without TP-1 (50 mg/m2 IM daily
In a randomized trial of 135 postoperative breast can- × 2 weeks, then twice weekly for a minimum of 3
cer patients with positive axillary nodes, patients months) following radical mastectomy for breast cancer
received L-phenylalanine mustard with levamisole [241]. Although details of the patients’ clinical charac-
(150 mg for 3 days every 2 weeks) or placebo for up to teristics were not reported, patients who received TP-1
2 years [270]. No significant effects of levamisole were exhibited a significant decrease in the incidence of
noted, although trends for improved survival were seen infections (mostly cystitis, conjunctivitis, and mucosi-
in postmenopausal patients and in those with four or tis), and an increase in Tind/Tc/s compared to the control
more positive lymph nodes. In other studies involving (no treatment) group. There was also a lower incidence
patients with stage III breast cancer treated by either of myelotoxicity in the TP-1 treated patients. These
radiotherapy or adjuvant chemotherapy (ADR, CTX, results have not been confirmed with a large-scale trial.
VCR), there was a high incidence of toxicity (primar-
ily transient agranulocytosis), requiring discontinua-
tion of levamisole. Follow-up is continuing on this
trial. In a study comparing postoperative radiation Current Status of Therapeutic
therapy and adjuvant chemotherapy (vincristine, adri-
amycin, and cyclophosphamide), levamisole adminis-
Alterations for Cancer-associated
tered concurrently appeared to improve both Immune Suppression
disease-free, as well as overall, survival for patients
The results with levamisole in combination with 5-FU
who received chemotherapy alone or combined radia-
as adjuvant therapy for Dukes C colon cancer have pro-
tion therapy plus chemotherapy [279]. Improvements
vided strong evidence that drugs with immunorestor-
were also noted in various immune parameters in the
ative properties can play a role in the treatment of human
levamisole treatment group [305]. Although this trial
cancer. What has been learned about the reversibility or
involved 150 patients, the multiple randomizations
prevention of cancer-associated immunosuppression?
made it difficult to analyze.
All of the agents discussed in this chapter, at least in
In other recent studies, levamisole was shown to
small phase I and II studies, appear to have the capabil-
improve the survival of resected patients with gastric
ity of improving immune cell numbers and/or functions
cancer treated with MIT and tegafur [367], but there
in immunosuppressed patients. The large-scale phase III
was a deleterious effect of levamisole in a trial of 140
surgical adjuvant trials for the most part have provided
patients with ovarian cancer who received adjuvant che-
only limited information concerning whether or not
motherapy following maximal surgical reduction of
perioperative immunosuppression could be prevented.
tumor [274].
Part of this relates to the fact that it is extremely difficult
– and probably impossible – to perform adequate, qual-
Biologicals ity control assays of immunity in multiple different
institutions participating in the same large-scale trial.
Transfer Factor The mechanism by which levamisole improved survival
The administration of transfer factor, prepared from leu- was not addressed, and it is not clear whether the drug
kocytes from household contacts, has been reported to exerted its effects as a result of immunomodulation or
improve survival for stage I and II patients with resected by other, as yet undefined, mechanisms. Several smaller
non-small-cell lung cancer who were treated further studies in patients with GI cancers suggested that certain
with a variety of adjuvant combination chemotherapy aspects of T-cell immunity (i.e., LPR) could be main-
regimens [162]. tained during the perioperative period by treatment with
levamisole. Since these studies, FA/5-FU and later
OK-432 FOLFOX regimens have become the regimen of choice
In a trial of 311 patients, OK-432 was shown to improve in adjuvant therapy of colon cancer.
survival of patients with resected stage I, II and III non- Studies with levamisole, TF5, Tα1, bestatin, and vita-
small-cell lung cancer when combined with three-drug min A have suggested that various immunorestorative
combination chemotherapy, compared to chemotherapy agents could ameliorate radiotherapy-induced depres-
[557]. sion of T-cell numbers or functions, or accelerate the
130 Therapeutic approaches to cancer-associated immune suppression

reconstitution of immunity following radiotherapy. In single-institution pilot studies to establish the pharma-
no case, however, did treatment totally normalize both cokinetics and immunomodulatory potential of the
T-cell numbers and functions, and so efficacy can be agent, and then in properly stratified, large-scale phase
considered partial at best. In several clinical trials, III randomized trials performed by a cooperative cancer
levamisole has not improved survival in irradiated group to establish clinical efficacy using the optimal
patients with non-small-cell lung cancer. Analysis of the immunomodulatory dose and schedule and an appropri-
trial in postradiotherapy patients with non-small-cell ate patient population.
lung cancer has suggested that Tα1 can improve overall
patient survival when used as an adjunct to radiotherapy. Acknowledgment Dr. Richard Schulof co-authored this
However, the mechanism by which Tα1 improved sur- chapter in the second edition, but died in an automobile
vival is not known. In addition to accelerating the recon- accident during the preparation of the third edition.
stitution of T-cell dependent immunity, it is possible that
thymic factors can protect bone marrow stem cells from
the myelotoxic effects of radiotherapy or chemotherapy
[241]. Such a mechanism has been proposed to explain
References
the lower incidence of myelotoxicity in breast cancer 1. Addison I, Baggage J, Gandossini M, Souhami R. Assessment of
patients who received TP-1 along with adjuvant CMF host defense against infection during chemotherapy of Hodgkin’s
disease. Cancer Chemother Pharmacol 1978; 1:129–133.
chemotherapy [241]. 2. Adler A, Stein JA, Ben-Efraim S. Immunocompetence, immuno-
Because radiation therapy results in a uniform and suppression, and human breast cancer, II. Further evidence of ini-
marked depression of immune cell numbers and func- tial immune impairment by integrated assessment effect of nodal
tions, patients who have received this treatment are ideal involvement (N) and of primary tumor size (T). Cancer 1980;
45:2061–2073.
candidates for studies of the immunorestorative effects
3. Adler A, Stein JA, Ben-Efraim S. Immunocompetence, immuno-
of BRMs. However, it must be recognized that if puta- suppression, and human breast cancer. III. Prognostic significance
tive immunorestorative agents are administered concur- of initial level of immunocompetence in early and advanced dis-
rently with radiation, their effects on immunity might be ease. Cancer 1980; 45:2074–2083.
negated to some degree. An agent could exert a benefi- 4. Ahmed K, Turk JL. Effect of anticancer agents neothramycin,
aclacinomycin, FK-565 and FK-156 on the release of interleukin-2
cial effect during radiation treatment only if it provided
and interleukin-1 in vitro. Cancer Immunol Immunother 1989;
a direct protective action on immune cells from the del- 28:87–92.
eterious effects of radiation. 5. Alberola V, Gonzalez-Molina A, Trenor A, et al. Mechanism of
Although a number of large-scale clinical trials have suppression of the depressed lymphocyte response in lung cancer
been performed with patients receiving concurrent che- patients. Allergol Immunopathol (Madr) 1985; 13:213–219.
6. Albin RJ, Gordon MJ, Soane WA. Levels of immunoglobulins in
motherapy, there are only very limited data concerning the serum of patients with carcinoma of the prostate. Neoplasma
the prevention of chemotherapy-induced immunosup- 1972; 19:57–60.
pression. Several studies have suggested that levamisole 7. Alexopoulos LG, Wiltshaw E. Immunological monitoring during
may have a role as a therapeutic adjunct for patients chemotherapy for advanced Hodgkin’s disease. Cancer 1978;
with hematopoietic malignancies who are receiving 42:2631–2640.
8. Alsabti EA. In vivo and in vitro assays of incompetence in bron-
maintenance chemotherapy, yet the mechanism by chogenic carcinoma. Oncology 1979; 36:171–175.
which levamisole exerts its effects remains unknown. 9. Amery WK, Butterworth BS. The dosage regimen of levamisole in
Although none of the immunorestorative agents have cancer: Is it related to efficacy and safety? Int J Immunopharmacol
proven effective when administered concurrently with 1983; 5:1–9.
10. Amery WK, Cosemans J, Gooszen HC, et al. Four-year results
intensive combination chemotherapy for patients with
from double-blind study of adjuvant levamisole treatment in resec-
advanced metastatic cancers, their potential role in table lung cancer. In: Terry WD, Rosenberg SA, eds. Immunotherapy
patients treated with adjuvant chemotherapy remains to of human cancer. New York: Elsevier/North-Holland, 1982:
be explored. 123–133.
The success or failure of future studies with BRMs 11. Amlot PL, Anger A. Binding of phytohaemagglutinin to serum sub-
stances and inhibition of lymphocyte transformation in Hodgkin’s
will be determined primarily, if not exclusively, by the
disease. Clin Exp Immunol 1976; 26:520–527.
selection of suitable phase II and III clinical models for 12. Anderson TC, Jones SE, Sochnlen BJ, et al. Immunocompetence
study and by the avoidance of confounding variables or and malignant lymphomas: Immunologic status before therapy.
inadequate sample sizes, which could lead to uninter- Cancer 1981; 48:2702–2709.
pretable results. It is likely that a two-phased approach 13. Anderson TM, Ibayashi Y, Tokuda Y, et al. Natural killer activity of
lymphocytes infiltrating human lung cancers following preoperative
will be necessary to evaluate BRMs with immunorestor- systemic recombinant interleukin-2. Arch Surg 1987; 122:12;
ative properties: focusing initially on phase I/II, 1446–1450.
Robert K. Oldham 131

14. Anderson TM, Ibayashi Y, Holmes EC, Golub SH. Modification of 34. Baral E, Blomgren H, Petrini B, et al. Prognostic relevance of
natural killer activity of lymphocytes infiltrating human lung can- immunologic variables in breast carcinoma. Acta Radiol Ther
cers. Cancer Immunol Immunother 1987; 25:65–68, Phys Biol 1977; 16:417–426.
15. Anthony HM, Templeman GH, Madsen KE, Mason MK. The 35. Baseler MW, Maxim PE, Veltri RW. Circulating IgA immune
prognostic significance of DHS skin tests in patients with carci- complexes in head and neck cancer, nasopharyngeal carcinoma,
noma of bronchus. Cancer 1974: 34(6) 1901–1906. lung cancer, and colon cancer. Cancer 1987; 59:1727–1731.
16. Anthony HM. Yorkshire trial of adjuvant therapy with levamisole 36. Baskies AM, Chretien PB, Weiss JF, et al. Serum glycoproteins in
in surgically treated lung cancer. In: Terry WD, Rosenberg SA, cancer patients: First report of correlations with in vitro and in vivo
eds. Immunotherapy of human cancer. New York: Elsevier/North- parameters and cellular immunity. Cancer 1980; 45:3050–3060.
Holland, 1982:135–440. 37. Bast RC, Reinherz EL, Maven C, et al. Contrasting effects of
17. Aparicio-Pages NM, Verspaget HW, Pena SA, Lamers CB. cyclophosphamide and prednisolone on the phenotype of human
Impaired local natural killer-cell activity in human colorectal peripheral blood leukocytes. Clin Immunol Immunopathol 1983;
carcinomas. Cancer Immunol Immunother 1989; 28:4; 301–304. 28:101–114.
18. Aparicio-Pages NM, Verspaget HW, Pena SA, Lamers CBHW. 38. Baumann MA, Milson TJ, Patrick CW, et al. Correlation of circu-
Impaired local natural killer cell activity in human colorectal lating natural killer cell count with prognosis in large cell lym-
carcinomas. Cancer Immunol Immunother 1989; 28:301–304. phoma. Cancer 1986; 57:2309–2312.
19. Aranha GV, McKhann CF, Simmons RL, Grage TB. Recall skin 39. Bean MA, Schelhammer PS, Her HW, et al. Immunocompetence
test antigens and the prognosis of stage I melanoma. J Surg Oncol of patients with transitional cell carcinoma as measured by DNCB
1976; 11:13–16. skin tests and in vitro lymphocyte function. Natl Cancer Inst
20. Arinaga S, Akiyoshi T, Tsuji H. Augmentation of the generation Monogr 1978; 49:111–114.
cell mediated cytotoxicity after a single dose of adriamycin in can- 40. Bedikian AY, Patt YZ, Murphy WK, et al. Prospective evaluation
cer patients. Cancer Res 1986; 46:4213–4216. of thymosin fraction V immunotherapy in patients with non-small
21. Arinaga S, Akiyoshi T, Tsuji H. Augmentation of the cell-mediated cell lung cancer receiving vindesine, doxorubicin, and cisplatin
cytotoxic response induced in mixed cell culture by adriamycin. (VAP) chemotherapy. Am J Clin Oncol 1984; 7:399–404.
Jpn J Cancer Res 1985; 76:414–419. 41. Benkovic B, Burek B, Jaksic B, Vitale B. Modulation of chronic
22. Armstrong D, Chmel M. Infectious complications of Hodgkin’s lymphocytic leukemia (CLL) lymphocyte phenotypes by in vitro
disease. In: Lacher MJ, ed. Hodgkin’s disease. New York: Wiley, incubation with alpha-1 thymosin. Blood Cells 1987; 12:2;
1976:267–290. 441–455.
23.Arnaud JP, Buyse M, Nordlinger B, et al. Adjuvant therapy of poor 42. Bennett A, Berstock A, Harris M, et al. Prostaglandins and their
prognosis colon cancer with levamisole: Results of an EORTC dou- relationship to malignant and benign breast tumors. In:
ble-blind randomized clinical trial. Br J Surg 1989; 76:284–289. Sammuelsson B, Ramuell PW, Paoletti R, eds. Advances in pros-
24. Ascher MS, Schneider WJ, Valentine FT, Lawrence HS. In vitro taglandin and thromboxane research vol 6. New York: Raven
properties of leukocyte dialysates containing transfer factor. Proc Press, 1980:595–599.
Natl Acad Sci USA 1974; 74:1178–1182. 43. Berd D, Macguire HC Jr, Mastrangelo MJ. Induction of cell-medi-
25. Ashorn RG, Vandenbard AA, Acott KM, Krohn KJ. Dialysable ated immunity to autologous melanoma cells and regression of
leukocyte extracts (transfer factor) augment non-specifically metastases after treatment with a melanoma cell vaccine preceded
keyhole limpet haemocyanin and horseshoe crab haemocyanin by cyclophosphamide. Cancer Res 1986; 46:2572–2577.
skin reactivity in immunized human recipients. Scand J Immunol 44. Berd D, Mastrangelo MJ, Enstron PF, et al. Augmentation of the
1976; 23:161–167. human immune response by cyclophosphamide. Cancer Res 1982;
26. Balaram P, Remani P, Padmanabhan TK, Vasudevan DM. Role of 42:4862–4866.
levamisole immunotherapy as an adjuvant to radiotherapy in oral 45. Berd D, Mastrangelo MJ. Effect of low dose cyclophosphamide on
cancer: Immune responses. Neoplasma 1988; 35:6; 617–625. the immune system of cancer patients: Reduction of T-suppressor
27. Balaram P, Padmanabhan TK, Vasudevan DM. Role of levamisole function without depletion of the CD8+ subset. Cancer Res 1987;
immunotherapy as an adjuvant to radiotherapy in oral cancer. II. 47:3317–3321.
Lymphocyte subpopulations. Neoplasma 1988; 35:2; 235–242. 46. Berd D, Maguire HC, Mastrangelo MJ. Impairment of concanavalin
28. Balch CM, Doughtery PA, Cloud GA, Tilden AB. Prostaglandin A-inducible suppressor activity following administration to patients
E2-mediated suppression of cellular immunity in colon cancer with advanced cancer. Cancer Res 1984; 44:1275–1280.
patients. Surgery 1984; 95:71–77. 47. Berenyl E, Kavai M, Szabolcsi M, Szegedi G. Levamisole in the
29. Balch CM, Tilden AB, Dougherty PA, Cloud GA. Depressed levels treatment of Hodgkin’s disease. Acta Med Acad Sci Hung 1979;
of granular lymphocytes with natural killer (NK) cell function in 36:177–185.
247 cancer patients. Ann Surg 1983; 198:192–199. 48. Bergmann L, Mitrou PS, Demmer-Dieckmann M, et al. Impaired
30. Balch CM, Dougherty PA, Tilden AB. Excessive prostaglandin E2 T- and B-cell functions in patients with Hodgkin’s disease. Cancer
production by suppressor monocytes in head and neck cancer Immunol Immunother 1987; 25:59–64.
patients. Ann Surg 1982; 196:645–650. 49. Berkarda B, Bouffard-Eyaboglu H, Derman U. The effect of cou-
31. Balm FA, Drexhage HA, von Blomberg M, et al. Mononuclear marin derivatives on the immunological system of man. Agents
phagocytic function in head and neck cancer. Cancer 1984; Actions 1983; 13:50–52.
54:1010–1015. 50. Berlinger NT. Deficient immunity in head and neck cancer due to
32. Bancewicz J, Calman K, MacPherson SG, et al. Adjuvant chemo- excessive monocyte production of prostaglandins. Laryngoscope
therapy and immunotherapy for colorectal cancer; preliminary 1984; 94:11, Pt.1; 1407–1410.
communication. J R Soc Med 1980; 73:197–199. 51. Bernengo MG, Fra P, Lisa F, Meregilli M, Zina G. Thymostimulin
33. Baral E, Blomgren H, Petrini B, Wasserman J. Blood lymphocytes therapy in melanoma patients: Correlation of immunologic effects
in breast cancer patients following radiotherapy and surgery. Int J with clinical course. Clin Immunol Immunopathol 1983;
Radiat Oncol Biol Phys 1977; 2:289–295. 28:311–324.
132 Therapeutic approaches to cancer-associated immune suppression

52. Bernengo MG, Lisa F, Meregalli M, Doveil GC. Changes in T- 70. Blomgren H, Edsmyr F, von Stedingk L-V, Wasserman J. Bestatin
and B-lymphocyte subpopulations before, during and after che- treatment enhances the recovery of radiation-induced impairments
motherapy for malignant melanoma. Int J Tissue React 1984; of the immunological reactivity of the blood lymphocyte popula-
6:505–511. tion in bladder and cancer patients. Biomed Pharmacother 1986;
53. Bicker U. Immunomodulation by 2-cyanaziridine derivatives. In: 40:50–54.
Serrou B, Rosenfeld C, Daniels JC, Saunder JP, eds. Current con- 71. Bobrove AM, Fuks Z, Strober S. Quantitation of T- and
cepts in human immunology and cancer immunomodulation. B-lymphocytes and cellular immune function in Hodgkin’s disease.
Amsterdam: Elsevier Biomedical, 1982:521–534. Cancer 1975; 36:169–179.
54. Bier J, Nicklisch U, Platz H. The doubtful relevance of nonspecific 72. Bockman RS. Prostaglandins in cancer: A review. Cancer Invest
immune reactivity in patients with squamous-cell carcinoma of the 1983; 1:485–493.
head and neck region. Cancer 1983; 52:1165–1172. 73. Boetcher DA, Leonard EJ. Abnormal monocyte chemotactic response
55. Bjorkholm M, Wedelin C, Holm G, et al. Lymphocytotoxic serum in cancer patients. J Natl Cancer Inst 1974; 52:1091–1098.
factors and lymphocyte functions in untreated Hodgkin’s disease. 74. Bollag W, Matta A. From vitamin A to retinoids in experimental
Cancer 1982; 50:2044–2048. and clinical oncology: Achievements, failures, and outlook. Ann
56. Bjorkholm M, Holm G, Mellstedt H. Persisting lymphocyte defi- NY Acad Sci 1981; 359:9–23.
ciencies during remission in Hodgkin’s disease. Clin Exp Immunol 75. Bolton PM, Teasdale C, Mander AM, et al. Immune competence in
1977; 28:389–393. breast cancer: Relationships of pretreatment immunologic tests to
57. Bjorkholm M, Holm G, Mellstedt H. Immunologic profile of diagnosis and tumor stage. Cancer Immunol Immunother 1976;
patients with cured Hodgkin’s disease. Scand J Hematol 1977; 1:251–258.
18:361–368. 76. Bolton PM, Mander AM, Davidson JM, et al. Cellular immunity in
58. Bjorkholm M, Holm G, Mellsted H, Johansson B. Immunodeficiency cancer: Comparison of delayed hypersensitivity skin tests in three
and prognosis in Hodgkin’s disease. Acta Med Scand 1975; common cancers. Br J Med 1975; 3(5974):18–20.
198:275–279. 77. Bone G, Appleton DR, Venebles CW. The prognostic value of the
59. Block JB, Haynes HA, Thompson WL, Neiman PE. Delayed cutaneous delayed hypersensitivity response to 2,4-dinitrochlo-
hypersensitivity in chronic lymphocytic leukemia. J Natl Cancer robenzene in gastrointestinal cancer. Br J Cancer 1974;
Inst 1969; 42:973–980. 29:403–406.
60. Blomgren H, Edsmyr F, Esposti PL, et al. Influence of bestatin, a 78. Borden EC, Davis TE, Crowley JJ, et al. Interim analysis of trial of
new immunomodifier, on human lymphoid cells. In: Serrou B, levamisole and 5-fluorouracil in metastatic colorectal carcinoma.
Rosenfeld C, Daniels JC, Saunders JP, eds. Current concepts in In: Terry WD, Rosenberg SA, eds. Immunotherapy of human can-
human immunology and cancer immunomodulation. Amsterdam: cer. New York: Elsevier/North-Holland, 1982:231–235.
Elsevier Biomedical, 1982:593–610. 79. Bosworth JL, Ghossein NA, Brooks TL. Delayed hypersensitivity
61. Blomgren H, Edsmyr F, Esposti PL, Naslund I. Immunological and in patients treated by curative radiotherapy. Its relation to tumor
haematological monitoring in bladder cancer patients receiving response and short-term survival. Cancer 1975; 36:353–358.
adjuvant bestatin treatment following radiation therapy. A prospec- 80. Braun DP, Harris JE, Rugenstein M. Relationships of arachidonic
tive randomized trial. Biomed Pharmacother 1984; 38:143–149. acid metabolism to indomethacin-sensitive immunoregulatory
62. Blomgren SE, Wolberg WH, Kisken WA. Effect of fluoropyrimi- function and lymphocyte PGE sensitivity in peripheral blood
dines on delayed cutaneous hypersensitivity. Cancer Res 1965; mononuclear cells of disseminated solid-tumor cancer patients. J
25:977–979. Immunopharmacol 1984; 6:227–236.
63. Blomgren H, Wasserman J, Rotstein S, et al. Possible role of pros- 81. Braun DP, Harris JE. Effects of combination chemotherapy on
taglandin-producing monocytes in the depression of mitogenic immunoregulatory cells in peripheral blood of solid-tumor cancer
response of blood lymphocytes following radiation therapy. patients: Correlation with rebound overshoot immune function
Radiother Oncol 1984; 1:255–261. recovery. Clin Immunol Immunopathol 1981; 20:193–214.
64. Blomgren H, Baral E, Edsmyr F, et al. Natural killer activity in 82. Braun DP, Harris JE. Modifications of the effects of cytotoxic
peripheral blood lymphocyte populations following local radiation chemotherapy on the immune responses of cancer patients with a
therapy. Acta Radiol Oncol 1980; 2:139–143. non-steroidal, anti-inflammatory drug, piroxicam. (Abstr.) Proc
65. Blomgren H, Wasserman J, Littbrand B. Blood lymphocytes after Am Soc Clin Oncol 1985; 4:223.
radiation therapy of carcinoma of prostate and urinary bladder. 83. Brenner BG, Friedman G, Margolese RG. The relationship of
Acta Radiol Ther Phys Biol 1974; 13:357–367. clinical status and therapeutic modality to natural killer cell activity
66. Blomgren H, Naslund I, Esposti PL, et al. Adjuvant bestatin immu- in human breast cancer. Cancer 1985; 56:1543–1548.
notherapy in patients with transitional cell carcinoma of the bladder. 84. Brooks WH, Caldwell HD, Mortara RH. Immune responses in
Clinical results of a randomized trial. Cancer Immunol Immunother patients with gliomas. Surg Neurol 1974; 2:419–423.
1987; 25:1; 46. 85. Brosman S, Elhilali M, Vescura C, Fahey J. Immune response in
67. Blomgren H, Strender LE, Petrini B, Wasserman J. Changes of the bladder cancer patients. J Urol 1979; 121:162–169.
spontaneous cytotoxicity of the blood lymphocyte population fol- 86. Brosman S, Hausman M, Shacks SJ. Studies on the immune status
lowing local radiation therapy for breast cancer. Eur J Cancer Clin of patients with renal adenocarcinoma. J Urol 1975; 114:375–380.
Oncol 1982; 18:637–643. 87. Browder JP, Cretien PB. Immune reactivity in head and neck
68. Blomgren H, Wasserman J, Baral E, Petrini B. Evidence for the squamous carcinoma and relevance to the design of immunotherapy
appearance of non-specific suppressor cells in the blood after local trials. Sem Oncol 1977; 4:4; 431–439.
irradiation therapy. Int J Radiol Oncol Biol Phys 1978; 88. Brown AE, Badger AM, Poste G. The effects of cimetidine on
4:249–253. immune cell function and host response to tumors. In: Serrou B,
69. Blomgren H, Strender LE, Edsmyr F. Bestatin treatment and the Rosenfeld C, Daniels JC, and Sanders JP, eds. Current concepts of
peripheral lymphocyte population in cancer patients. Recent human immunology and cancer immunomodulation. Amsterdam:
Results Cancer Res 1980; 75:133–138. Elsevier Biomedical, 1982:513–519.
Robert K. Oldham 133

89. Bruckner HW, Mokyr MB, Mitchell MS. Effect of imidazole-4- 107. Chahinian AP, Goldberg J, Holland JF, et al. Chemotherapy vs.
carboxamide-5-(3,3-dimethyl-1-triazeno) on immunity in patients Chemoimmunotherapy with levamisole or Corynebacterium parvum
with malignant melanoma. Cancer Res 1974; 34:181–183. in advanced lung cancer. Cancer Treat Rep 1982; 66:1291–1297.
90. Bukowski RM, Deodhar S, Hewlett JS, Greenstreet R. 108. Cheema AR, Hersh EM. Patient survival after chemotherapy and
Randomized controlled trial of transfer factor in stage II malig- its relationship to in vitro lymphocyte blastogenesis. Cancer
nant melanoma. Cancer 1983; 51:269–272. 1971; 28:851–855.
91. Buroker TR, Moertel CG, Fleming TR, et al. A controlled evalu- 109. Chihara G, Hamuro J, Maeda YY, et al. Antitumor and metastasis-
ation of recent approaches to biochemical modulation or enhance- inhibitory activities of lentinan as an immunomodulator: An
ment of 5-fluorouracil therapy in colorectal carcinoma. J Clin overview. Cancer Detect Prev Suppl 1987; 1:423–443.
Oncol 1985; 3:1624–1631. 110. Chirhara G. The antitumor polysaccharide lentinan: An overview.
92. Burtin C, Noirot C, Scheinmann P, et al. Clinical improvement in In: Aoki T, Urushizaki I, and Tsubara E, eds. Manipulation of
advanced cancer disease after treatment combining histamine host defense mechanisms. Excerpta Medica 27(9) 1981:1–16.
and H2-antihistaminics (rantidine or cimetidine). Eur J Cancer 111. Chirigos MA, Mastrangelo MJ. Immunorestoration by chemi-
Clin Oncol 1988; 24:2; 161–167. cals. In: Milhich E, ed. Immunological approaches to cancer
93. Busutti L, Blotta A, Mastrorilli M, et al. Transfer factor adjuvant therapeutics. New York: Wiley, 1982:191–239.
therapy in non-small-cell lung carcinoma (NSCLC) after surgery 112. Chisesi T, Capnist G, Rancan L, et al. The effect of thymic sub-
and radiotherapy. J Exp Pathol 1987; 3:4; 565–568. stances on T-circulating cells of patients treated for Hodgkin’s
94. Butterworth C, Oon CJ, Westbury G, Hobbs JR. T-lymphocyte disease. J Biol Regul Homeost Agents 1988; 2:4,193–198.
response in patients with malignant melanoma. Eur J Cancer 113. Chlebowski RT, Nystrom S, Reynolds R, et al. Long-term survival
1977; 10:639–646. following levamisole or placebo adjuvant treatment of colorectal
95. Cairo MS, Mallett C, Vande Ven C, et al. Impaired in vitro poly- cancer: A Western Cancer Study Group trial. Oncology 1988;
morphonuclear function secondary to the chemotherapeutic 45:3:141–143.
effects of vincristine, adriamycin, cyclophosphamide, and actino- 114. Clemens LE, Siiteri PK, Stities DP. Mechanism of immunosup-
mycin D. J Clin Oncol 1986; 5:798–804. pression of progesterone of natural lymphocyte activation during
96. Cameron DJ, O’Brien P, Majeski JA. Effects of in vivo treatment pregnancy. J Immunol 1979; 122:5; 1978–1985.
with ibuprofen on macrophage function in breast and colon cancer 115. Cochran AJ, Mackie RM, Grant RM, et al. An examination of the
patients. J Clin Lab Immunol 1986; 20:1; 23–28. immunology of cancer patients. Int J Cancer 1976; 18:298–309.
97. Cannon GB, Dean JH, Herberman RB, et al. Association of 116. Cohen MH, Chretien PB, Johnston-Early A, et al. Thymosin frac-
depressed postoperative lymphoproliferative responses to alloan- tion V prolongs survival of intensively treated small-cell lung
tigens with poor prognosis in patients with stage I lung cancer. Int cancer patients. In: Terry WD, Rosenberg SA, eds. Immunotherapy
J Cancer 1980; 25:9–17. of human cancer. New York: Elsevier/North-Holland,
98. Carmignani G, Belgrano E, Puppo P, Cornaglia P. T-lymphocyte- 1982:141–145.
inhibiting factors in renal cancer: Evaluation of differences 117. Cohen JHD, Danel L, Cordier G, et al. Sex steroid receptors in
between arterial and renal venous blood levels of E-rosette- peripheral T-cell: Absence of androgen receptors to OKT8-
forming cells. J Urol 1978; 120:673–675. positive cells. J Immunol 1983; 131:2767–2771.
99. Carpenter JT Jr, Smalley RV, Raney M, et al. Ineffectiveness of 118. Cohen MH, Chretien PB, Indo DC, et al. Thymosin fraction V
levamisole in prolonging remission or survival of women and intensive combination chemotherapy prolonging the survival
treated with cyclophosphamide, doxorubicin, and 5-fluorouracil of patients with small-cell lung cancer. JAMA 1979;
for good-risk metastatic breast carcinoma: A Southeastern 241:1813–1815.
Cancer Study Group Trial. Cancer Treat Rep 1986; 70:9; 119. Colozza M, Tonato M, Belsanti V, et al. 5-Fluorouracil and iso-
1073–1079. prinosine in the treatment of advanced colorectal cancer: A lim-
100. Case DC, Hansen JA, Corrales E, et al. Depressed in vitro lym- ited phase I, II evaluation. Cancer 1988; 62:1049–1052.
phocyte responses to PHA in patients with Hodgkin’s disease in 120. Cone L, Uhr JW. Immunological deficiency disorders associated
continuous long remissions. Blood 1977; 49:771–778. with chronic lymphocyte leukemia and multiple myeloma. J Clin
101. Catalona WJ, Chretien PB. Abnormalities of quantitative dinitro- Invest 1964; 43:2241–2249.
chlorobenzine sensitization in cancer patients: Correlation with 121. Consolini R, Cei B, Cini P, et al. Circulating thymic hormone activity
tumor stage and histology. Cancer 1973; 31:353–356. in young cancer patients. Clin Exp Immunol 1986; 66:1; 173–180.
102. Catalona WJ, Sample WF, Chretien PB. Lymphocyte reactivity in 122. Costanzi JJ, Fletcher WS, Balcerzak SP, et al. Combination che-
cancer patients: Correlation with tumor histology and clinical motherapy plus levamisole in the treatment of disseminated
stage. Cancer 1973; 31:65–71. malignant melanoma. A Southwest Oncology Group Study.
103. Catalona WJ, Tarpley JL, Potvin C, Chretien PB. Correlations Cancer 1984; 53:833–836.
among cutaneous reactivity to DNCB, PHA-induced lymphocyte 123. Cozzolino F, Torcia M, Carossino AM, et al. Characterization of
blastogenesis and peripheral blood E-rosettes. Clin Exp Immunol cells from invaded lymph nodes in patients with solid tumors.
1975; 19:327–332. J Exp Med 1987; 166:303–318.
104. Catalona WJ, Ratliff TL, McCool R. Concanavalin-A-activated 124. Cullen BF, Van Belle G. Lymphocyte transformation and changes
suppressor cell activity in peripheral blood lymphocytes of urologic in leukocyte count. Effect of anesthesia and operation.
cancer patients. J Natl Cancer Inst 1980; 65:553–557. Anesthesiology 1975; 43:563–569.
105. Caulfield MJ, Luce KJ, Proffitt MR, Cevy J. Induction of idiotype- 125. Dagan S, Tzehoval E, Fridkin M, Feldman M. Tuftsin and tuftsin
specific suppressor T-cells with antigen/antibody complexes. J Ex conjugates potentiate immunogenic processes: Effects and pos-
Med 1983; 157:1713–1725. sible mechanisms. J Biol Response Mod 1987; 6:6; 625–636.
106. Cervical Cancer Immunotherapy Study Group. Immunotherapy 126. Danel L, Sonweine G, Monier JC, Saez S. Specific estrogen bind-
using the streptococcal preparation OK-432 for the treatment of ing sites in human lymphoid cells and thymic cells. J Steroid
uterine cervical cancer. Cancer 1987; 60:2394–2402. Biochem 1983; 18:5; 559–563.
134 Therapeutic approaches to cancer-associated immune suppression

127. Danish Breast Cancer Cooperative Group. Increased breast cancer 146. Eilber FR, Morton DL. Impaired immunologic reactivity and
recurrence rate after adjuvant therapy with levamisole. Lancet recurrence following cancer surgery. Cancer 1970; 25:362–367.
1980; 2:824–827. 147. Eilber FR, Morton DL, Ketcham AS. Immunologic abnormalities
128. Das SN, Khanna NN, Khanna S. A multiparametric observation of in head and neck cancer. Am J Surg 1974; 128:534–538.
immune competence in breast cancer and its correlation with tumor 148. Eisenthal A, Monselise J, Zinger R, Adler A. The effect of cime-
load and prognosis. Ann Acad Med Singapore 1985; 14:374–381. tidine on PBL from healthy donors and melanoma patients:
129. Davis S, Mietlowski W, Rohweder JJ, et al. Levamisole as an Augmentation of T-cell responses to TCGF mitogens and alloan-
adjuvant to chemotherapy in extensive bronchogenic carcinoma. tigens and of TCGF production. Cancer Immunol Immunother
A Veterans Administration Lung Cancer Group study. Cancer 1986; 21:2; 141–147.
1982; 50:646–651. 149. Ershler WB, Hacker MP, Burroughs BJ, et al. Cimetidine and the
130. Dawson DE, Everts EC, Vetto RM, Burger DR. Assessment of immune response. In vivo augmentation of nonspecific and specific
immunocompetent cells in patients with head and neck squamous- immune response. Clin Immunol Immunopathol 1983; 26:10–17.
cell carcinoma. Ann Otol Rhinol Laryngol 1985; 94:342–345. 150. Estervez ME, Ballart IJ, Diez RA, et al. Normalization of mono-
131. DeBoer KP, Braun DP, Harris JE. Natural cytotoxicity and anti- cyte candidacidal deficiency by cyclooxygenase inhibitors in
body-dependent cytotoxicity in solid-tumor cancer patients: Hodgkin’s disease. Cancer 1985; 55:2774–2778.
Regulation by adherent cells. Clin Immunol Immunopathol 1982; 151. Eura M, Maehara T, Tsutomo I, Ishikawa T. Suppressor cells in
23:133–144. the effector phase of autologous cytotoxic reactions in cancer
132. Del Giacco GS, Cengiarotti L, Mantovani G, et al. Advanced patients. Cancer Immunol Immunother 1988; 27:147–153.
lung cancer treated with combination chemotherapy with or with- 152. Ezdinli EZ, Simonson KL, Smith RA. Comparison of the effects
out thymostimulin. In: Byrom NA, Hobbs JR, eds. Thymic factor of single vs. multiple agent chemotherapy on lymphocytes
therapy Serono Symposium Publications. New York: Raven assayed by the rosette techniques. Cancer 1978; 42:2234–2243.
Press, 1982:321–328. 153. Faguet GB, Davis HC. Survival in Hodgkin’s disease: The role of
133. Dellon AL, Potvin C, Chretien PB. Thymus-dependent lympho- immunocompetence and other major risk factors. Blood 1982;
cyte levels during radiation therapy for bronchogenic and esoph- 59:938–945.
ageal carcinoma: Correlations with clinical course in responders 154. Fisher RI, Bostick-Bruton F. Depressed T-cell proliferative
and nonresponders. Am J Roentgenol Radiat Ther Nucl Med responses in Hodgkin’s disease: Role of monocyte-mediated sup-
1975; 123:500–511. pression via prostaglandins and hydrogen peroxide. J Immunol
134. Dillman RO, Beauregard JC, Zavanelli MI, et al. In vivo immune 1982; 129:1770–1774.
restoration in advanced cancer patients after administration of 155. Fisherman EW. Does the allergic diathesis influence malignancy?
thymosin fraction V or thymosin alpha-1. J Biol Response Mod J Allergy 1960; 31:74–78.
1983; 2:139–147. 156. Flodgren P, Sjogren HO. Influence in vitro on NK and K-cell
135. Dimitrov NV, Arnold D, Munson J, et al. Phase II study of thy- activities by cimetidine and indomethacin with and without
mosin fraction 5 in the treatment of metastatic renal cell carcinoma. simultaneous exposure to interferon. Cancer Immunol
Cancer Treat Rep 1985; 69:137–138. Immunother 1985; 19:28–34.
136. Dizon QS, Southam CM. Abnormal cellular response in skin 157. Florentin I, Kraus L, Mathé G, Hadden JW. In vivo study in mice
abrasion in cancer patients. Cancer 1963; 16:1288–1292. of the immunopharmacological properties of NPT-15392. In:
137. Domellof L. Athlin L, Berghem L. Effects of long-term combina- Serrou B, Rosenfeld C, Daniels JB, Saunders JP, eds. Current
tion chemotherapy on the reticuloendothelial system. Cancer concepts of human immunology and cancer. Amsterdam: Elsevier
1984; 53:2073–2078. Biomedical, 1982:463–469.
138. Donoran AJ. Reticuloendothelial function in patients with cancer: 158. Forbes JT, Greco FA, Oldham RK. Human natural cell-mediated
Initial observations. Am J Surg 1967; 111:230–238. cytotoxicity: II. Levels in neoplastic disease. Cancer Immunol
139. Droller MJ, Schneider MU, Perlmann P. A possible role of pros- Immunother 1982; 11:147–153.
taglandins in the inhibition of natural and antibody-dependent 159. Ford RJ, Tsao J, Kouttab NM, et al. Association of an interleukin
cell-mediated cytotoxicity against tumor cells. Cell Immunol abnormality with the T-cell defect in Hodgkin’s disease. Blood
1978; 39:165–177. 1984; 64:386–392.
140. Droller MJ, Perlmann P, Schneider MU. Enhancement of natural 160. Fracchia A, Pacetti M, Barberis M, et al. Determination of
and antibody-dependent lymphocyte cytotoxicity by drugs which T-lymphocyte subpopulations in patients with lung cancer. A com-
inhibit prostaglandin production by tumor target cells. Cell parison between lung lavage and peripheral blood by monoclonal
Immunol 1978; 39:154–164. antibodies and flow cytometry. Respiration 1987; 51:3; 161–169.
141. Ducos J, Mingueres J, Colombio P, et al. Lymphocyte response to 161. Frydecka I. Natural killer-cell activity during the course of disease
PHA in patients with lung cancer. Lancet 1970; 1:111–112. in patients with Hodgkin’s disease. Cancer 1985; 56:2799–2803.
142. Dupont B, Ballon M, Hansen JA, et al. Effects of transfer factor 162. Fujisama T, Yamaguchi Y, Kimura H, et al. Adjuvant immuno-
therapy on mixed lymphocyte culture reactivity. Proc Natl Acad therapy of primary resected lung cancer with transfer factor.
Sci USA 1974; 71:867–871. Cancer 1984; 54:663–669.
143. Edelman GM, Cunningham BA, Gall WE, et al. The covalent 163. Fujisawa T, Yamaguchi Y, Kimura H. Transfer factor in restoration
structure of and entire gamma G immunoglobulin molecule. Proc and cell-mediated immunity in lung cancer patients. Jpn J Surg
Natl Acad Sci USA 1969; 63:78–85. 1983; 13:304–311.
144. Ehrke MJ, Mihich E, Berd D, Mastrangelo MJ. Effects of anti- 164. Fuks Z, Strober S, Kaplan HS. Interaction between serum factors
cancer drugs on the immune system in humans. Sem Oncol 1989; and T-lymphocytes in Hodgkin’s disease. N Eng Med 1976;
16:3; 230–253. 295:1273–1278.
145. Eilber FR, Nizze HA, Morton DL. Sequential evaluation of gen- 165. Fuks Z, Strober W, Bobrove AM, et al. Long-term effects of
eral immune competence in cancer patients: Correlation with radiation on T- and B-lymphocytes in peripheral blood of patients
clinical course. Cancer 1975; 35:660–665. with Hodgkin’s disease. J Clin Invest 1976; 58:803–814.
Robert K. Oldham 135

166. Garay GE, Pavlovsky S, Sasiain MC, et al. Immunocompetence 186. Hadden JW. Chemically-defined immunotherapeutic drugs under
and prognosis in children with acute lymphoblastic leukemia: development for the treatment of cancer. In: Serrou B, Rosenfeld
Combination of two different maintenance therapies. Med Pediatr C, Daniels JC, and Saunders JP, eds. Current concepts in human
Oncol 1976; 2:403–415. immunology and cancer immunomodulation. Amsterdam:
167. Gatti RA, Good RA. Occurrence of malignancy in immunodefi- Elsevier Biomedical, 1982:439–448.
ciency disease: A literature review. Cancer 1971; 28:89–98. 187. Hadden JW. Recent advances in the preclinical and clinical immu-
168. Geffner JR, Serebrinsky G, Isturiz MA. Normal human serum nopharmacology of interleukin-2: emphasis on IL-2 as an immu-
restores the expression of Fc8 receptors in immune complex: Blocked norestorative agent. Cancer Detect Prev 1988; 12:537–552.
human mononuclear cells. Immunology 1986; 59:239–243. 188. Hadjipetrou-Kourounakis L, Manikou H, Tsougranis A. Restoration
169. Giulivi A, Cilano L, Roncoroni L, et al. Effects of cimetidine on of immunosuppression in lung cancer by normal sera. J Clin Lab
in vitro transformation of peripheral monocytes to macrophages Immunol 1985; 16:149–153.
in healthy volunteers and cancer patients. Int J Immunopharmacol 189. Haim N, Meidav A, Samuelly B, et al. Prostaglandin-related and
1986; 8:5; 517–523. adherent cell suppressor system in apparently cured Hodgkin’s
170. Goldenberg GJ, Brandes LJ, Lau WH, et al. Cooperative trial of disease patients. J Biol Response Mod 1984; 3:219–225.
immunotherapy for nasopharyngeal carcinoma with transfer factor 190. Hakim AA. Peripheral blood lymphocytes from patients with can-
from donors with Epstein-Barr virus antibody activities. Cancer cer lack interleukin-2 receptors. Cancer 1988; 61:4; 689–701.
Treat Rep 1985; 69:761–767. 191. Halili M, Bosworth J, Romney S, et al. The long-term effect of
171. Goldrosen MH, Han T, Jung D, et al. Impaired lymphocyte blas- radiotherapy on the immune status of patients cured of a gyneco-
togenic response in patients with colon adenocarcinoma: Effects logic malignancy. Cancer 1976; 37:2875–2878.
of disease and age. J Surg Oncol 1977; 9:229–234. 192. Hall HRS, Goldstein AL. The endocrine thymus. In: Becker KC,
172. Goldsmith HS, Levin AG, and Southam CM. A study of cellular ed. Principles and practice of endocrinology and metabolism.
responses in cancer patients by quantitative and qualitative New York: Lippincott, 1990.
Rebuck tests. Surg Forum 1965; 16:102–104. 193. Han T, Takita H. Indomethacin-mediated enhancement of lym-
173. Goldstein AL, Low TLK, Thurman GB, et al. Thymosins and phocyte response to mitogens in healthy subjects and lung cancer
other hormone-like factors of the thymus gland. In: Mihich E, ed. patients. Cancer 1980; 46:2416–2420.
Immunological cancer therapeutics. New York: Wiley, 1982: 194. Han T, Nemoto T, Ledesma EJ, Bruno S. Enhancement of
137–190. T-lymphocyte response to mitogens by indomethacin in breast and
174. Golub SH, Rangel DM, Morton D. In vitro assessment of immu- colorectal cancer patients. Int J Immunopharmacol 1983; 5:11–15.
nocompetence in patients with malignant melanoma. Int J Cancer 195. Herr HW. Adherent suppressor cells in the blood of patients with
1977; 20:873–880. bladder cancer. J Urol 1981; 126:457–460.
175. Goodwin JS, Messner RP, Bankhurst AD, et al. Prostaglandin- 196. Hakim AA. Lipid-like agent from human neoplastic cells sup-
producing suppressor cells in Hodgkin’s disease. N Engl J Med presses cell-mediated immunity. Cancer Immunol Immunother
1977; 297:963–968. 1980; 8:1330.
176. Gottlieb AA, Farmer JL, Matzura CT, et al. Modulation of human 197. Han T, Takita H. Immunologic impairment in bronchogenic car-
T-cell production of migration inhibitory lymphocytes by cytok- cinoma: A study of lymphocyte response to phytohemagglutinin.
ines derived from human leukocyte dialysates. J Immunol 1984; Cancer 1972; 30:616–620.
132:1; 256–260. 198. Hancock BW, Bruce L, Sokol RJ, Clark A. Transfer factor in
177. Green AA, Borella L. Immunologic rebound after cessation of Hodgkin’s disease: A randomized clinical and immunological
long-term chemotherapy in acute leukemia. II. In vitro response study. Eur J Cancer Clin Oncol 1988; 24:5; 929–933.
to phytohemagglutinin and antigens by peripheral blood and 199. Harris J, Copeland D. Impaired imunoresponsiveness in tumor
bone marrow lymphocytes. Blood 1973; 42:99–110. patients. Ann NY Acad Sci 1974; 230:56–85.
178. Greenwald RJ, et al. The B7 family revisited. Annu Rev Immunol 200. Harris JE, DeBoer KP, Vahey AC, Braun DP. The measurement
2005; 23:515–548. of leukocyte subsets in the peripheral blood of cancer patients
179. Groveman DS, Borden EC. In vitro and in vivo effects of levam- using monoclonal antibody reagents. Med Pediatr Oncol 1982;
isole on monocyte chemotaxis in normal donors and patients 10:185–194.
with colorectal carcinoma. J Biol Response Mod 1983; 2: 201. Harris JE, Stewart THM. Recovery of mixed lymphocyte reactivity
167–174. (MLR) following cancer chemotherapy in man. In: Schwartz
180. Grzelak I, Olszewski WL, Engeset A. Decreased suppressor cell MR, ed. Proceedings of the Sixth Leukocytes Culture Conference.
activity after surgery. J Clin Lab Immunol 1985; 16:201–205. New York: Academic Press, 1979:555–580.
181. Grzelak I, Olszewski WL, Engeset A. Suppressor cell activity in 202. Haskell CM. Immunologic aspects of cancer chemotherapy. Ann
peripheral blood in cancer patients after surgery. Clin Exp Rev Pharmacol Toxicol 1977; 17:179–195.
Immunol 1983; 51:149–156. 203. Hausman MS, Brossman SA, Snyderman R, et al. Defective
182. Guiliano AE, Rangel D, Golub SH, et al. Serum-mediated immu- monocyte function in patients with genitourinary carcinoma.
nosuppression in lung cancer. Cancer 1979; 43:917–924. J Natl Cancer Inst 1975; 55:1047–1054.
183. Gupta S, Cunningham-Rundles S. Deficient concanavalin-A- 204. Hayashi Y, Nishida T, Yoshida H, et al. Peripheral T-gamma lym-
induced suppressor cell activity in women with untreated breast phocyte population in head and neck cancer. Cancer Immunol
cancer. J Clin Lab Immunol 1982; 9:159–161. Immunother 1984; 17:160–164.
184. Gupta S, Good RA. Subpopulations of human T-lymphocytes: II. 205. Heal JM, Chuang C, Blumberg N. Perioperative blood transfu-
Effect of thymopoietin, corticosteroids, and irradiation. Cell sions and prostate cancer recurrence and survival. Am J Surg
Immunol 1977; 34:10–18. 1988; 156:374–379.
185. Haas GS, Halperin E, Doseretz D, et al. Differential recovery of 206. Hersey P, Bindon C, Czerniecki M, et al. Inhibition of interleukin-2
circulating T-cell subsets after nodal irradiation for Hodgkin’s production by factors released from tumor cells. J Immunol
disease. J Immunol 1984; 132:1026–1030. 131:2837–2842.
136 Therapeutic approaches to cancer-associated immune suppression

207. Hersh EM, Gutterman JU, Mavligit GM, Reed RC. Immunologic 230. Hoon DS, Irie RF, Cochran AJ. Gangliosides from human mela-
aspects of chemotherapy. In: Cancer Chemotherapy-Fundamental noma immunomodulate responses of T-cells to interleukin-2.
Concepts and Recent Advances. Chicago, Illinois: Yearbook Cell Immunol 1988; 111:2; 410–419.
Medical, 1975: 279–294. 231. Horiguchi M, Saito A, Sonoda H. A gastric cancer showing
208. Hersh EM. Vinca alkaloids. In: Sartorelli AC, Johns DG, eds. marked improvement (stage IV) with lentinan immunotherapy
Antineoplastic and immunosuppressive agents, part I. New York: and 5-FU, MMC-chemotherapy. Gan To Kagaku Ryoho 1988;
Springer Verlag, 1974:601–602. 15:6; 1973–1977.
209. Hersh EM, Gutterman JU, Mavligit GM. Immunotherapy of 232. Horrino N. Impaired PWM-induced polyclonal B-cell activation
leukemia. Med Clin North Am 1976; 60:1019–1042. in patients with malignancies treated with various intermittent
210. Hersh EM, Gutterman JU, Mavligit GM, et al. Serial studies of combination chemotherapies including doxorubicin. Cancer
immunocompetence of patients undergoing chemotherapy for 1982; 50:659–667.
acute leukemia. J Clin Invest 1974; 54:401–408. 233. Hortobagyi GN, Hersh EM, Papadopoulos EJ, et al. Initial clini-
211. Hersh EM, Whitecan JP Jr, McCredie JB, et al. Chemotherapy, cal studies with copovithane. J Biol Response Mod 1986;
immunocompetence, immunosuppression and prognosis in acute 5:319–329.
leukemia. N Engl J med 1971; 285:1211–1216. 234. Hsu CC, Logerfo P. Correlation between serum alpha globulin and
212. Hersh EM, Irwin WS. Blastogenic responses of lymphocytes plasma inhibitory effect of PHA-stimulated lymphocytes in colon
from patients with untreated and treated lymphomas. Lymphology cancer patients. Proc Soc Exp Biol Med 1972; 139:575–578.
1969; 2:150–160. 235. Huang AT, Mold NG, Fisher Sr, et al. A prospective study of
213. Hersh EM. Immunosuppressive agents. In: Sartorelli AC, Johns squamous head and neck carcinoma. Cancer 1987; 59:1721–1726.
DG, eds. Antineoplastic and immunosuppressive agents, part I. 236. Hubbard GW, Wanebo H, Fukuda M, Pace R. Defective suppressor-
New York: Springer Verlag, 1974:555–576. cell activity in cancer patients: A defect in immune regulation.
214. Hersh EM, Oppenheim J. Inhibition of in vitro lymphocyte transfor- Cancer 1981; 47:2177–2184.
mation during chemotherapy in man. Cancer Res 1979; 27:98–105. 237. Ninneman JL. Melanoma-associated immunosuppression through
215. Herskovic A, Bauer M, Seydel G, et al. Postoperative thoracic B-cell activation of suppressor T-cells. J Immunol 1978;
irradiation with or without levamisole in non-small-cell lung can- 120:1573–1579.
cer: Results of a radiation therapy oncology group study. Int J 238. Hughes NR. Serum concentrations of gamma G, gamma A and
Radiation Oncology Biol Phys 1988; 14:1; 37–42. gamma M immunoglobulins in patients with carcinoma, mela-
216. Hilgard P, Pohl J, Stekar J, Voegeli R. Oxazaphosphorines as bio- noma, and sarcoma. J Natl Cancer Inst 1971; 46:1015–1028.
logical response modifiers: Experimental and clinical perspective. 239. Humphrey LJ, Humphrey MA, Singla O, Volenec FJ. Immunologic
Cancer Treat Rev 1985; 12:155–162. responsiveness of patients with cancer: Relationship to tumor
217. Hillinger SM, Herzig GP. Impaired cell-mediated immunity in type, stage and prognosis. Ann Surg 1981; 193:574–578.
Hodgkin’s disease mediated by suppressor lymphocytes and 240. Hyman GA. Increased incidence of neoplasia in association with
monocytes. J Clin Invest 1978; 61:1620–1627. chronic lymphocytic leukemia. Scand J Haematol 1969;
218. Hiraoka N, et al. Prevalence of FOXP3 regulatory T cells increases 6:99–104.
during progression of pancreatic ductal adenocarcinoma and its 241. Iaffaioli RV, Frasci G, Tortora G, et al. Effect of thymic extract
premalignant lesion. Clin Cancer Res 2006; 12:5423–5434 ‘thymostimulin’ on the incidence of infections and myelotoxicity
219. Hirsh B, Johnson JT, Rabin BS, Thearle PB. Immunostimulation during adjuvant chemotherapy for breast cancer. Thymus 1988;
of patients with head and neck cancer. In vitro and preliminary 12:69–75.
clinical experience. Arch Otolaryngol 1983; 109:298–301. 242. Ichimura O, Suzuki S, Saito M, et al. Augmentation of interleukin-1
220. Hirshaut Y, Pinsky CM, Frydecka I, et al. Effect of short-term and interleukin-2 production by OK-432. Int J Immunopharmacol
levamisole therapy on delayed hypersensitivity. Cancer 1980; 1985; 7:263–270.
45:362–366. 243. Inagaki T, Morise K, Matsunaga H. Effects of endoscopic intratu-
221. Hoagland JG, Scoggin S, Giavazzi R, et al. Tumor-derived moral injection of lentinan in patients with gastric cancer. Gan To
suppressor factors (TDSFs) in normal and neoplastic colon and Kagaku Ryoho 1988; 15:2; 319–324.
rectum. J Surg Res 1986; 40:5; 467–474. 244. Intergropu 5-FU/levamisole study positive; untreated arms stopped,
222. Hodgson WJB, Lowenfels AB. Blood transfusion and recurrence clinical alert sent. Clin Trials – Clin Cancer Lett 1989; October.
rates in colonic malignancy. Lancet 1982; 2:1047. 245. Israel L, Mugica J, Chahinian PH. Prognosis of early broncho-
223. Hojo H, Hashimoto Y. Cytotoxic cells induced in tumor-bearing rats genic carcinoma: Survival cures of 451 patients after resection of
by a streptococcal preparation (OK-432). Gann 1981; 72:62–69. lung cancer in relation to the results of preoperative tuberculin
224. Holm G, Mellstedt H, Bjorkholm M, et al. Lymphocyte abnor- skin test. Biomed Exp 1973; 19:68–72.
malities in untreated patients with Hodgkin’s disease. Cancer 246. Itoh K, Pellis NR, Balch C. Monocyte-dependent serum-borne sup-
1976; 37:751–762. pressor of induction of lymphokine-activated killer cells in lympho-
225. Holmes EC. Immunology of tumor-infiltrating lymphocytes. Ann cytes from melanoma patients. Cancer Immunol Immunother 1989;
Surg 1985; 201:158–163. 29:57–62.
226. Holmes E, Sibbitt WL, Bankhurst AD. Serum factors which sup- 247. Jacobson DR, Zolla-Pazner S. Immunosuppression and infection
press natural cytotoxicity in cancer patients. Int Arch Allergy in multiple myeloma. Sem Oncol 1986; 13:3; 282–290.
Appl Immunol 1986; 80:39–43. 248. Jansen HM, The TH, deGast GC, et al. The primary immune
227. Holmes EC, Golub SH. Immunologic defects in lung cancer response of patients with different stages of squamous-cell bron-
patients. J Thoracic Cardiovasc Surg 1976; 71:161–168. chial carcinoma. Thorax 1978; 33:755–760.
228. Hoon DSB, Bowker RJ, Cochran AJ. Suppressor-cell activity in 249. Jenkins VK, Ray P, Ellis HN, et al. Lymphocyte response in
melanoma-draining lymph nodes. Cancer Res 1987; 47:1529–1533. patients with head and neck cancer: Effect of clinical stage and
229. Hoon DSB, Korn EL, Cochran AJ. Variations in functional radiotherapy. Arch Otolaryngol 1976; 102:596–600.
immunocompetence of individual tumor-draining lymph nodes 250. Jerrells TR, Dean JH, Richardson GL, et al. Role of suppressor
in humans. Cancer Res 1987; 47:1740–1744. cells in depression of in vitro lymphoproliferative responses of
Robert K. Oldham 137

lung cancer and breast cancer patients. J Natl Cancer Inst 1978; 272. Kenady DE, Chretien PB, Potvin C, et al. Effect of thymosin in vitro
61:1001–1009. on T-cell levels during radiation therapy. Cancer 1972; 39:642–652.
251. Jerrells TR, Dean JH, Herberman RB. Relationship between 273. Kerman RH, Stefani SS. Phytohemagglutinin stimulation of lym-
T-lymphocyte levels and lymphoproliferative responses to mito- phocytes in lung cancer patients. Oncology 1977; 34:10–12.
gens and alloantigens in lung and breast cancer patients. Int J 274. Khoo SK, Whitaker SV, Jones IS, Thomas DA. Levamisole as
Cancer 1978; 21:282–290. adjuvant to chemotherapy of ovarian cancer. Results of a ran-
252. Job G, Pfeundschuh M, Bauer M, et al. The influence of radiation domized trial and 4-year follow-up. Cancer 1984; 54:986–990.
therapy on T-lymphocyte subpopulations defined by monoclonal 275. Kikuchi Y, Kizawa I, Oomori K, et al. Effects of cimetidine on
antibodies. Int J Radiat Oncol Biol Phys 1984; 10:2077–2081. interleukin-2 production by peripheral blood lymphocytes in
253. Joensun H, Nordman E, Toivanen A. Long-term effect of levami- advanced ovarian carcinoma. Eur J Cancer Clin Oncol 1988;
sole on the immune functions in melanoma patients. Strahlenther 24:7; 1185–1190.
Onkol 1986; 162:12; 753–756. 276. Kirsh MM, Orringer MB, McAuliffe S, et al. Transfer factor in
254. Joensun H, Toivanen A, Nordman E. Effect of tamoxifen on the treatment of carcinoma of the lung. Ann Thorac Surg 1984;
immune function. Cancer Treat Rep 1986; 7:3; 381–382. 38:140–145.
255. Johnson JD, Summersgill JT, Raff MJ. The in vitro effects of 277. Klefstrom P, Holsti P, Grohn P, et al. Levamisole in the treatment
methotrexate on the phagocytosis and intracellular killing of of stage II breast cancer. Five-year follow-up of a randomized
Staphylococcus aureus by human neutrophils. Cancer 1986; double-blind study. Cancer 1985; 55:2753–2757.
57:2343–2345. 278. Klefstrom P. Combination of levamisole immunotherapy and
256. Johnston-Early A, Cohen MH, Fossieck BE, et al. Delayed hyper- polychemotherapy in advanced breast cancer. Cancer Treat Rep
sensitivity skin testing as a prognostic indicator in patients with 1980; 64:65–72.
small-cell lung cancer. Cancer 1983; 52:1395–1400. 279. Klefstrom P, Nuortio L, Taskinen E. Post-operative radiation
257. Jondal M, Merrill J, Ullberg M. Monocyte-induced human natural therapy and adjuvant chemoimmunotherapy in breast cancer.
killer cell suppression followed by increased cytotoxic activity Aspects of timing and immune competence. Acta Radiol [Oncol]
during short-term in vitro culture in autologous serum. Scand J 1986; 25:3; 161–166.
Immunol 1981; 14:555–563. 280. Klefstrom P, Grohn P, Heinonen E, et al. Adjuvant postoperative
258. Jones SE, Griffith K, Dombrowski P, Gaines JA. Immunodeficiency radiotherapy, chemotherapy, and immunotherapy in stage III
in patients with non-Hodgkin’s lymphomas. Blood 1977; breast cancer. II. Five-year results and influence of levamisole.
49:335–344. Cancer 1987; 60:5; 936–942.
259. Jose DG, Seshadri R. Circulating immune complexes in neuro- 281. Klefstrom P, Holsti P, Grohn P, Heinonem E. Combination of
blastoma: Direct assay and role in blocking specific cellular levamisole immunotherapy with conventional treatments in
immunity. Int J Cancer 1974; 13:824–838. breast cancer. In: Terry WD, ed. Proceedings of Second
260. Jubert AV, Lee ET, Hersh EM, McBridge CM. Effects of surgery, International Conference on Immunotherapy of Cancer, Bethesda,
anesthesia and intraoperative blood loss on immunocompetence. Maryland, 1982; 19:4128–4130.
J Surg Res 1973; 15:399–403. 282. Kleinerman ES, Zwelling LA, Schwartz R, et al. Effect of
261. Jung TT, Berlinger NT, Juhn SK. Prostaglandins in squamous-cell L-phenylalanine mustard, adriamycin and actinomycin D, and
carcinoma of the head and neck: A preliminary study. Laryngoscope 4′-(9-acridinylamino) methanesulfon-m-anisidide on naturally
1985; 95:307–312. occurring human spontaneous monocyte-mediated cytotoxicity.
262. Kadish AS, Doyle AT, Steinhaur EH, Ghossein NA. Natural cyto- Cancer Res 1982; 42:1692–1695.
toxicity and interferon production in human cancer: Deficient 283. Koba F, Akiyoshi T, Tsuji H. Natural killer-cell activity in the
natural killer activity and normal interferon production in patients perigastric lymph nodes from patients with gastric carcinoma or
with advanced disease. J Immunol 1981; 127:1817–1822. benign lesions. J Clin Lab Immunol 1987; 23:4; 191–195.
263. Kaneko T. Effects of lentinan on cytotoxic functions of human lym- 284. Kova J, Ninger E, Zemanov D, Lauerov P. A prospective study of
phocytes. Immunopharmacol Immunotoxicol 1988; 10:2; 157–163. lymphocyte responses to phytohemagglutinin in melanoma (lack
264. Kaplan HS. Hodgkin’s disease. Cambridge, Massachusetts: of prognostic value of correlation with minimal tumor burden).
Harvard University Press, 1980. Neoplasma 1980; 27:575–582.
265. Kaplan HS. Hodgkin’s disease. Cambridge, Massachusetts: 285. Kraus S, Comas F, Perez C, et al. Treatment of inoperable non-
Harvard University Press, 1980. small-cell carcinoma of the lung with radiation therapy, with or
266. Karavodin LM, Guilliano AE, Golub SH. T-lymphocyte subsets in without levamisole. Am J Clin Oncol 1984; 7:405–412.
patients with malignant melanoma. Cancer Immunol Immunother 286. Kreienberg R, Boerner D, Melchert F, and Lemmell EM. Reduction
1981; 11:251–254. of the immunosuppressive action of chemotherapeutics in patients
267. Kaszubowski PA, Husky G, Tung KS, Williams RC Jr. with mammary carcinoma by Azimexon. J Immunopharmacol
T-lymphocyte subpopulations in peripheral blood and tissues of 1983; 5:49–64.
cancer patients. Cancer Res 1980; 40:4684–4657. 287. Kripke ML. Immunoregulation of carcinogenesis: Past, present,
268. Katz AE. Immunobiologic staging of patients with carcinoma of and future. J Natl Cancer Inst 1988; 80:722–727.
the head and neck. Laryngoscope 1983; 93:445–463. 288. Kumano N, Suzuki S, Oizumi K, et al. Imbalance of T-cell sub-
269. Kawaguchi T, Suematsu M, Koizumi HM, et al. Activation of sets in cancer patients and its modification with bestatin, a small
macrophage function by intraperitoneal administration of the molecular immunomodifier. Tohoku J Exp Med 1985;
streptococcal anti-tumor agent OK-432. Immunopharmacology 147:125–133.
1983; 6:1077–1089. 289. Kurosu Y, Tanaka N, Furusho Y, Morita K. Cimetidine-mediated
270. Kay RG, Mason BH, Stephans EJ, et al. Levamisole in primary augmentation of lymphocyte responses to phytohemagglutinin in
breast cancer: A controlled study in conjunction with L-phenylalanine gastric cancer patients. Jpn J Clin Oncol 1989; 19:1; 56–61.
mustard. Cancer 1983; 51:1992–1997. 290. Lauria F, Raspadori D, Tura S. Effect of a thymic factor on
271. Keller SM, Groshen S, Martini N, Kaiser LR. Blood transfusion T-lymphocytes in B-cell chronic lymphocytic leukemia: In vitro
and lung cancer recurrence. Cancer 1988; 62:606–610. and in vivo studies. Blood 1984; 64:667–671.
138 Therapeutic approaches to cancer-associated immune suppression

291. Lauria F, Foa R, Gobbi M, et al. Increased proportion of suppres- 312. Luzi G, Troopea F, Seminara R, et al. Clinical and immunological
sor/cytotoxic (OKT8+) cells in patients with Hodgkin’s disease evaluation in non-resectable lung cancer patients treated with
in long-lasting remission. Cancer 1983; 52:1385–1388. thymostimulin. In: Byrom NA, Hobbs JR, eds. Thymic factor
292. Lauria JA, Moertel CG, Fleming TR, et al. Surgical adjuvant therapy. New York: Raven Press, 1984:309–320.
therapy of large-bowel carcinoma: An evaluation of levamisole 313. Maca RM, Panje WR. Indomethacin-sensitive suppressor-cell
and the combination of levamisole and fluorouracil. J Clin Oncol activity in head and neck cancer patients pre- and post-irradiation
1989; 7:1447–1456. therapy. Cancer 1982; 50:483–489.
293. Laurie J, Moertel C, Fleming T, et al. Surgical adjuvant therapy 314. Macchiarini P, Danesi R, Del Tacca M, Angeletti CA. Effects of thymo-
of poor prognosis colorectal cancer with levamisole alone or stimulin on chemotherapy-induced toxicity and long-term survival in
combined levamisole and 5-fluorouracil (5-FU) (Abstract). Proc small-cell lung cancer patients. Anticancer Res 1989; 9:1; 193–196.
ASCO 1986; 5:81. 315. Mackay JR, Goodyear MD, Riglar C, et al. Effect on immunologic
294. Lawrence HS. The transfer in humans of delayed skin sensitivity and other indices of adjuvant cytotoxic chemotherapy including
to streptococcal M substance and to tuberculin with disrupted melphalan in breast cancer. Cancer 1984; 53:2619–2627.
leukocytes. J Clin Invest 1955; 34:219–230. 316. Maguire LC, Roszman TL, Lackey S. Failure of cimetidine as an
295. Lee ET, Ishmael R, Bottomley RH, Murray JL. An analysis of skin immunomodulator in cancer patients and normal subjects. South
tests and their relationship to recurrence and survival in stage III Med J 1985; 78:1078–1080.
and stage IV melanoma patients. Cancer 1982; 49:2336–2341. 317. Mahaley MS Jr, Brooks WH, Roszman TL, et al. Immunobiology
296. Lee YT. Delayed cutaneous hypersensitivity, lymphocyte count, of primary intracranial tumors. I. Studies of the cellular immunity
and blood tests, in patients with breast carcinoma. J Surg Oncol of brain tumor patients. J Neurosurg 1977; 46:467–476.
1984; 27:135–140. 318. Maisel RH, Ogura JH. Dinitrochlorobenzene skin sensitization
297. Lee AKY, Rowley M, MacKay IR. Antibody-producing capacity and peripheral lymphocyte count: Predictors of survival in head
in human cancer. Br J Cancer 24:454–463. and neck cancer. Ann Otol Rhinol Laryngol 1976; 85:517–522.
298. Lee YT, Sparks FC, Eilber FR, Morton DL. Delayed cutaneous 319. Mandel MA, Dvorak K, Decasse JJ. Salivary immunoglobulins
hypersensitivity and peripheral lymphocyte counts in patients in patients with oropharyngeal and bronchopulmonary cancer.
with advanced cancer. Cancer 1975; 35:748–755. Cancer 1973; 31:1408–1413.
299. Leibler GA, Concannon JP, Magorern GJ, et al. Immunoprofile 320. Mandeville R, Lamoureux G-T, Poisson SL, Poisson R. Biological
studies for patients with bronchogenic carcinoma. J Thorac markers and breast cancer: A multiparameter study. II. Depressed
Cardiovasc Surg 1977; 74:506–518. immune competence. Cancer 1982; 50:1280–1288.
300. Levin RH, Landy M, Frei E. The effect of 6-mercaptopurine on 321. Mantovani G, Floris C, Maccio A, et al. Role of interleukin-2
immune response in man. N Engl J Med 1964; 271:16–22. (IL-2) in cancer-related immune deficiency: In vitro response to
301. Levo Y, Rotter V, Ramot B. Restoration of cellular immune response IL-2, production of IL-2, and IL-2 receptor expression in patients
by levamisole in patients with Hodgkin’s disease. Biomedicine with advanced cancer. Cancer Detect Prev 1988; 12:149–159.
1975; 23:198–200. 322. Marshall ME, Mendelsohn L, Butler K, et al. Treatment of meta-
302. Lewinski UH, Mavligit GM, Hersh EM. Cellular immune modulation static renal cell carcinoma with coumarin (1,2-benzopyrone) and
after a single high dose of levamisole. Cancer 1980; 46:2185–2194. cimetidine: A pilot study. J Clin Oncol 1987; 5:6; 862–866.
303. Liberati AM, Bragia M, Edwards BS, et al. Immunorestorative 323. Marshall ME, Riley LK, Rhoades J, et al. Effects of coumarin
properties of thymostimulin (TS) in patients with Hodgkin’s dis- (1,2-benzopyrone) and cimetidine on peripheral blood lympho-
ease in clinical remission. Cancer Immunol Immunother 1985; cytes, natural killer cells, and monocytes in patients with
19:136–141. advanced malignancies. J Biol Response Mod 1989; 8:1; 62–69.
304. Liberata AM, Ballatori E, Fizzotti M, et al. A randomized trial to 324. Martelli MF, Velardi A, Rambotti P, et al. The in vivo effect of a thy-
evaluate the immunorestorative properties of thymostimulin in mic factor (thymostimulin) in immunologic parameters of patients
patients with Hodgkin’s disease in complete remission. Cancer with untreated Hodgkin’s disease. Cancer 1982; 50:490–497.
Immunol Immunother 1988; 26:87–93. 325. Mastrangelo MJ, Berd D, Maguire H Jr. Cell-mediated immu-
305. Liberata AM, Voekel JG, Borden EC, et al. Influence of non-spe- nity: The immunoaugmenting effects of cancer chemotherapeutic
cific immunologic factors on prognosis in advanced bronchogenic agents. Sem Oncol 1986; 13:2; 186–194.
carcinoma. Cancer Immunol Immunother 1982; 13:140–144. 326. Mathé G, Amiel JL, Schwarzenberg L, et al. Active immunother-
306. Lichtenstein A, Zighelboim J, Dorey R, et al. Comparison of apy for acute lymphoblastic leukemia. Lancet 1969; 1:697–699.
immune derangements in patients with different malignancies. 327. Mathé G. Do tuftsin and bestatin constitute a biopharmacological
Cancer 1980; 45:2090–2095. immunoregulatory system? Cancer Detect Prev Suppl 1987;
307. Liu W, et al. CD127 expression inversely correlates with FoxP3 1:445–455.
and suppressive function of human CO4+ Treg cells. J Exp Med 328. Mathé G, Umezawa H, Misset JL, et al. Immunomodulating
2006; 203:1701–1711. properties of bestatin in cancer patients. A phase II trial. Biomed
308. Lotan R. Effects of vitamin A and its analogs (retinoids) on normal Pharmacother 1986; 40:10; 379–382.
and neoplastic cells. Biochim Biophys Acta 1980; 605:33–91. 329. Mavligit GM. Immunologic effects of cimetidine: Potential uses.
309. Loutfi A, Shakr A, Jerry M, et al. Double-blind randomized pro- Pharmacotherapy 1987; 7:120S–124S.
spective trial of levamisole/placebo in stage I cutaneous malignant 330. Mavligit GM, Raphael LS, Calvo DB III, Wong WL.
melanoma. Clin Invest Med 1987; 10:4; 325–328. Indomethacin-induced monocyte-dependent restoration of local
310. Ludwig CU, Hartmann D, Landmann R, et al. Unaltered immu- graft-versus-host reaction among cells from cancer patients. J
nocompetence in patients with nondisseminated breast cancer at Natl Cancer Inst 1980; 65:317–320.
the time of diagnosis. Cancer 1985; 55:1673–1678. 331. McGeorge MB, Russel EC, Mohanakumar T. Immunologic eval-
311. Lundy J, Raaf JH, Deakins S, Wanebo HJ. The acute and chronic uation of long-term effects of childhood ALL chemotherapy:
effects of alcohol on the human immune system. Surg Gynecol Analysis of in vitro NK- and K-cell activities of peripheral blood
Obstet 1975; 141:212–218. lymphocytes. Am J Hematol 1982; 12:19–27.
Robert K. Oldham 139

332. McGinnes K, Florence J, Penny R. The effect of radiotherapy on 352. Moertel CG, Ritts RE, O’Connell MJ, Silvers A. Nonspecific
the natural killer (NK) cell activity of cancer patients. J Clin immune determinants in the patient with unresectable gastroin-
Immunol 1987; 7:3; 210–217. testinal carcinoma. Cancer 1979; 43:1483–1492.
333. Menconi E, Barzi A, Greco M, et al. Immunological profile of 353. Monson JR, Ramsden C, Guillou PJ. Decreased interleukin-2
breast cancer patients in early of advanced disease. Experimentia production in patients with gastrointestinal cancer. Br J Surg
1979; 35:820–822. 1986; 73:6; 483–486.
334. Merrin C, Han T. Immune responses in bladder cancer. J Urol 354. Monte JE, Bukowski RM, James RE, et al. A critical review of
1974; 111:170–172. immunotherapy of disseminated renal adenocarcinoma. J Surg
335. Messerschmidt GL, Henry DH, Snyder HW Jr, et al. Protein A Oncol 1982; 21:5–8.
immunoadsorption in the treatment of malignant disease. J Clin 355. Morales A, Eidinger D. Immune reactivity in renal cancer. J Urol
Oncol 1988; 6:2; 203–212. 1976; 115:510–513.
336. Metzher Z, Hoffeld JT, Oppenheim JJ. Evidence for the involve- 356. Morin A, Ballet J, Touraine J, Hadder JW. Current status of iso-
ment of monocyte-derived toxic oxygen metabolites in the lym- prinosine. In: Serrou B, Rosenfeld C, Daniels JC, Saunders JP,
phocyte dysfunction of Hodgkin’s disease. Clin Exp Immunol eds. Current concepts in human immunology and cancer immu-
1981; 46:313–320. nomodulation. Amsterdam: Elsevier Biomedical, 1982:479–489.
337. Micksche M, Cerni C, Kokron K, et al. Stimulation of the immune 357. Morton DL, Eilber FR, Malmgren RA, Wood WC. Immunological
response in lung cancer patients by vitamin A. Oncology 1977; factors which influence response to immunotherapy in malignant
34:234–239. melanoma. Surgery 1970; 68:158–164.
338. Micksche M, Colot M, Kucera H. Radioimmunotherapy in 358. Mukherji B, Guha A, Loomis R, Ergin MT. Cell-mediated amplifi-
advanced cervical cancer. In: Dubois JB, Serrou B, Rosenfeld C, cation and down regulation of cytotoxic immune response against
eds. Immunopharmacologic effects of radiation therapy. New autologous human cancer. J Immunol 1987; 138:1987–1991.
York: Raven Press, 1981:499–518. 359. Mukhopadhyaya R, Advani SH, Gangal SG. Functional evalua-
339. Mihich E. Biological response modifiers in cancer therapeu- tion of T-lymphocytes from peripheral blood and spleens in
tics: Potentialities and limitations. Cancer Detect Prev 1988; Hodgkin’s disease. Br J Cancer 1987; 56:800–802.
12:531–535. 360. Munster AM. Post-traumatic immunosuppression is due to acti-
340. Mihich E, Ehreke MJ, Ishizuka M. Immunomodulation by antibi- vation of suppressor T-cells. Lancet 1976; 1:1329.
otics. In: Mihich E, Sakurai Y, eds. Biological responses in can- 361. Munzarova M, Mechl Z, Kovarik J, Kolcova V. Skin tests with
cer, vol 3: immunomodulation by anti-cancer drugs. New York: autologous cholesteryl hemisuccinate-treated tumor cells, DNCB
Plenum Press, 1985:71–73. and PPD and their relationship to prognosis in malignant mela-
341. Miller HC, Vito C. Action of a thymic cytokine TsIF in reversing noma patients. Neoplasma 1988; 35:2; 229–234.
the autoimmune disease state of the MRL/1pr mouse. Mol 362. Murphy RM, Colton CK, Yarmush ML. Staphylococcal protein A
Biother 1989; 1:4; 213–217. adsorption in neoplastic disease: Analysis of physiochemical
342. Miller LL, Spitler LE, Allen RE, Minot DR. A randomized, aspects. Mol Biother 1989; 1:4; 186–207.
double-blind, placebo-controlled trial of transfer factor as 363. Murray JL, Kollmorgen GM. Inhibition of lymphocyte response
adjuvant therapy for malignant melanoma. Cancer 1988; by prostaglandin-producing suppressor cells in patients with
61:1543–1549. melanoma. J Clin Immunol 1983; 3:268–276.
343. Miller FR, Kataoka T. Interactions of antimetabolites with tumors 364. Murray JL, Reuben JM, Smith TL, et al. A pilot clinical trial of
and the immune system. In: Mihich E, Sakurai Y, eds. Biological the toxicity and immunorestorative effects of T-cell reconstituting
responses in cancer, vol 3: immunomodulation by anti-cancer factor (SR 270258) in immunosuppressed cancer patients.
drugs. New York: Plenum Press, 1985:33–69. J Biol Response Mod 1987; 6:1; 56–68.
344. Mitchell MS, Kempf RA, Harel W, et al. Effectiveness and toler- 365. Najjar VA. Biochemistry and physiology of tuftsin – Thr-Lys-
ability of low-dose cyclophosphamide and low-dose intravenous Pro-Arg. In: Sbarra S, Strauss R, eds. The reticuloendothelial
interleukin-2 disseminated melanoma. J Clin Oncol 1988; 6:3; system. New York: Plenum Press, 1980:45–71.
409–424. 366. Nathanson SD, Zamfirescu PL, Portaro JK, et al. Acute effects of
345. Mitchell MS, Defonti RC. Immunosuppression by 5-fluorouracil. orally administered levamisole on random monocyte motility and
Cancer 1970; 26:884–889. chemotaxis in man. J Natl Cancer Inst 1978; 61:301–306.
346. Mitchell MS, Wade ME, Deconti RC, et al. Immunosuppressive 367. Niimoto M, Hattori T, Ito I, et al. Levamisole as postoperative adju-
effects of cytosine arabinoside and methotrexate in man. Ann Int vant immunochemotherapy for gastric cancer. A randomized con-
Med 1969; 70:535–547. trolled study of the MMC+ Tegafur regimen with or without
347. Miwa H, Orita K. Immunotherapy of gastrointestinal cancer levamisole. Report I. Cancer Immunol Immunother 1984; 18:13–18.
patients with levamisole. Acta Med Okayama 1979; 368. Noda T, Asano M, Yoshie O, et al. Interferon-γ induction of
33:29–42. human peripheral blood mononuclear cells by OK-432, a killed
348. Miwa H, Orita K. Cancer immunotherapy with levamisole. Acta preparation of Streptococcus pyogenes. Microbiol Immunol
Med Okayama 1978; 32:239–245. 1985; 30:81–88.
349. Miwa H, Orita K. Reactivating effect of levamisole on cell-medi- 369. Noma T, Yoshimura N, Yata J. Depressed lymphocyte functions
ated immunity in gastrointestinal cancer patients. Acta Med of cancer patients and their correlation by bestatin (product of
Okayama 1977; 31:325–329. Streptomyces olivereticuli). In: Serrou S, Rosenfeld C, Daniels
350. Modini C, Ziparo V, Cicconetti F, et al. Role of delayed cutane- JC, Saunders JP, eds. Current concepts in human immunology
ous hypersensitivity reaction in classifying patients with bron- and cancer immunomodulation. Amsterdam: Elsevier Biomedical,
chial carcinoma. Tumori 1985; 71:277–281. 1982:611–616.
351. Moertel CG, Fleming TR, MacDonald JS, et al. Levamisole and 370. Nordman E, Saarimaa H, Tolvanen A. The influence of 5-fluorou-
fluorouracil for adjuvant therapy of resected colon carcinoma. N racil on cellular and humoral immunity in cancer patients. Cancer
Engl J Med 1990; 322:6; 352–358. 1978; 41:64–69.
140 Therapeutic approaches to cancer-associated immune suppression

371. Nordman E, Tolvanen A. Effects of irradiation of the immune International Conference on Immunotherapy of Cancer.
function in patients with mammary, pulmonary or head and neck Amsterdam: Elsevier/North-Holland, 1980:47–54.
carcinoma. Acta Radiol Oncol Radiat Phys Biol 1978; 17:3–9. 392. Payne JE, Meyer JH, Macpherson JG, et al. The value of lympho-
372. Norris RW, Byrom NA, Nagvekar NM, et al. Thymostimulin plus cyte transformation in carcinoma of the colon and rectum. Surg
surgery in the treatment of primary truncal malignant melanoma: Gynecol Obstet 1980; 150:687–693.
Preliminary results of a U.K. multicentre clinical trial. In: Byrom 393. Penn I. Tumors of the immunocompromised patient. Annu Rev
NA, Hobbs JR, eds. Thymic factor therapy. New York: Raven Med 1988; 39:63–73.
Press, 1984:341–348. 394. Penn I. Occurrence of cancer in immune deficiency. Cancer 1974;
373. Oka S. A review of clinical studies of bestatin. Recent results. 34:858–866.
Cancer Res 1980; 75:126–132. 395. Perez CA, Bauer M, Emami BN, et al. Thoracic irradiation with or
374. Olivari AJ, Glait HM, Guardo A, et al. Levamisole in squamous- without levamisole (NSC #177023) in unresectable non-small-cell
cell carcinoma of the head and neck. Cancer Treat Rep 1979; carcinoma of the lung: A phase III randomized trial of the RTOG.
63:983–990. Int J Radiation Oncology Biol Phys 1988; 15:6; 1337–1346.
375. Onsrud M. Whole pelvic irradiation in stage I endometrial carci- 396. Petrini B, Wasserman J, Blomgren H, Rotstein S. T-helper/
noma: Changes in numbers and reactivities of some blood lym- suppressor ratios in chemotherapy and radiotherapy. Clin Exp
phocyte subpopulations. Gynecol Oncol 1982; 13:283–292. Immunol 1983; 53:255–256.
376. Orita K, Miwa H, Fukuda H, et al. Preoperative cell-mediated 397. Petrini B, Wasserman J, Glas U, Blomgren H. T-lymphocyte sub-
immune status of gastric cancer patients. Cancer 1976; populations in blood following radiation therapy for breast cancer.
38:2343–2348. Eur J Cancer Clin Oncol 1982; 18:921–924.
377. Osband M, Shen Y, Schlesinger M, et al. Successful tumor immu- 398. Petrini B, Wasserman J, Biberfeld G, et al. The effect of in vitro irra-
notherapy with cimetidine in mice. Lancet 1981; 1:636–638. diation on PHA-mediated cytotoxicity and lymphocytes with recep-
378. Oshimi K, Kano S, Takaku F, Okumura K. Augmentation of tors for the Fc pat of Ig. J Clin Lab Immunol 1979; 2:333–336.
mouse natural killer cell activity by a streptococcal preparation 399. Pierri I, Cordone G, Rogna S, et al. T-lymphocyte phenotype and
OK-432. J Natl Cancer Inst 1980; 65:1265–1269. functions in patients with head and neck cancer. Laryngoscope
379. Ota K, Kurita S, Yamada K, et al. Immunotherapy with bestatin 1985; 95:577–581.
for acute non-lymphocytic leukemia in adults. Cancer Immunol 400. Pines A. BCG plus levamisole following irradiation of advanced
Immunother 1986; 23:1; 5–10. squamous bronchial carcinoma. Int J Radiation Oncology Biol
380. Ozer H. Effects of alkylating agents on immunoregulatory mech- Phys 1980; 6:1041–1042.
anisms. In: Mihich E, Sakurai Y, eds. Biological responses in 401. Pinsky CM, El-Domeiri A, Caron AA, et al. Delayed hypersensi-
cancer, vol 3: immunomodulation by anti-cancer drugs. New tivity reactions in patients with cancer. In: Mathé G, Weiner R,
York: Plenum Press, 1985:95–129. eds. Recent results in cancer research, 1970; 35: 37–41.
381. Paavonen T, Anderson LC, Adlercrentz H. Estradiol enhances 402. Pinsky CM, Wanebo H, Mike V, Oettgen H. Delayed cutaneous
human B-cell maturation via inhibition of suppressor T-cells in hypersensitivity reactions and prognosis in patients with cancer.
pokeweed mitogen-stimulated cultures. J Exp Med 1981; Ann NY Acad Sci 1976; 276:407–410.
154:1935–1945. 403. Pompidou A, Soubrane C, Cour V, et al. Immunological effects of
382. Padmanabhan TK, Balaram P, Vasudevan DM. Role of levamisole isoprinosine as a pulse immunotherapy in melanoma and ARC
immunotherapy as an adjuvant to radiotherapy in oral cancer. I. A patients. Cancer Detect Prev Suppl 1987; 1:457–462.
three-year clinical follow-up. Neoplasma 1987; 34:5; 627–632. 404. Posner MR, Reinherz E, Lane H, et al. Circulating lymphocyte
383. Paik YK, Kimura L, Yanagihora E, et al. Inhibition of the phyto- populations in Hodgkin’s disease after mantle and para-aortic
hemagglutinin stimulation of leukocytes by C-reactive protein. J irradiation. Blood 1983; 61:705–708.
Reticuloendothel Soc 1972; 11:420. 405. Powles TJ, Dady DJ, Williams J, et al. Use of inhibitor of prostaglan-
384. Papatestas AE, Kark AE. Peripheral lymphocyte counts in breast din synthesis and patients with breast cancer. In: Sammuelsson B,
carcinoma: An index of immune competence. Cancer 1978; Ramwel PW, Paoletti R, eds. Advances in prostaglandin and throm-
34:2014–2017. boxane research, vol 6. New York: Raven Press, 1980 511–516.
385. Parkinson DR, Cano PO, Jerry LM, et al. Complications of can- 406. Prehn RT, Prehn LM. The flip side of tumor immunity. Arch Surg
cer immunotherapy with levamisole. Lancet 1977; i:1129–1132. 1989; 124:102–106.
386. Passwell J, Lavanon M, Davidsohn J, Ramot B. Monocyte PG E2 407. Prehn RT, Prehn LM. The autoimmune nature of cancer. Cancer
secretion in Hodgkin’s disease and its relation to decreased cel- Res February 15, 1987; 47: 927–932.
lular immunity. Clin Exp Immunol 1983; 51:61–68. 408. Pritchard DJ, Ritts RE Jr, Taylor WF, Miller GC. A prospective
387. Paterson AHG, Natting M, Takata L, et al. Chemoimmunotherapy study of immune responsiveness in humans. I. Assessment of ini-
with levamisole in metastatic breast carcinoma: a controlled tial pretreatment status with stage of disease. Cancer 1978;
clinical trial. Cancer Clin Trials 1980; 3:5–10. 41:2165–2173.
388. Patt YZ, Hersh EM, Reuben J, et al. A phase-I study of intrave- 409. Pross HF, Baines MG. Spontaneous human lymphocyte-mediated
nous azimexon therapy in human cancer. J Biol Response Mod cytotoxicity against tumor target cells: I. The effect of malignant
1986; 5:4; 313–318. disease. Int J Cancer 1976; 18:593–604.
389. Pauwels R, Van der Straeter M. Delayed hypersensitivity in patients 410. Pui C-H, Ip SH, Kung P, et al. High serum interleukin-2 receptor
with bronchial carcinoma. Scand J Resp 1975; 56:160–164. levels are related to advanced disease and a poor outcome in child-
390. Pavlovsky S, Muriel FS, Garay G, et al. Chemoimmunotherapy hood non-Hodgkin’s lymphoma. Blood 1987; 70:3; 624–628.
with levamisole in acute lymphoblastic leukemia. Cancer 1981; 411. Pulay TA, Csomor S. Effect of levamisole treatment on immuno-
48:1500–1507. logical parameters and the early course of cervical cancer.
391. Pavlovsky S, Garay G, Sackman MF, Svarch E. Levamisole ther- Neoplasia 1980; 29:81–86.
apy during maintenance of remission in patients with acute lym- 412. Quirt I, Shelley W, Bodurtha A, et al. Adjuvant levamisole
phoblastic leukemia. In: Terry WD, ed. Proceedings of Second improves survival and disease-free survival in patients with poor
Robert K. Oldham 141

prognostics with malignant melanoma. (Abstr.) Proc ASCO of cancer: Present status on trials in man. New York: Raven Press,
1986; 5:130. 1978:635–646.
413. Raben M, Walch N, Galili U, Schlesinger M. The effect of radia- 432. Romagnani S, Maggi E, Del Prete G, et al. Short- and long-term
tion therapy on lymphocyte subpopulations in cancer patients. effects of radiation on T-cell subsets in peripheral blood of
Cancer 1976; 37:1417–1421. patients with Hodgkin’s disease. Cancer 1980; 46:2590–2595.
414. Ramot B, Biniaminov M, Shoham C, Rosenthal E. The effect of 433. Romagnani S, Ferrini PLR, Ricci M. The immune derangement
levamisole on E-rosette-forming cells in vivo and in vitro in in Hodgkin’s disease. Semin Oncol 1985; 22:41–55.
Hodgkin’s disease patients. N Engl Med 1976; 194:809–811. 434. Rosenberg SA, Seipp CA, White DE, Wesley R. Perioperative
415. Ramot B, Rosenthal E, Biniaminov M, Ben-Bassat I. Effect of blood transfusions are associated with increased rates of recurrence
levamisole, thymic humoral factor, and indomethacin on E-rosette and decreased survival in patients with high-grade soft-tissue sar-
formation of lymphocytes in Hodgkin’s disease. Isr J Med Sci comas of the extremities. J Clin Oncol 1985; 3:5; 698–709.
1981; 17:232–235. 435. Rossen RD, Morgan AC. Blockage of the humoral immune
416. Rangel DM, Golub SH, Morton DL. Demonstration of lympho- response: Immune complexes in cancer. Cancer Immunol 1980;
cyte blastogenesis-inhibiting factors in sera of melanoma patients. 210–280.
Surgery 1977; 82:224–232. 436. Roth JA, Osborne BA, Ames RS. Immunoregulatory factors
417. Rapson NT, Cornbleet MA, Chessells JM, Hardisity RM. derived from human tumors. II. Partial purification and further
Immunosuppression and serious infections in children with acute immunobiochemical characterization of a human sarcoma-derived
lymphoblastic leukemia: A comparison of the three chemotherapy immunosuppressive factor expressing HLA-DR and immuno-
regimens. Br J Hematol 1980; 45:41–52. globulin-related determinants. J Immunol 1983; 130:303–308.
418. Ravikumar T, Steele G, Rodrick M, et al. Effects of tumor growth 437. Roth JA, Golub SH, Grimm EA, et al. Effects of operation on
on interleukins and circulating immune complexes. Cancer 1984; immune response in cancer patients: Sequential evaluation of in vitro
53:1373–1378. lymphocyte function. Surgery 1976; 79:47–51.
419. Reed DM. On the pathological changes in Hodgkin’s disease 438. Rotstein S, Blomgren H, von Stedingk LV, et al. Long-term effects
with special references to its relation to tuberculosis. Johns on the immune system following local irradiation for breast can-
Hopkins Hosp Rep 1902; 10:133–198. cer. Pokeweed mitogen-induced immunoglobulin secretion by
420. Rees JC, Rossio JL, Wilson HG, et al. Cellular immunity in neopla- blood lymphocytes and serum immunoglobulin levels. Eur J Surg
sia. Antigen and mitogen responses in patients with bronchogenic Oncol 1985; 11:137–141.
carcinoma. Cancer 1975; 36:2010–2015. 439. Rotstein S, Blomgren H, Nilsson B, et al. Prognostic value of various
421. Remacle-Bonnet MM, Rommier GJ, Luc C, et al. Nonspecific immunological tests in breast cancer patients treated with adjuvant
suppressive and cytostatic activities mediated by human colonic postoperative chemotherapy. Chemotherapy 1984; 3:227–231.
carcinoma tissue or cultured cell extract. J Immunol 1978; 440. Rotstein S, Blomgren H, Petrini, et al. Blood lymphocyte counts
121:44–52. with subset analysis in operable breast cancer. Cancer 1985;
422. Renoux G, Renoux M. DTC. A summary of current status. In: 56:1413–1419.
Serrou B, Rosenfeld C, Daniels JC, Saunder JP, eds. Current con- 441. Ryan RE Jr, Neel HB, Ritts RE. Correlation of pre-operative
cepts in human immunology and cancer immunomodulation. immunologic test results with recurrence in patients with head
Amsterdam: Elsevier Biomedical 1982:575–584. and neck cancer. Otolaryngol Head Neck Surg 1980; 88:58–63.
423. Renoux G, Renoux M. Effect immunostimulant d’un imidothiazole 442. Saijo N, Shimizu E, Shibuya M, et al. Effect of chemotherapy on
dans l’immunization des souris contre l’infecction par Brucella natural killer (NK) activity and antibody-dependent cell-mediated
abortus. C R Acad Sci (Paris) 1971; 272:349–350. cytotoxicity in carcinoma of the lung. Br J Cancer 1982; 46:
424. Ress JC, Rossio HL, Wilson HE, et al. Cellular immunity in neo- 180–189.
plasia. Antigen and mitogen response in patients with broncho- 443. Saito M, Ebina T, Koi M, et al. Induction of interferon-gamma in
genic carcinoma. Cancer 1975; 36:2010–2015. mouse spleen cells by OK-432, a preparation of Streptococcus
425. Retsas S, Thomas C, Hobbs JR. The effect of vindesine therapy pyogenes. Cell Immunol 1982; 68:187–192.
on the in vitro immune response of patients with advanced malig- 444. Sakaguchi S. Naturally arising CO4(+) regulatory T cells for
nant melanoma. Clin Oncol 1981; 7:33–37. immunologic self-tolerance and negative control of immune
426. Richtsmeier WJ, Eisele D. In vivo anergy reversal with cimeti- responses. Annu Rev Immunol 2004; 22:531–562.
dine in patients with cancer. Arch Otolaryngol Head Neck Surg 445. Salmon SE, Hunt A, Bonnet JD, et al. Alternating combination
1986; 112:10; 1074–1077. chemotherapy and levamisole improves survival in multiple
427. Rijswijk REN, Sybesma JPH, Kater L. A prospective study of the myeloma: A Southwest Oncology Group study. J Clin Oncol
changes in the immune status before, during and after multiple 1983; 1:453–461.
agent chemotherapy for Hodgkin’s disease. Cancer 1983; 446. Sample WF, Gertner HR, Chretien PB. Inhibition of phytohe-
51:637–644. magglutinin-induced in vitro lymphocyte transformation by
428. Rijswijk REN, Sybesma JPH, Kater L. A prospective study of the serum from patients with carcinoma. J Natl Cancer Inst 1971;
changes in immune status following radiotherapy for Hodgkin’s 46:1291–1297.
disease. Cancer 1984; 53:62–69. 447. Santos GW, Owens AH, Sensenbrenner LL. Effects of selected
429. Roberts MM. Lymphocyte transformation in breast cancer. Br J cytotoxic agents on antibody production in man: A preliminary
Surg 1970; 57:381. report. Ann NY Acad Sci 1964; 114:404–423.
430. Robinson BW. Asbestos and cancer: Human natural killer cell 448. Santos LB, Yamada FT, Scheinberg MA. Monocyte and lympho-
activity is suppressed by asbestos fibers but can be restored by cyte interaction in patients with advanced cancer. Evidence for
recombinant interleukin-2. Am Rev Respir Dis 1978; 139:4; deficient IL-1 production. Cancer 1985; 56:1553–1558.
897–901. 449. Sarin PS, San DK, Thornton AH, et al. Neutralization of HTLV-
431. Rojas AF, Feirstan JN, Glait HM, Olivari AJ. Levamisole action III/LAV replication by antiserum to thymosin-alpha-one. Science
in breast cancer. In: Terry WD, Windhorst D, eds. Immunotherapy 1986; 232:1135–1137.
142 Therapeutic approaches to cancer-associated immune suppression

450. Sato E, et al. Intraepithelial CO8(+) tumor-infiltrating lympho- 470. Seder RH, Vaughn CW, Oh S-K, et al. Tumor regression and tem-
cytes and a high CO8(+)/regulatory T cell ratio are associated porary restoration of immune response after plasmapheresis in a
with a favorable prognosis in ovarian cancer. Proc Natl Acad Sci patient with recurrent oral cancer. Cancer 1987; 60:318–325.
USA 2005; 102:18538–18543. 471. Sedman PC, Ramsden CW, Brennan TG, et al. Effects of low-
451. Scambia G, Panici PB, Mancuso S, et al. Effects of antiestrogen dose perioperative interferon on the surgically induced suppres-
and progestin on immune functions in breast cancer patients. sion of antitumor immune response. Br J Surg 1988;
Cancer 1988; 61:2214–2218. 75:976–981.
452. Schantz SP, Savage HE, Racz T, et al. Immunologic determinants 472. Sen L, Borella L. Expression of cell surface markers on T- and
of head and neck cancer response to induction chemotherapy. J B-lymphocytes after long-term chemotherapy of acute leukemia.
Clin Oncol 1989; 7:7; 857–864. Cell Immunol 1973; 9:84–95.
453. Schlechter GP, Wahl LM, Oppenheim JJ. Suppressor monocytes 473. Serdengecti S, Buyukunal E, Molinas N, et al. Overall survival
in human disease: A review. Adv Exp Med Biol 1979; 121B: results of non-small cell lung cancer patients: Chemotherapy
283–298. alone versus chemotherapy with combined immunomodulation.
454. Schechter GP, Soehlen F. Monocyte-mediated inhibition of lym- Chemioterapia 1988; 7:2; 122–126.
phocyte blastogenesis in Hodgkin’s disease. Blood 1978; 474. Sertoli MR, Guarneri D, Rubagotti A, et al. Adjuvant immuno-
52:261–271. chemotherapy in colorectal cancer. Dukes C. Oncology 1987;
455. Scher HI, Shank B, Chapman R, et al. Randomized trial of com- 44:2; 78–81.
bined modality therapy with and without thymosin fraction V in 475. Shafir M, Bekesi JG, Papatestas A, et al. Preoperative and post-
the treatment of small cell lung cancer. Cancer Res 1988; operative immunological evaluation of patients with colorectal
48:1663–1670. cancer. Cancer 1980; 46:700–705.
456. Schindler L, Leroux M, Zimmerman GF, et al. Reversal of defec- 476. Shank B, Scher H, Hilaris BS, et al. Increased survival with high-
tive lymphoproliferation in postoperative patients with colon dose multified radiotherapy and intensive chemotherapy in limited
cancer. J Cancer Res Clin Oncol 1987; 113:2; 166–170. small cell carcinoma of the lung. Cancer 1985; 56:2771–2778.
457. Schlelhammer PF, Bracken RB, Bean M, et al. Immune evaluator 477. Shiraki S, Mori H, Kadomoto N, et al. Adjuvant immunotherapy
with skin testing. A study of testicular, prostatic, and bladder of carcinoma colli with levamisole. Prevention of immunological
neoplasms. Cancer 1976; 38:149–156. depression following surgical therapy and radiotherapy. Int J
458. Schlumberger HD. BAY I 7433: a synthetic polymer with antitu- Immunopharmacol 1982; 4:73–80.
mor activity. In: Hersh EM, Cirigos MA, Mastrangelo, MJ, eds. 478. Shoham J, Theodore E, Brenner HJ, et al. Enhancement of the
Augmenting agents in cancer therapy. New York: Raven Press, immune system of chemotherapy-treated cancer patients by
1981:373–390. simultaneous treatment with thymic extract, TP-1. Cancer
459. Schulof RS, Lloyf MJ, Cleary PA, et al. A randomized trial to evalu- Immunol Immunother 1980; 9:173–180.
ate the immunorestorative properties of thymosin-alpha-one in 479. Shukla HS, Hughes LE, Whitehead RH, Newcomb RG. Long-term
patients with lung cancer. J Biol Response Mod 1985; 4:147–158. follow-up of general immune competence in breast cancer. II.
460. Schulof RS, Bockman RS, Garofalo JA, et al. Multivariate analy- Sequential pre- and post-treatment levels: A 10-year study. Cancer
sis of T-cell functional defects and circulating serum factors in Immunol Immunother 1986; 21:6–11.
Hodgkin’s disease. Cancer 1981; 48:964–973. 480. Sibbit WL, Bankhurst AD, Williams RC. Studies of cell subpop-
461. Schulof RS, Michitsch RW, Livingston PO, Gupta S. Monocyte- ulations mediating mitogen hyporesponsiveness in patients with
mediated suppression on lymphocyte cytotoxic activity for cultured Hodgkin’s disease. J Clin Invest 1978; 61:55–63.
autologous melanoma cells. Cell Immunol 1981; 57:529–532. 481. Silverman NA, Alexander JC, Potvin C, Chretien PB. In vitro
462. Schulof RS, Chorba TC, Cleary PA, et al. T-cell abnormalities lymphocyte reactivity and T-cell levels in patients with mela-
after mediastinal irradiation for lung cancer. The in vitro influ- noma: Correlations with clinical and pathological stage. Surgery
ence of synthetic thymosin alpha-1. Cancer 1985; 55:974–983. 1976; 79:332–339.
463. Schulof RS. Thymic peptide hormones: Basic properties and 482. Sinclair T, Ezdinli EZ, Boonlayangoor P, et al. Rosette and blas-
clinical applications in cancer. CRC Critical Rev Oncol Hematol togenesis inhibition by plasma from Hodgkin’s disease and other
1985; 3:309–376. malignancies. Positive correlation in stage I and II Hodgkin’s disease.
464. Schulof RS, Lee BJ, Lacher MJ, et al. Concanavalin-A-induced Cancer 1983; 51:238–244.
suppressor cell activity in Hodgkin’s disease. Clin Immunol 483. Slade MS, Simmons RL, Yunis E, Greenberg LJ. Immunodepression
Immunopathol 1980; 16:454–462. after major surgery in normal patients. Surgery 1975;
465. Schulof RS, Lloyd MJ, Ueno WM, et al. Phase II trial of thymosin 78:363–372.
fraction 5 in advanced renal cancer. J Biol Response Mod 1984; 484. Slater JM, Ngo E, Lau BHS. Effect of therapeutic irradiation on
3:151–159. the immune response. Am J Roentgenol Radium Ther Nucl Med
466. Schulof RS, Lacher MJ, Gupta S. Abnormal phytohemaggluti- 1976; 126:313–320.
nin-induced T-cell proliferative responses in Hodgkin’s disease. 485. Smalley RV, Talmadge J, Oldham RK, Thurman GB. The thy-
Blood 1981; 57:607–613. mosins: Preclinical and clinical studies with fraction V and alpha-1.
467. Seddiki N, et al. Expression of interleukin (IL)-2 and IL-7 recep- Cancer Treat Rev 1984; 11:69–84.
tors discriminates between human regulatory and activated T 486. Smith RB, Dekernion J, Baron B, et al. Levamisole in the treat-
cells. J Exp Med 2006; 203:1693–1700. ment of non-invasive and invasive bladder cancer: A preliminary
468. Seder RH, Katz AE, Keggins JJ, et al. Plasma exchange in six report. J Urol 1978; 119:347–349.
patients with advanced cancers of the head and neck. J Clin 487. Snyderman R, Siegler HF, Meadew R. Abnormalities of mono-
Apheresis 1985; 2:238–249. cyte chemotaxis in patients with melanoma: Effects of immuno-
469. Seder RH, Katz AE, Keggins JJ, et al. Plasma exchange in six therapy and tumor removal. J Natl Cancer Inst 1977; 58:37–41.
patients with advanced cancers of the head and neck. J Clin 488. Sokal JE, Aungst CW. Response to BCG vaccination and survival
Apheresis 1985; 2:238–249. in advanced Hodgkin’s disease. Cancer 1969; 24:128–134.
Robert K. Oldham 143

489. Soto A, Hall T, Reed S. Trial of antiviral action of isoprinosine reaction of cancer patients. Clin Immunol Immunopathol 1982;
against rhinovirus infection of volunteers. Antimicrob Agent 24:155–160.
Chemother 1973; 3:332–334. 509. Tamura K, Shibata Y, Matsuda Y, Ishida N. Isolation and charac-
490. Soulillou JP, Douillard JU, Vie H, et al. Defects in lectin-induced terization of an immunosuppressive acidic protein from ascitic
interleukin-2 (IL-2) production by peripheral blood lymphocytes fluids of cancer patients. Cancer Res 1981; 41:3244–3252.
of patients with Hodgkin’s disease. Eur J Cancer Clin Oncol 510. Taramelli D, Mazzocchi A, Clemente C, et al. Lack of suppres-
1985; 21:935. sive activity of human primary melanoma cells on the activation
491. Spitler LE, Sagebiel R. A randomized trial of levamisole versus of autologous lymphocytes. Cancer Immunol Immunother 1988;
placebo as adjuvant therapy in malignant melanoma. N Engl J 26:1; 61–66.
Med 1980; 303:1143–1147. 511 Tarpley JL, Twomey PL, Catalona WJ, Cretien PB. Suppression
492. Spreafico F, Vecchi A. The imunomodulatory activity of certain of cellular immunity by anesthesia and operation. J Surg Res
cancer chemotherapeutic agents. In: Mihich E, Sakurai Y, eds. 1977; 22:195–201.
Biological responses in cancer, vol. 3: immunomodulation by 512. Tartter PI, Burrows L, Kirshner P. Perioperative blood transfu-
anti-cancer drugs. New York: Plenum Press, 1985:131–149. sion adversely affects prognosis after resection in stage I (subset
493. Steele G, Wang BS, Mannick JA. Immune RNA and transfer factor. No) non-oat cell lung cancer. J Thorac Cardiovasc Surg 1984;
In: Mihich E, ed. Immunological approaches to cancer therapeutics. 88:659–662.
New York: Wiley, 1982:257–276. 513. Tartter PI, Martinelli G. Lymphocyte subsets, natural killer cyto-
494. Stefani S, Kerman R, Abbate J. Serial studies of immunocompe- toxicity, and perioperative blood transfusion for elective colorectal
tence in head and neck cancer patients undergoing radiation cancer surgery. Cancer Detect Prev (Suppl) 1987; 1:571–576.
therapy. Am J Roentgenol Radium Ther Nucl Med 1976; 514. Tartter PI, Burrows L, Papatestas AE, et al. Perioperative blood
126:880–886. transfusion has prognostic significance for breast cancer. Surgery
495. Stefano S, Kerman R, Abbate J. Serial studies of immunocompe- 1985; 97:225–230.
tence in head and neck cancer patients undergoing radiation 515. Taylor SA, Gordon MY, Currie GA, Shepherd VB. Irradiation
therapy. Am J Roentgenol 1976; 126:880–886. and monocyte function in patients with breast cancer. Int J Rad
496. Stein JA, Adler A, Efraim SB, Maor M. Immunocompetence, Oncol 1979; 5:2063–2067.
immunosuppression and human breast cancer. Cancer 1976; 516. Theofilopoulos A, Dixon FJ. The biology and detection of
38:1171–1187. immune complexes. Adv Immunol 1979; 28:89–220.
497. Stephens EJW, Wood HF, Mason B. Levamisole as adjuvant to 517. Thienpont D, Vanparjis OFJ, Racymakers AHM, et al. Tetramasole
cyclic chemotherapy in end-stage mammary carcinoma. Recent (R8299): A new potent broad spectrum anthelmintic. Nature
Results Cancer Res 1978; 68:139–145. 1966; 209:1084–1086.
498. Stratton JA, Byfield PE, Byfield JE, et al. A comparison of the 518. Thornes RD, Lynch G, Sheehan MV. Cimetidine and coumarin
acute effects of radiation therapy, including or excluding the thymus, therapy of melanoma. Lancet 1982; 2:328.
or the lymphocyte subpopulations of cancer patients. J Clin 519. Thornes RD, Lynch G. Combination of cimetidine with other
Invest 1975; 56:88–97. drugs for the treatment of cancer. N Engl J Med 1983; 308:591.
499. Stratton JA, Fast PE, Weintraub I. Recovery of lymphocyte func- 520. Tilden AB, Balch CM. Immune modulatory effects of indometha-
tion after radiation therapy for cancer in relationship to prognosis. cin in melanoma patients are not related to prostaglandin
J Clin Lab Immunol 1982; 7:147–153. E2-mediated suppression. Surgery 1982; 92:528–532.
500. Strender LE, Blomgren H, Wasserman J, et al. Influence of adju- 521. Toge T, Hamamoto S, Itagaki E, et al. Concanavalin-A-induced
vant radiation therapy in breast cancer on PWM-induced Ig and spontaneous suppressor cell activities in peripheral blood
secretion by blood lymphocytes in vitro. Anticancer Res 1983; lymphocytes and spleen cells from gastric cancer patients. Cancer
3:41–45. 1983; 52:1624–1631.
501. Strender LE, Blomgren H, Petrini B, et al. Immunologic monitor- 522. Toivanen A, Granberg I, Nordman E. Lymphocyte subpopulations
ing in breast cancer patients receiving postoperative adjuvant in patients with breast cancer after postoperative radiotherapy.
chemotherapy. Cancer 1981; 48:1996–2002. Cancer 1984; 54:2919–2923.
502. Sugden PJ, Lilleyman JS. Impairment of lymphocyte transforma- 523. Tom BH, Macek CM, Subramanian C, et al. In vitro expression of
tion by plasma from patients with advanced Hodgkin’s disease. suppressogenic and enhancing activities in human colon cancer
Cancer 1980; 45:899–905. cells. J Biol Response Mod 1984; 3:435–444.
503. Symoens J, Veys E, Mielants W, Pinals R. Adverse reactions to 524. Tonnesen H, Knigge U, Bulow S, et al. Effect of cimetidine on
levamisole. Cancer Treat Rep 1978; 62:1721–1730. survival after gastric cancer. Lancet 1988; 29:2(8618); 990–992.
504. Taguchi T. Clinical efficacy of lentinan on patients with stomach 525. Treurniet-Donker AD, Meischke-de Jongh ML, van Putten WLJ.
cancer: Endpoint results of a four-year follow-up survey. Cancer Levamisole as adjuvant immunotherapy in breast cancer. Cancer
Detect Prev Suppl 1987; 1:333–349. 1987; 59:1590–1593.
505. Takusugi M, Ramseyer A, Takasugi J. Decline of natural non- 526. Tsang KY, Fudenberg HH, Pan JF, et al. An in vitro study on the
selective cell-mediated cytotoxicity in patients with tumor progres- effects of isoprinosine on immune responses in cancer patients.
sion. Cancer Res 1977; 37:413–418. Int J Immunopharmacol 1983; 5:481–490.
506. Talmadge JE, Lenz BF, Pennington R, et al. Immunomodulatory 527. Tsuyuguchi I, Shiratsuchi H, Fukuoka M. T-lymphocyte subsets
and therapeutic properties of bestatin in mice. Cancer Res 1986; in primary lung cancer. Jpn J Clin Oncol 1987; 17:1; 13–17.
46:9; 4505–4510. 528. Uchida A, Kolb R, Micksche M. Generation of suppressor cells
507. Talmadge JE, Lenz B, Schneider M, et al. Immunomodulatory for natural killer activity in cancer patients after surgery. J Natl
and therapeutic properties of FK-565 in mice. Cancer Immunol Cancer Inst 1982; 68:735–741.
Immunother 1989; 28:93–100. 529. Uchida A, Mickskche M. Generation of suppressor cells f or
508. Talpaz M, Medina JE, Patt YZ, et al. The immune restorative natural killer (NK) activity in cancer patients after surgery.
effect of cimetidine administration in vivo on the local graft-vs-host Cancer Immunol Immunother 1981; 10:203–210.
144 Therapeutic approaches to cancer-associated immune suppression

530. Uchida A, Hoshino T. Clinical studies on well-mediated immunity 549. Wanebo HJ. Observations on the effects of adjuvant radiation on
in patients with malignant disease. I. Effects of immunotherapy immune tests of patients with colorectal cancer and head and
with OK-432 on lymphocyte subpopulation and phytomitogen neck cancer. In: Dubois JB, Serrou B, Rosenfeld C, eds.
responsiveness in vitro. Cancer 1980; 45:476–483. Immunopharmacologic effects of radiation therapy. New York:
531. Umeda T, Yokoyama H, Kobayashi K, et al. Subsets of Raven Press, 1980:241–252.
T-lymphocytes of patients with malignancies of the urogenital 550. Wanego HJ, Riley T, Katz D, et al. Indomethacin-sensitive sup-
tract. Cell Mol Biol 1979; 25:95–99. pressor-cell activity in head and neck cancer patients. Cancer
532. Uracz W, Mytar B, Zembala M, et al. Activated monocytes in 1988; 61:462–474.
gastric cancer patients. II. Suppressor and cytostatic activity in 551. Wara WM, Phillips TL, Wara DW, et al. Immunosuppression fol-
vitro. J Cancer Res Clin Oncol 1982; 104:307–313. lowing radiation therapy for carcinoma of the nasopharynx. Am J
533. Van Rijswijk REN, DeMeijer A, Bob Sybesma JPH, Kater L. Roentgenol Radium Ther Nucl Med 1975; 123:482–485.
Five-year survival in Hodgkin’s disease. The prospective value of 552. Wara WM, Wara DW, Philips TL, Amman AJ. Elevated IgA in
immune status at diagnosis. Cancer 1986; 57:1489–1496. cancer of the nasopharynx. Cancer 1975; 35:1313–1315.
534. Van-Stolen K, Wiernick PH, Schiffer CA, Schimpff SC. 553. Wara WM, Neely MH, Amman AJ, Wara DW. Biological modifi-
Evaluation of levamisole as an adjuvant to chemotherapy for cation of immunologic parameters in head and neck cancer patients
treatment of ANLL. Cancer 1983; 51:1576–1580. with thymosin fraction V. In: Goldstein AL, Chirigos MA, eds.
535. Vanhaelen CPJ, Fisher RI. Increased sensitivity of T-cells to reg- Lymphokines and thymic hormones: Their potential utilization in
ulation by normal suppressor cells persists in long-term survivors cancer therapeutics. New York: Raven Press, 1981:257–262.
with Hodgkin’s disease. Am J Med 1982; 72:385–390. 554. Warren RP, McCall CA, Urban RW. Augmentation of natural killer
536. Vaudaux P, Kiefer B, Forni M, et al. Adriamycin impairs phago- cell activity by ImuVert: A biological response modifier derived
cytic function and induces morphologic alterations in human from Serratia marcescens. Mol Biother 1989; 1:3; 145–151.
neutrophils. Cancer 1984; 59:400–410. 555. Wasserman J, Wallgren A, Blomgren H, et al. Prognostic rele-
537. Venkataraman M, Rao DS, Levin RD, Westerman MP. vance of post-irradiation lymphocyte reactivity in breast cancer
Suppression of B-lymphocyte function by T-lymphocytes in patients. Cancer 1986; 58:348–351.
patients with advanced lung cancer. J Natl Cancer Inst 1985; 556. Wasserman J, Blomgren H, Petrini B, et al. Effect of radiation
74:37–41. therapy and in vitro x-ray exposure on lymphocyte subpopula-
538. Verhaegen H, DeCree J, DeCock W, et al. Levamisole therapy in tions and their functions. Am J Clin Oncol 1982; 5:195–208.
patients with colorectal cancer. In: Terry WD, Rosenberg SA, 557. Watanabe Y, Takashi I. Clinical value of immunotherapy for lung
eds. Immunotherapy of human cancer. New York: Elsevier/ cancer by the streptococcal preparation OK-432. Cancer 1984;
North-Holland, 1982:225–229. 53:248–253.
539. Vinzenz K, Matejka M, Watzek G, et al. Modulation of NK activ- 558. Webster DJ, Whitehead RH, Richardson G, Hughes LE.
ity in regional lymph nodes by preoperative immunotherapy with Levamisole: A double-blind immunological study. Anticancer
OK-432 in patients with cancer of the oral cavity. Cancer Detect Res 1982; 2:29–32.
Prev Suppl 1987; 1:463–475. 559. Weiden PL, Bean MA, Schultz P. Perioperative blood transfusion
540. Von Roenn J, Harris JE, Braun DP. Suppressor cell function in does not increase the risk of colorectal cancer recurrence. Cancer
solid-tumor cancer patients. J Clin Oncol 1987; 5:1; 150–159. 1987; 60:870–874.
541. Voogt PJ, van de Velde CJH, Brand A, et al. Perioperative blood 560. Wells SA Jr, Burdick JF, Christiansen C, et al. Demonstration of
transfusion and cancer prognosis: Different effects of blood tumor-associated delayed cutaneous hypersensitivity reactions in
transfusion on prognosis of colon and breast cancer patients. lung cancer patients and in patients with carcinoma of the cervix.
Cancer 1987; 59:836–843. Natl Cancer Inst Monogr 1973; 37:197–203.
542. Vose BM, Vanky F, Argrov S, Klein E. Natural cytotoxicity in 561. Wells SA Jr, Burdick JF, Christiansen C, et al. Demonstration of
man: Activity of lymph node and tumor-infiltrating lymphocytes tumor-associated delayed cutaneous hypersensitivity reactions in
and regional lymphocytes. Eur J Immunol 1977; 7:753–757. lung cancer patients and in patients with carcinoma of t he cervix.
543. Wagner DK, Kiwanuka J, Edwards BK, et al. Soluble interleu- Natl Cancer Inst Monogr 1973; 37:197–203.
kin-2 receptor levels in patients with undifferentiated and lym- 562. Wells SA Jr, Burdick JF, Joseph WL, et al. Delayed cutaneous
phoblastic lymphomas: Correlation with survival. J Clin Oncol hypersensitivity reactions to tumor cell antigens and to nonspe-
1987; 5:8; 1262–1274. cific antigens. J Thorac Cardiovasc Surg 1973; 66:557–562.
544. Wagner V, Janku O, Wagnerora M, et al. The production of com- 563. Weppelmann B, Monkemeir D. The influence of prostaglandin
plete and incomplete antibodies in patients with neoplastic dis- antagonists on radiation therapy of carcinoma of the cervix.
ease. Neoplasma 1972; 19:75–87. Gynecol Oncol 1984; 17:196–199.
545. Wakasugi H, Kasahara T, Minato N, et al. In vivo potentiation of 564. Weese J, Oldham RK, Tormey DC, et al. Immunologic monitor-
human natural killer cell activity by a streptococcal preparation: ing in carcinoma of the breast. Surg Gynecol Obstet 1977; 145:
Interferon and interleukin-2 participation in the stimulation with 209–218.
OK-432. J Natl Cancer Inst 1982; 69:807–812. 565. Wesselius LJ, Wheaton DL, Manahan-Wahl LJ, et al. Lymphocyte
546. Walter RJ, Danielson JR. Defective monocyte chemotaxis in subsets in lung cancer. Chest 1987; 91:5; 725–729.
patients with epidermoid tumors of the head and neck. Arch 566. White JE, Chen T, Reed R, et al. Limited squamous-cell carci-
Otolaryngol 1985; 11:530–540. noma of the lung: A Southwest Oncology Group randomized
547. Wanebo HJ, Rao B, Miyazawa N, et al. Immune reactivity in pri- study of radiation with or without doxorubicin and with or without
mary carcinoma of the lung and its relation to prognosis. J Thorac levamisole immunotherapy. Cancer Treat Rep 1982; 66:113–120.
Cardiovasc Surg 1976; 72:339–347. 567. Whitehead RH, Thatcher J, Teasdale C, et al. T- and B-lymphocytes
548. Wanebo HJ, Pace R, Hargett S, et al. Production of and response in breast cancer: Stage relationship and abrogation of T-lymphocyte
to interleukin-2 in peripheral blood lymphocytes of cancer depression by enzyme treatment in vitro. Lancet 1976;
patients. Cancer 1980; 57:656–662. 1:330–333.
Robert K. Oldham 145

568. Wilkins SA Jr, Olkowskii ZL. Immunocompetence of cancer Rosenfeld C, Daniels JC, Saunders JP, eds. Current concepts in
patients treated with levamisole. Cancer 1977; 39:487–493. human immunology and cancer immunomodulation. Amsterdam:
569. Windle R, Bell PR, Shaw D. Five-year results of a randomized Elsevier Biomedical, 1982:471–478.
trial of adjuvant 5-fluorouracil and levamisole in colorectal 576. Yokoyama H, Umeda T, Kobayashi K, et al. Restoration of
cancer. Br J Surg 1987; 74:7; 569–572. E-rosette formation by bestatin in patients with bladder cancer. C
570. Windle R, Wood RFM, Bell PRF. The effect of levamisole on Cancer Res Clin Oncol 1980; 98:195–201.
post-operative immunosuppression. Br J Surg 1979; 66:507–509. 577. Young RC, Corder MP, Haynes HA, DeVita VT. Delayed hyper-
571. Wleklik MS, Luczak M, Najjajr VA. Tuftsin-induced tumor sensitivity in Hodgkin’s disease. Am J Med 1972; 52:63–72.
necrosis activity. Mol Cell Biochem 1987; 75:2; 169–174. 578. Zacharski LR, Linman JW. Lymphocytopenia: Its causes and
572. Wolf GT, Lovett EJ III, Peterson KA, et al. Lymphokine pro- significance. Mayo Clin Proc 1971; 46:168–173.
duction and lymphocyte subpopulations in patients with head 579. Zamkoff KW, Reeves WG, Paolozzi FP, et al. Impaired interleukin
and neck squamous carcinoma. Arch Otolaryngol 1984; regulation of the phytohemagglutinin response in Hodgkin’s dis-
110:731–735. ease. Clin Immunol Immunopathol 1985; 35:111–124.
573. Wrzesinki C, et al. Hematopoietic stem cells promote the expan- 580. Zamkoff KW, Reeves WQ, Paolozzi FP, et al. Impaired interleukin
sion and function of adoptively transferred antitumor CD8(+) T regulation of the phytohemagglutinin response in Hodgkin’s
cells. J Clin Invest 2007; 117:492–501. disease. Clin Immunol Immunopathol 1985; 35:111.
574. Wybran J, Govaerts P. Levamisole and human lymphocyte sur- 581. Zanker KS, Blumel G, Lange J, et al. Coumarin in malignant
face markers. Clin Exp Immunol 1977; 27:319–321. melanoma patients: An experimental and clinical study. Drug
575. Wybran J, Serrou B, Belpomme D, et al. Immunomodulating of Exp Clin Res 1984; 10:767–774.
T-cell and NK function by NPT-15392 in cancer patients. A coop- 582. Zou W. Regulatory T. Cells, tumour immunity and immunotherapy.
erative study by the tumor immunology group. In: Serrou B, Nat Rev Immunology 2006; 6:295–307.
7 Cancer vaccines
KENNETH A. FOON AND MALEK M. SAFA

Introduction gens on the surface of the cells being recognized (Fig. 1)


[1–3]. The proteins from which the peptides are derived
Immune approaches to the therapy of cancer have sub- maybe cell surface or cytoplasmic proteins [4, 5]. MHC
stantially evolved over recent years, from treating antigens are highly polymorphic, and different alleles
patients with nonspecific immune stimulants to a focus have distinct peptide-binding capabilities. The sequenc-
on the use of tumor-associated antigens (TAAs) either ing of peptides derived from MHC molecules has led to
by passive immune therapy with antibodies targeted the discovery of allele-specific motifs that correspond to
directly to tumor cells or by active immune therapy via anchor residues that fit into specific pockets on MHC
vaccinations with tumor cells, tumor cell lysates, pep- class I or II molecules [6, 7].
tides, carbohydrates, gene constructs encoding proteins, There are two T lymphocyte, helper and cytotoxic,
or anti-idiotype antibodies that mimic TAAs. which recognize antigens through a specific TCR com-
posed of both α and β subunits arranged in close conjunc-
tion to the CD3 molecule, which is responsible for
Approaches to Cancer Vaccines signaling. CD4 helper T cells secrete cytokines and lym-
phokines that enhance immunoglobulin production as
Specific active-immunotherapy differs from nonspecific well as activate CD8 CTLs. CD4 helper T cells are acti-
immune-based therapies such a BCG in that the goal is vated by binding via their TCR to class II molecules,
not general but rather specific activation of the immune which contain 14–25 amino acid (mer) peptides in their
system to eliminate tumor cells and not affect surround- antigen-binding cleft [8–10]. Extracellular proteins are
ing normal tissue. Theoretically, specific immunotherapy endocytosed and degraded (exogenous processing into
through vaccines activates a unique lymphocyte (B-and/ 14–25 mer peptides in endocytic compartments (acidified
or T-cell) response, which has an immediate antitumor endosomes) ) and bind to newly synthesized MHC class II
effect as well as memory response against future tumor molecules. The MHC peptide complex is transported to
challenge. The first and most obvious type of vaccines is the cell membrane, where it can be recognized by specific
autologous or allogeneic tumor cell preparations (Table 1). CD4 helper T cells. In most cased the MHC class II anti-
Alternatively, membrane preparations from tumor cells gen-containing peptide is presented to the CD4 helper T
have been used. In either instance these vaccines have cells by a specialized cell called an antigen presenting cell
been combined with a variety of cytokines. Gene- (APC). More specifically, a variety of cells are capable of
modified tumor cells expressing antigens designed to processing and presenting exogenous antigen including B
increase immunogenicity or gene modified to secrete cells, monocytes, macrophages, and the bone marrow
cytokines have been a valuable tool for vaccination. In derived derndritic cells (DC). DCs are the most efficient
addition, increase in our knowledge of TAA biology has APCs and express high levels of MHC class I and II mol-
led to the use of purified TAAs, DNA-encoding protein ecules, costimulatory molecules such as CD80 and CD86,
antigens, and/or protein-derived peptides. All of these and specific markers such as CD83. After antigen uptake,
approaches are being tested in the clinic. DCs migrate peripherally to lymph nodes, where antigen
Mechanistically, the ultimate aim of a vaccine is to presentation to CD4 helper T cells takes place [11, 12].
activate a component of the immune system such as There are two types of CD4 helper T cell capable of
antibodies or lymphocytes against TAAs presented by generating either an antibody-or a cell-mediated immune
the tumor (Table 2). Antibodies must recognize antigens response. This is based on the type of signaling they
in the native protein state at the cell surface. Once receive. Th1 CD4 helper T cells stimulate cell-mediated
bound, these molecules can mediate ADCC or comple- immunity by activating CTLs thought the release of lym-
ment-mediated cytotoxicity. T lymphocytes, on the phocytokines such as IL-2. Th2 CD4 helper T cells medi-
other hand, recognize proteins as fragments or peptides ate an antibody response through the release of
of varying size, presented in the context of MHC anti- lymphocytokines such IL-4 and IL-10. In some instances

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 147
© Springer Science + Business Media B.V. 2009
148 Cancer vaccines

Table 1. Tumor vaccines the generation of one type of response may serve to inhibit
Autologous tumor vaccines the generation of the other (ref. 13, i.e., IL-10 secretion by
Allogeneic whole cell vaccines Th2 helper T cells inhibits the generation of CTLs).
Dendritic cell vaccines CD8-positive CTLs are activated in most cases by pep-
Viral oncolysates tides derived from intracellular proteins that are cleaved
Polyvalent shed antigen vaccines
Peptide vaccines to 9–10 mer peptides in the cytosol of tumor cells or
Anti-idiotype vaccines APCs by proteosomes. The peptides are then transported
Genetically modified vaccines via specialized transporter molecules called TAP proteins
Recombinant viral vaccines to the endoplasmic reticulum, where they become associ-
DNA vaccines
ated with newly synthesized MHC class I molecules [14].
The complex is then transported via the Golgi apparatus
to the cell surface membrane, where the complex is rec-
Table 2. Characteristics of different vaccines
ognized by CD8 cytotoxic T cells via a specific TCR. Any
Response characteristics endogenously processed protein can be presented to the
Multiple Single Antibody T cell immune system in this way. Several reports suggest a
Vaccine antigens antigen response response subset of APCs can present exogenously processed pro-
Autologous cells + + + + teins on MHC class I molecules to CTLs [15–19].
Allogeneic cells + − + +
Shed antigens + − + +
Carbohydrate − + + −
Peptide − + + + Pitfalls in Developing Cancer
Anti-idiotype − + + +
antibody Vaccines
Dendritic cell NA NA + +
DNA − + + +
Tumor cells have developed a variety of mechanisms to
escape immune surveillance. DC’s are actively inhibited
+, present; −, absent; NA, not applicable in the tumor milieu. Both immature and defective DCs
Vaccine (cells, lysates, protein, peptide, cDNA)

14 25mer
peptide CD4 T -cell
TCRS
Antigen Presenting Cell CD
(APC)
MHC IL - 2
Class II
TC
CD R
Exogenous protein S IL - 4
from lysed tumor 9 10 mer
MHC
peptide CD8 T -cell IL - 5
Class
IL - 10
R
TC S
CD
Tumor cell
undergoing lysis
Tumor cell

B -cell

Figure 1. T cell activation: T cells recognize antigens as fragments of proteins (peptides) presented with major histocompatibility
complex (MHC) molecules on the surface of cells. The antigen-presenting cell processes exogenous protein from the vaccine or
from the lysed tumor cell into a peptide, and presents the 14/25-mer peptide to CD4 helper T cells on a class II molecule. There
are also data to suggest that exogenous proteins can be processed into 9/10-mer peptides that may be presented on MHC class I
molecules to CD8 cytotoxic T cells. Activated Th1 CD4 helper T cells secrete Th1 cytokines such as IL-2 that up-regulate CD8
cytotoxic T cells. Activated Th2 CD4 hepler T cells secrete Th2 cytokines such as IL-4, IL-5, and IL-10 that activate B cells
Kenneth A. Foon and Malek M. Safa 149

are described in a variety of tumors. In addition, DC’s antigen 4 (CTLA4) on T-reg cells has also been tested to
that present tumor antigens, may fail to reach the T cells enhance the anti-tumor immunity to vaccines [29].
in lymph nodes that generate active immune responses Cyclophosphamide has been used for many decades to
against tumors. The immune response may be skewed boost immune response [30–32], as have other chemo-
toward a Th2 response or T cells may be anergic. Immune therapy agents [33, 34].
regulatory cells may contribute to immune tolerance.
CD4 positive T cells (T-reg) with a high affinity receptor
for CD25 that co-express the intracellular marker Foxp3
also play an important role in immune tolerance [20,
Cancer Prevention Vaccines
21]. Mutation or down regulation of immunodominant A major success story in cancer vaccinology is cancer
tumor antigens, MHC molecules, or molecules involved prevention targeting the human papillomavirus (HPV)
in the antigen processing machinery may also in part to prevent cervical cancer. Cervical cancer is the second
explain the escape of tumor cells from immune recogni- most common cancer in women responsible for over
tion [22–24]. Down regulation or mutation of pro-apop- 250,000 deaths annually worldwide. Seventy percent of
totic molecules and expression of anti-apoptotic cervical cancers are caused by the two most common
molecules may also render tumor cells resistant to apop- oncogenic HPV types, HPV-16 and HPV-18, while
tosis. Tumor cells may acquire mechanisms that may another 10% are caused by HPV-45 and HPV-31 [35].
actively contribute to immune tolerance. For instance, The Food and Drug Administration has approved the
Fas ligand (FasL) – expressing tumors can deliver an cancer prevention vaccine HPV-16/18/6/11 (Gardisil,
apoptotic signal to activated T cells and natural killer Merck & Co. Inc, Whitehouse Station, NJ). This vaccine
(NK) cells expressing Fas receptor. The tumor micro- uses recombinant DNA technology to develop subunit
environment may also contain soluble factors that vaccines which include only the epitopes from the
inhibit T cell function. Factors such as TGF-beta, pros- pathogen recognized by the immune system. Copies of
taglandins, IL-10, and catabolizing enzymes are pro- the L1 viral capsid protein, the same protein which anti-
duced by tumor cells themselves or by stromal cells that bodies are generated against in the natural immune
may lead to T cell hyporesponsiveness. Countering these response to HPV, spontaneously self-assemble into non-
various tumor escape mechanisms is a key component to infectious virus-like particles which are used as the antigen
successful vaccine therapy. in the prophylactic vaccine. The vaccine is formulated
with ASO4 (aluminum hydroxide and monophosphoryl
lipid A) adjuvant [36, 37]. Only half the women with
HPV infection develop protective immunity because the
Mechanisms to Improve natural infection by HPV evades detection of the
the Immune Response immune system. Clinical studies demonstrated seropos-
itivity in women who receive the HPV 16/18/6/11 vaccine
Potent adjuvants improve the effectiveness of vaccines was 100% for HPV-16 and HPV-18 1 month after vac-
by accelerating the generation of immune responses and cination and remained at 100% 4.5 years after vaccination
sustaining responses for extended periods of time. [38–40]. Efficacy against cervical intraepithelial neo-
Commonly used adjuvants such as alum or Freund’s are plasia associated with HPV-16 was 100%. Of course,
effective in elevating antibody titers but do not elicit many women are already infected with HPV and will
significant Th1 responses or activate cytolytic T lym- develop cervical intraepithelial neoplasia and remain at
phocytes (CTL’s). The current focus is on the generation risk for developing cervical carcinoma. A number of HPV
of adjuvants that are designed to specifically elicit cel- based cervical vaccines are in development that are designed
lular immune responses. Toll-like receptors (TLRs) are to eliminate HPV induced disease after infection.
known to be involved in the initiation of immune
responses. CpG stimulates TLR9 and has been used
with vaccines to augment immune responses [25]. TLR8
may be involved in the activity of T-reg cells. Strategies
Therapeutic Cancer Vaccines
aimed at TLR8 are proposed to neutralize T- reg cells There has not yet been a therapeutic cancer vaccine
[26]. One approach to decrease the role of T-reg cells is approved by the US FDA, however, there are a number
to use immunotoxin directed against the high affinity of success stories and near success stories that should lead
IL-2 receptor [27, 28]. Monoclonal antibodies specific to approval in the near future. Melacine is composed of
for the negative regulatory signals mediating the CTL lyophilized melanoma lysates from two melanoma cells
150 Cancer vaccines

lines and the adjuvant Detox. In a phase 3 trial of 604 Table 3. Selected therapeutic vaccines in phase 3 development
patients with resected stage 3 melanoma, patients were Vaccine Company Indication
administered Melacine and low dose interferon alpha-
Antigen-loaded
2b versus high dose interferon alpha-2b [41]. Patient’s dendritic cells
were stratified by sex and number of nodes and ran- Sipuleucal-T Dendreon Corp Prostate
domly assigned to receive either 2 years of treatment DCVax-prostate Northwest Prostate cancer
with Melacine and low dose interferon alpha-2b or high Biotheraputics
Inc
dose interferon alpha-2b alone for 1 year. The median
Monified tumor cells
overall survival exceeded 84 months on the melancine OncoVax-CL Intracel Corp Colon cancer
low dose interferon alpha-2b (arm 1) versus 83 months GVAX Cell Genesys Inc Prostate cancer
in the high dose interferon alpha-2b (arm 2) (p = 0.56). Idiotype vaccines
Five year overall survival was 61% in arm 1versus 57% Favid Favrille Inc Follicular
lyphoma
in arm 2 and estimated 5 year relapse-free survival was BiovaxID Biovest Follicular
50% in arm 1 versus 48% in arm 2 with median relapse- International Inc lyphoma
free survival times of 58 months in arm 1 and 50 months MyVax Genitope Corp Follicular
in arm 2. Overall survival and relapse-free survival were lymphoma
clearly indistinguishable in the two arms. The incidence Viral vector
INGN-201 Introgen Head and neck
of neuropsychiatric severe adverse experiences were Therapeutics Inc cancer
similar, although they were more severe in the high dose TroVax Oxford BioMedica Renal cell cancer
interferon alpha-2b arm. The primary aim of this study Peptides
was that Melacine plus low dose interferon alpha-2b TV-1001 GemVax AS Pancreatic
would prolong overall survival compared with high dose (telomerase) cancer
BLP-25 (MUC1) Merck KGA Non-small cell
interferon alpha-2b. Unfortunately, this primary aim lung cancer
was not met, and their results failed to demonstrate Carbohydrate
rejection of the null hypothesis, with nearly identical GMK Progenics Melanoma
survival curves. Melacine was approved in Canada Pharmaceuticals
Inc
based on quality of life improvements.
DC vaccines are an attractive approach to vaccine
therapy although they are labor intensive requiring
prostate cancer. In the first study, patients were treated
unique autologous DC preparations from individual
at two dose levels with median survival at the low-dose
patients (Table 3). Sipuleucel-T and DCVax-Prostate
of 24 months and at the high dose of 35 months. A second
are both vaccines based on DC’s engineered to present
study used five vaccine doses. The median survival at
T-cell antigens associated with prostate cancer [42, 43].
the low and middle doses was 23 months and 20 months
Sipuleucel-T consists of autologous DC and a fusion
respectively and was not yet reached at the high dose
prostatic acid of prostates and phosphatase and granulo-
(>29 months). Phase 3 trials are ongoing.
cyte macrophage colony stimulating factor (GM-CSF).
There are a number of non-cell based patient specific
A randomized phase 3 placebo controlled trial in patient
vaccines that are currently in phase 3 trials. Favid,
with metastatic asymptomatic androgen-independent
BiovaxID and MyVax are idiotype vaccines that use KLH
prostate cancer was completed. Eighty-two patients
as a carrier. All three are patient specific for patient’s with
were randomized in a 2:1 ratio [44]. The primary end-
B-cell lymphoma. Anti-idiotype immune responses have
point of this study which was progression-free survival
been shown to correlate with better clinical outcomes in
was not reached (p = 0.052). However, overall survival
follicular lymphoma patients who received idiotype vac-
was significantly different for vaccine (26 months) ver-
cines [47, 48]. MyVax is also being studied in phase 2
sus placebo (21 months) with a hazard ratio of 1.7 and p
trials for mantle cell lymphoma, diffuse large B-cell lym-
= 0.01. A second study showed a trend toward increased
phoma and chronic lymphocytic leukemia.
survival (19 months versus 16 months) but did not reach
INGN-201 is a recombinant adenovirus-p53-based
significance. A phase 3 clinical trial using survival as an
vaccine for head and neck cancer. The adenovirus theo-
end point is ongoing.
retically functions to deliver the p53 protein in large
GVAX consists of two irradiated allogeneic prostate
quantities to the tumor cells. While this is classified as a
cancer cell lines engineered to secrete GM-CSF [45,
gene therapy, there is evidence that suggests an immune
46]. Two phase 2 trials have been completed in patients
response is elicited by p53 which is overexpressed in
with asymptomatic metastatic androgen-independent
Kenneth A. Foon and Malek M. Safa 151

endocytosis of anti-Id by APC

processed to processed to
9/10 mer peptides 15/25 mer peptides

MHC class II
MHC class I+
peptide MHC class I MHC class II+
CD8 peptide
APC TCR
TcR
IL-2r
CD4

CD8 T cell Th1


CD4
cytokines
T cell
clonal proliferation (IL-2, IFN-γ)
TCR
Mφ Th2 CD4 MHC class II+
NK cytokines peptide
T (IL-4, IL-10)
direct ADCC
Activated lysis B cell recognition
CD8 T cell TcR MHC of anti-Ιd and
processed to
class II endocytosis of anti-Ιd
15/25 mer
CD8
peptides
MHC class I+ Anti-anti-
peptide Id (Ab11) B Cell
Granzymes
Preforin
INF-γ
TNF-β Tumor cell mediates ADCC and
direct anti-metastatic effect

Figure 2. Putative immune pathways for anti-id vaccines

the tumor. Therefore, p53 may be considered a tumor- [51]. This vaccine is in phase 3 trials for melanoma.
associated antigen and may aid in the elimination of BLP-25 is a liposome-encapsulated synthetic peptide
tumor cells [49]. Interestingly, an adenovirus-p53 gene that corresponds to the variable number 10 tandem
therapy vector (Gendicine) has been approved in China repeat region of the mucin (MUC)-1 molecule. MUC-1
for head and neck cancer. TroVax is a vaccine in which is overexpressed and underglycosylated on tumor cells.
the tumor associated antigen 5T4 is expressed in a mod- The MUC-1 target is highly represented on most epithe-
ified vaccinia Ankara (MVA) vector, which induces lial tumors. There is currently a phase 3 trial in non-small
strong immune response similar to the live virus. It is cell lung cancer using BLB-25.
expected that the recombinant 5T4 antigen will be
included in the antiviral immune response. This vaccine
is in phase 3 trials for renal cell cancer. TV-1001 is a
telomerase peptide based vaccine that is in phase 3 trials
Conclusion
for pancreatic cancer. Telomerase is over expressed in The platform for therapeutic vaccines is broad including
many cancers and is theoretically a good target antigen. a variety of antigens, both non specific antigens repre-
GMK is a ganglioside conjugate vaccine in which gan- sented by whole cell based vaccine approaches and
glioside GM2 is coupled to KLH and formulated with recombinant antigens as represented by the protein and
QS-21 adjuvant [50]. The goal of this vaccine is to virus based approaches. DC’s are an extremely appeal-
induce an antibody response rather than a T-cell response ing vaccine approach, however, they are limited by the
152 Cancer vaccines

difficulties associated with patients specific cell therapies. 19. Rock KL, Rothstein L, Gamble S. Characterization of antigen-
No specific approach to vaccine therapy has stood out as presenting cells that present exogenous antigens in association
with calls I MHC molecules. J Immonol (1993) 150:438–46.
clearly superior. Strategies to enhance the immune response 20. Tanaka H, Tanaka J, Kjaargaard J, Shu S. Depletion of CD4 +
will be the next most important step in therapeutic can- CD25 + regulatory cells augments the generation of specific
cer vaccines. Monoclonal antibodies inhibiting T-regs, immune T cells in tumor-draining lymph nodes. J Immunother
the use of a variety of cytokines and TLR stimulation (2002) 25(3):207–17.
21. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady
are among the strategies that will be employed.
G, Wahl SM. Conversion of peripheral CD4 + CD25- naïve T cells
to CD4 + CD25 + regulatory T cells by TGF-β induction of tran-
scription factor Foxp3. J exp Med (2003) 198(12):1875–86.
References 22. Pawelec G, Koch S, Griesemann H, Rehbein A, Hahnel K,
Gouttefangeas C. Immunosenescence, suppression and tumour
1. Brodsky FM, Guafgliardi LE. The cell biology of antigen process- progression. Cancer Immunol Immunother (2006) 55:981–86.
ing and presentation and presentation. Annu Rev Immunol (1991) 23. Garrido F, Carera T, Concha A, Glew S, Ruiz-Cabello F, Stern PL.
9:707–44. Natural history of HLA expression during tumour development.
2. Osanto S. Vaccine trials for the clinician: prospects for tumor anti- Immunonol Today (1993) 14(1):491–99.
gens. Oncologist (1997) 2:284–99. 24. Garrido F, Ruiz-Cabello F, Cabrera T, Perez-Villar JJ, Lopez-Botet
3. Parham P. Antigen processing. Transporters of delight. Nature M, Duggan-Keen M, Stern PL. Implications for immunosurveil-
(Lond.) (1900) 348:674–5. lance of altered HLA class I phenotypes in human tumours.
4. Monaco J. A molecular model of MHC class-I-restricted antigen Immunol Today (1997) 18(2):89–95.
processing. Immunol Today (1992) 13:173–9. 25. Speiser DE, Lienard D, Rufer N, Rubio-Gudoy V, Rimoldi D,
5. Townsend SE, Allison JP. Tumor rejection after direct constimula- Lejeune F, Krieg AM, Cerottini JC, Romero P. Rapid and strong
tion of CD8+ T cells by B7-transfected melanoma cells. Science human CD8 + T cell responses to vaccination with peptide, IFA,
(Washington DC) (1993) 259:368–70. and CpG oligodeoxynucleotide 7909. J Clin Invest (2005)
6. Falk K, Rotzchke O. Consensus motifs and peptide ligands of 115(3):739–746.
MHC class I molecules. Semin Immunol (1993) 5:81–7. 26. Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, Wang DY, Li Y,
7. Falk K, Rotzchke O, Stevanovic J. Allele-specific motifs revealed Wang HY, Wang RF. Toll-like receptor 8-mediated reversal of CD4
by sequencing of self-peptides eluted from MHC molecules. + regulatory T cells. J Clin Invest (2005) 309(5739):1380–84.
Nature (Lond.) (1991) 351:290–4. 27. Dannull J, Su Z, Rizzieri D, Yang BK, Coleman D, Yancey D,
8. Kast WM, Bronklhorst AM, DeWaal LP, Melief CJ. Cooperation Zhang A, Dahm P, Chao N, Gilboa E, Vieweg J. Enhancement of
between cytotoxic and helper T lymphocytes in protection against vaccine-mediated antitumor immunity in cancer patients after
lethal Sendai virus infection. J Exp Med (1986) 164:723–38. depletion of regulatory T cells. J Clin Invest (2005)
9. Keene J, Forman J. Helper activity is required for the in vitro genera- 115(12):3623–33.
tion of cytotoxic T lymphocytes. J Exp Medi (1982) 150:1134–42. 28. Attia P, Maker AV, Haworth LR, Rogers-Freezeer L, Rosenberg
10. Raulet D, Bevan M. Helper T cells for cytotoxic T lymphocytes SA. Inability of a fusion protein of IL-2 and diptheria toxin
need not be region restricted. J Exp Med (1982) 155:1766–84. (Denileukin Diftitox, DAB389IL-2, ONTAK) to eliminate regula-
11. Allison JP, Hurwitz AA, Leach Dr. Manipulation of co-stimulatory tory T lyphocytes in patients with melanoma. J Immunother (2005)
signals to enhance antitumor T-cell responses. Curr Opin Immunol 28(6):582–92.
(1995) 7:682–6. 29. Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL,
12. Tai T, Paulson JC, Cahan CD et al. Ganglioside CM2 as a human Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA, Freezer
tumor antigen (OFA-I-1). Proc Natl Acad Sci USA (1983) LJ, Morton KE et al. Cancer regression and autoimmunity induced
80:5392–6. by cytotoxic T lymphocyte-associated antigen 4 blockade in
13. Fitch FW, Laneki DW, Gajewski TF. T-cell mediated immune regu- patients with metastatic melanoma. Proc Natl Acad Sci USA
lation: help and suppression. In: Paul W, Nita E, eds. Fundamental (2003) 100(14):8372–77.
Immunology. New York: Raven Press (1993) 733–61. 30. MacLean GD, Miles DW, Rubens RD, Reddish MA, Longenecker
14. Zinkernagel R, Doherty P. MHC-restricted cytotoxic cell: studies BM. Enhancing the effect of THERATOPE STn-KLH cancer vac-
on the biological role of polymorphic major transplantation anti- cine in patients with metastatic breast cancer by pretreatment with
gens determining T cell restriction-specificity, function and low-dose intravenous cyclophosphamide. J Immunother with
responsiveness. Adv Immunol (1979) 27:51–106. Emphasis Tumor Immunol (1996) 19(4):309–316.
15. Grant EP, Rock KL. MHC class I-restricted presentation of exog- 31. Glaser M. Regulation of specific cell-mediated cytotoxic response
enous antigen by thymic antigen-presenting cells in vitro and in against SV40-induced tumor associated antigens by depletion of
vivo. J Immonul (1992) 148:13–18. suppressor T cells with cyclophosphamide in mice. J Exp Med
16. Harding CV, Song R. Phagocytic processing of exogenous particu- (1979) 149(3):774–779.
late antigens by macrophages for presentation by class I MHC 32. Mitchell MS, Kan-Mitchell J, Kempf RA, Harel W, Shau HY, Lind
molecules. J Immunol (1994) 153:4925–33. S. Active specific immunotherapy for melanoma: phase 1 trial of
17. Kovacsovic-Bankowski M, Rock KL. A phagosome-to-cytosol allogeneic lysates and a novel adjuvant. Cancer Res (1988)
pathway for exogenous antigens presented on MHC class I mole- 48(20):5883–93.
cules. Science (Washington DC) (1995) 267:243–5. 33. Velu T, Ramlau R, Quoix E, Pawlicki M, Pless M, Lena H, Levy E,
18. Rock KL, Gamble S, Rothstein L. Presentation of exogenous anti- Krzakowski M, Limacher J-M, Bizouarne N. A phase II study
gen with class I major histocompatibility complex molecules. evaluating the clinical efficacy of TG4010 (MVA-MUC1-IL2)
Science (Washington DC) (1990) 249:918–21. in association with chemotherapy in patients with non small cell
Kenneth A. Foon and Malek M. Safa 153

lung cancer. American Society of Clinical Oncology, Orlando, FL 42. Small EJ, Fratesi P, Reese DM, Strang G, Laus R, Peshwa MV,
(2005) Abs 7132. Valone FH. Immunotherapy of hormone-refractory prostate cancer
34. Nowak AK, Robinson BW, Lake RA. Synergy between chemo- with antigen-loaded dendritic cells. J Clin Oncol (2000)
therapy and immunotherapy in the treatment of established murine 18(23):3894–3903.
solid tumors. Cancer Res (2003) 63(15):4490–4496. 43. Knutson KL. Technology evaluation: DCVax, Northwest biothera-
35. Munoz N, Bosch FX, Castellsague X, Diaz M, de Sanjose S, peutics. Curr Opin Mol Ther (2002) 4(4):403–407.
Hammouda D, et al. Against which human papillomavirus types 44. Small EJ, Schellhammer PF, Higano CS, et al. Placebo-controlled
shall we vaccinate and screen? The international perspective. Int J phase III trial of immunologic therapy with Sipuleucel-T
Cancer (2004) 111:278–85. (ACP8015) in patients with metastatic, asymptomatic hormone
36. Giannimi SL, Hanon E, Moris P, Van Mechelen M, Morel S, Dessy refractory prostate cancer. J Clin Oncol (2006) 24:3089–94.
F, et al. Enhanced humoral and memory B cellular immunity using 45. Simons JW, Sacks N. Granulocyte-macrophage colony-stimulating
HPV 16/18 L1 VLP vaccine formulated with the MPL/aluminium factor-transduced allogeneic cancer cellular immunotherapy: the
salt combination (AS04) compared to aluminium salt only. Vaccine GVAX vaccine for prostate cancer. Urologic Oncology (2006)
(2006) 24:5937–49. 24:419–24.
37. Hilleman MR. Overview of vaccinology with special reference to 46. Michael A, Ball G, Quatan N, et al. Delayed disease progression
papillomavirus vaccines. J Clin Virol (2000) 19:79–90. after allogeneic cell vaccination in hormone-resistant prostate
38. Harper DM, Franco EL, Wheeler CM, Moscicki AB, Romanowski B, cancer and correlation with immunologic variables. Clin Cancer
Rotelli-Martins CM, et al. Sustained efficacy up to 4.5 years of a biva- Res (2005) 11:4469–78.
lent L1 virus-like particle vaccine against human papillomavirus types 47. Weng W-K, Czerwinski D, Timmerman J, Hsu FJ, Levy R. Clinical
16 and 18: follow up from a randomized control trial. Lancet (2006) outcome of lymphoma patients after idiotype vaccination is
376:1247–55. correlated with humoral immune response and immunoglobulin
39. Harper DM, Franco EL, Wheeler C, Ferris DG, Jenkins D, Schuind A, G Fc receptor genotype. J Clin Oncol (2004) 22:4717–24.
et al. Efficacy of a bivalent L1 virus-like particle vaccine in prevention 48. Inoges S, Rodriguez-Calivillo M, Zabalegui N, Lopez-Diaz de
of infection with human papillomavirus types 16 and 18 in young Cerio A, Villanueava H, Soria E, et al. Clinical benefit associated
women: a randomized controlled trial. Lancet (2004) 364:757–65. with idiotype vaccination in patients with follicular lymphoma.
40. Villa LL, Cost RL, Petta CA, Andrade RP, Ault KA, Giuliano AR, J Natl Cancer Inst (2006) 98:1292–301.
et al. Prophylactic quadrivalent human papillomavirus (types 6,11, 49. Tokunaga N, Murakami T, Endo Y, Nishizaki M, Kagawa S,
16, and 18) L1 virus-like particle vaccine in young women: a ran- Tanaka N, Fujiwara T. Human monocyte-derived dendritic cells
domized double-blind placebo-controlled multicentre phase II pulsed with wild-type p53 protein efficiently induced CTLs against
efficacy trial. Lancet Oncol (2005) 6:271–8. p53 overexpressing human cancer cells. Clin Cancer Res (2005)
41. Mitchell MS, Abrams J, Thompson JA, Kashan-Sabet M, DeConti 11(3):1312–18.
RC, Hwu W, Atkins MB, Whitman E, Ernstoff MS, Haluska FG, 50. Knutson KL. GMK(progenics pharmaceuticals). Curr Opin Invest
Jakowatz JG, Das Gupta TK, Richards JM, Samlowski WE, Drugs (2002) 3(1):159–164.
Costanzi JJ, Aronson FR, Deisseroth AB, Dudek AZ, Jones VE. 51. Chapman PB, Morrisey D, Panageas KS, Willimas L, Lewis JJ,
Randomized trial of an allogeneic melanoma lysate vaccine with Isreal RJ, Hamilton WB, Livingston PO. Vaccination with bivalent
low-dose interferon alfa-2b compared with high-dose interferon G(M2) and G(D2) ganglioside conjugate vaccine: a trial comparing
alfa-2b for resected stage III cutaneous melanoma. J Clin Oncol doses of G(D2)-keyhole limpet hemocyanin. Clin Cancer Res
(2007) 25(15):2078–85. (2000) 6(12):4658–62.
8 Cytokines
WALTER M. LEWKO AND ROBERT K. OLDHAM

Cytokines are regulatory proteins, produced and secreted book. Table 1 summarizes cell sources and effects of
by various cells, which control immune response, cytokines discussed.
hematopoiesis, inflammation, wound repair and tissue
morphogesis. Cytokines may be secreted or membrane
bound. Secreted cytokines may act locally as autocrine
or paracrine factors or over some distance as would a
Cytokine Receptors: Many
hormone. Membrane bound cytokines act by cell–cell Belong to Receptor Families
contact, communicating information from one cell to
Cytokines usually induce their effects by binding to
another, often bidirectionally. There are cell surface
target cell receptors, which generate intracellular signals.
receptors for each cytokine that bind the cytokine spe-
Often the result is gene activation. The receptors for the
cifically. Receptor subunits may be shared between dif-
various cytokines tend to be grouped into families
ferent cytokines. Binding the cytokine brings about
reflecting common genetic origin, structure, signaling
signaling and a series of cell activating events. For many
and cell response. Some of the major receptor groups
cytokine receptors (not all), this involves increased
include the IL-1 family, cytokine/hematopoietin recep-
phosphorylation of certain tryrosine residues on key cel-
tor family, the IL-6 family, the IL-17 family and the
lular proteins. Kinases are the enzymes that carry out
tumor necrosis factor family. For example, the receptors
phosphorylation. Receptors may themselves be kinases,
for IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-9, IL-13, IL-15,
which activate upon binding. More typically, the acti-
G-CSF, GM-CSF and Prolactin are all members of the
vated receptor may recruit cytosolic kinases, for exam-
hematopoietin receptor family. Receptors for these
ple, mitogen activated protein kinase (p38 MAPK)1 and
cytokines have been cloned and analyzed. Amino acid
Janus kinases (Jak1, Trk and Jak 3) [97, 533, 556, 1009,
sequence reveals segments that are similar between
1699]. As the kinases bind the receptor complex, their
members of the same family [1472]. The IL-6 family
enzymatic activities increase and an array of cellular
members share the gp130 signaling subunit [1943].
proteins is phoshorylated, in certain cases including the
Receptors are generally composed of two or more sub-
receptor itself. These modifications bring about changes,
units. In a basic model, one subunit binds the ligand; the
increases or decreases, in each protein’s activity. Among
other subunit generates the signal; together in a complex,
these proteins are the STAT proteins that control gene
the subunits bind the cytokine with higher affinity than
expression. When phosphorylated, STAT proteins trans-
they do separated. Different receptors may share subunits
locate from the cytoplasm to the nucleus and bind spe-
that carry out common functions. For example, several
cific enhancer segments allowing the expression of the
receptors (IL-2, IL-4, IL-7, IL-9, IL-13, IL-15, IL-21)
genes needed to bring about a cytokine’s response.
share a common gamma subunit (γc) that is involved in
In this chapter, several of the major cytokines will be
signaling. The importance of this gamma chain’s function
discussed. Regulation of cellular immunity will be empha-
is shown in various forms of combined immunodefi-
sized for its role in the elimination of tumor and virus
ciency disease that appear due to mutations [259, 1097,
infected cells. Inflammation is discussed for its important
1699]. Since this receptor subunit is shared by so many
role in not only in immune response but also cancer pro-
cytokines, these genetic defects have far reaching and
gression. Unfortunately, it is not possible to cover all of
debilitating results in the immune response system.
the interesting cytokine research in detail. We have tried to
Cytokine activity is of necessity transient. Uncontrolled
summarize past results and highlight certain interesting
activity may result in inflammatory diseases, allergy or
trends. The interferons, colony stimulating factors and
autoimmunity. There are several levels at which regula-
certain growth factors are covered in other chapters in this
tion may occur, from cytokine secretion to receptor bind-
ing to intracellular signaling to target cell responsiveness.
For example, there are cellular proteins that regulate
1
Abbreviations are listed at the end of the chapter. signaling [492, 1413, 1882, 2256]. Among them are the

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 155
© Springer Science + Business Media B.V. 2009
156 Cytokines

Table 1. Summary of cytokine sources and effects related to the growth and treatment of cancera
Cytokine Cell source Cells influenced Effectb
IL-1 Mono/Macro Macrophages Activation, TNF, IL-6, PGE2, NO (w/IFNγ)
B cells B cells Growth/Ig (w/IL-4, IL-6), chemotaxis
T cells T cells Growth (w/IL-2), migration, IL-2, IL-2R, IFNγ
NK Th2 Growth, IL-4, IL-5, IL-6
DC Mast cells IL-3, IL-4, IL-5, IL-6, IL-9 (Th2-like cytokines)
Some tumor cells LAK Induction (w/IL-2)
Fibroblasts NK IFNγ (w/1L-12), cytotoxicity (w/IFNγ)
Keratinocytes Tumor cells Growth/inhibition/metastasis/no effect
Endothelial Fibroblasts Growth, PGE2, collagen
Glial cells Chondrocytes ↓ Growth, ↓ collagen, ↑ protease
Eosinophils Keratinocytes Growth
Neurons Endothelial PGE2, VCAM, ICAM, angiogenesis
Epithelial Vascular smooth muscle Growth, IFNβ
Synovial Eosinophils Degranulation
Basophils Degranulation, histamine release
Liver Metabolism, protein production
Synovial PGE2, collagenase, phospholipase A2
Glial cells Growth
Neurons Survival
Pancreatic Islet Insulin
Muscle Negative protein balance (w/insulin, IL-1, TNF)
DC Maturation (w/GM-CSF)
Stem cells Viability/survival (w/IL-3, G-, M-, GM-CSF)
Hypothalamus Corticotrophin releasing factor
Some tumors ↓ Growth
Melanoma B16 ↑ Metastasis (↑ endothelial VCAM)
IL-2 T cells (active) NK Activation/cytotoxicity
LAK Activation/cytotoxicity
T cells Growth/cytotoxicity/tolerance/AICD
PBL Growth/CTL generation
B cells Growth/differentiation (w/IL-6, IL-12)
Macrophages Activation, TNFα, β, NO
DC Proliferation
Keratinocyte Proliferation
Eosinophils Eosinophilia
Tumor cells Growth/inhibition/no effect ↓ ICAM/↓ MHCI
IL-3 T cells (active) Stem cells Growth
Thymic epithelium Megakaryocyte Platelet production
Mast cells RBC precursors RBC production
Keratinocytes Mast cells Survival/growth (w/IL-4)
Neurons Natural cytotoxic Growth/TNF
Eosinophils Granulocytes Growth/survival/differ
Macrophages Growth/differ/activation
NK Activation (w/IL-2)
T cells Activation/growth (w/IL-2)
Osteoclasts ↑ or ↓
Hematopoietic tumor Growth stimulation/inhibition/no effect
IL-4 Th2 B cells Growth, Ig secretion (IgG, IgE), MHCII
Mast cells T cells Growth stimulation/inhibition/no effect
Basophils Th0 Differentiation to Th2, inhibition formation Th1
NK Mono/Macro Growth/activation/AP/HLA DR
↓ Inflammatory cytokines
Eosinophils Endothelial Growth/VCAM
DC Fibroblasts Growth
γδ T cells PBL Growth (IL2-primed)
NK T cells NK Growth/IL5 secretion (w/IL-2, IL-12)
Mast cells Growth/ICAM (w/IL-3)
LAK Activation (IL-2 primed)
DC Growth/activation (w/IL-2, IL-7, GM-CSF)
(continued)
Walter M. Lewko and Robert K. Oldham 157

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
Hematopoietic cells Growth (+/−)
Certain tumors ↓ Growth, ↓ COX-2, ↓ PGE2
IL-5 Th2 Eosinophils Growth/differ/survival/activity/chemotaxis
Eosinophils B cells Growth/survival/Ig (IGA, IgM)
Mast cells Mast cells Growth (w/other factors)
NK T cells Growth/differentiation (w/IL-2), IL-2R
LAK Activation (w/IL-2)
NK Activation (w/IL-2), IL-2R
IL-6 Fibroblasts B cells Differentiation/Ig (w/IL-2)/survival
Macrophages T cells Growth/activation (w/IL-2)
Epithelial Megakaryocytes Growth/platelets
Endothelial NK Activation/cytotoxicity
Eosinophils Hepatocytes Acute phase proteins, fibrinogen
Neutrophils Intestine cells Acute phase proteins
Mast cells Fibroblasts Growth/collagen
B cells Osteoclasts Growth/activation
Keratinocytes Endothelial Growth
Th2/CD8 T Neurons Regeneration
Osteoblasts Melanoma ↓ Growth
Synoviocytes Leukemia/lymphoma↓ ↓ Growth
growth
Megakaryocytes Breast cancer ↓ Growth
Langerhans cells Cervical cancer ↓ Growth
Astrocytes DC Antigen presentation, especially self antigen
DC
Some tumors
IL-7 Stroma, marrow Pre-B cells Growth/differ (w/SCF/FLT3L)
Stroma, thymus Pre-T cells Growth/differ/survival (w/SCF/FLT3L)
Keratinocyte Mono/macro Growth/activation/antitumor activity
B cells NK Activation
Intestinal epithelium LAK Activation
DC T cells (w/IL-2) Growth/different/survival/memory/cytotoxicity
Endothelial DC Growth/antigen presentation
Some carcinomas TIL Activation/proliferation
Leukemia/lymphoma Pre-eosinophils Growth
Melanoma ICAM
Some leukemia/lymp Growth
IL-8 Macrophages Neutrophils Chemotaxis, superoxide, degranulation,
hydrolase
Endothelial T cells Chemotaxis
Neutrophils Macrophages Chemotaxis
Eosinophils Endothelial Chemotaxis/angiogenesis
Mast cells Eosinophils Chemotaxis
Epithelial cells NK Chemotaxis
Fibroblasts Cancer Autocrine growth/metastasis/angiogenesis
Keratinocyte
Some melanomas
Some carcinomas
IL-9 Th2 T cells (active) Growth (w/IL2), IL-22
Mast cells Fetal thymocytes Growth (w/IL2)
Mast cells Survival, growth (w/IL-3, IL-4), IL-22
Pre erythroid Growth (w/erythropoietin)
B cells IgE
Lymphoma Growth/survival
IL-10 Th2, mice T cells ↓ Growth/IFNγ/cytotoxicity/chemotaxis.
Th1 Th2, humans NK ↓ Cytotoxicity/IFNγ
Tr1 Mono/macro ↓ Activivity/IL12/AP/collagenase; ↑ TIMP
nTreg Mast cells Growth (w/IL-3, IL-4)
B cells B cells Growth/differ/survival/MHCII (w/IL-2)
(continued)
158 Cytokines

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
Monocytes Activated B cells Apoptosis
Eosinophils Fibroblasts ↓ Collagen/fibrosis
Mast cells DC ↓ Antigen presentation
Keratinocyte
Some tumors
IL-11 Stroma, marrow Progenitor cells Growth/colony formation
Fibroblasts Megakaryocytes Thrombopoiesis (w/IL-3)
Epithelial B cells Growth
Chondrocytes Macrophages ↓ IL-12
Synovial Th2 ↓ IL-4, IL-5, IL-13
Smooth muscle Fibroblasts ↓ Differ to adipocytes
Liver Acute phase proteins
Synovial cells TIMP
Osteoclasts Growth/differentiation
IL-12 B cells NK Growth/cytotoxicity/IFN (w/IL-1)
Macrophages CD8 T cells Growth/cytotoxicity/IFN (w/IL-2)
DC Th1 IFNγ, IL-2, growth
Neutrophils Th2 ↓ IL-4, IL-10, growth
Microglial PBL LAK induction/CTL growth cytotoxicity
Keratinocytes TIL Growth/cytotoxicity
Macrophages IFNγ
B cell Growth/differentiation (w/IL-2)
DC Activation/antigen presentation, IFNγ
IL-13 Th2 cells Mono/macro ↓ Cytokines/NO/PGE2/CD14/ADCC
Mast cells ↑MHCII/CD11b c/IL-1RA/CD23
Basophils B cells Growth/differ/Ig/IgE switching/CD23
Eosinophils Neutrophils ↑ IL1Ra, ↓ CD14
DC NK IFNγ, cytotoxicity
Some B lines Endothelial VCAM/MCP-1/angiogenesis
Tumor cells DC Growth/maturation,↓ IL-23, IL-1, IL-6.
Th17 Suppression
Epithelial ↓ NO
Synovial ↓ IL-1β, TNF
Macrophages ↓ HIV production
Renal ca ↓ Growth
IL-14 CD8 T cells B cells Growth/differentiation/memory
Th1, Th2 B cell lymphoma Autocrine growth
NKT cells
Follicular DC
B lymphoma
IL-15 Epithelial T cells Growth/cytotoxicity/memory
Stroma, marrow γδ T cells Growth/activation
PBMC B cells Growth/IgG, IgA, IgM
Fibroblasts NK Growth/cytotoxicity/chemotaxis
Keratinocyte LAK Induction/cytotoxicity
TIL Growth
Mast cells Growth/response
Keratinocyte Growth; psoriasis
IL-16 CD8 T CD4 T cells Migration/growth w/IL-2, IL-15
Macrophages Mono/macro Migration/activation/antigen presentation
CD4 T cells DC Migration/antigen presentation/tolerance (w/TPO)
B cells Eosinophils Migration
Fibroblasts HIV ↓ Replication/↓entry
Eosinophils NK Cytokines/migration
Mast cells
DC (immature)
Epithelial
Brain
IL-17 Th17 Macrophages Inflammatory cytokines, IL-10, IL-12.
Neutrophils Fibroblasts Inflammatory cytokines, GM-CSF
(continued)
Walter M. Lewko and Robert K. Oldham 159

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
CD8+ T cells Epithelial Inflammatory cytokines
Endothelial Inflammatory cytokines
Keratinocyte Inflammatory cytokines, ICAM
DC Maturation/cytokine production
Chondrocytes Inflammatory cytokines/MMP/NO
Granulocytes Production (w/GM-CSF)
Synoviocytes IL-6, LIF
CH ovary cells Invasiveness
Monocytes Osteoclast differentiation
Osteoclasts Activation
IL-17B Pancreas Monocytes IL-1β, TNFα
Intestine Neutrophils Migration (indirect effect)
Stomach
IL-17C Prostate Monocytes IL-1β, TNFα.
Fetal kidney
IL-17F CD4 T cells Endothelial ↓ Angiogenesis. ↑ IL-2, TGFβ, MCP-1
Monocytes Bronchial epithelium ICAM, IL-6, IL-8 (neutrophil recruitment)
Basophils
Mast cells
IL-18 Macrophages Th1 (w/IL-2, IL-12) IFNγ, growth/differ/IL-2,
FAS/apoptosis/escape, IL-13
DC B cells IFNγ (w/IL-12), IgG
Kupffer cells NK (w/IL-2, IL-12) IFNγ, growth, anti tumor activity
FAS/apoptosis/escape, IL-13
Keratinocyte Macrophages IFNγ (w/IL-2)
Airway epithelium DC IFNγ (w/IL-2)
Adrenal cortex NK T cells IL-4
Neutrophils Degranulation, cytokines, CD11b (complement R)
Endothelial Migration/regulation of angiogenesis
IL-19 Monocytes Th2 Activation, IL-4
Keratinocytes Monocytes Activation, IL-6, TNF, ROS
PBMC PBMC IL-10, IL-19
IL-20 Keratinocytes Keratinocyte Proliferation, psoriatic character
Monocytes Hemato progenitors Proliferation
Glial cells Endothelium bFGF, VEGF, IL-8, angiogenesis
Synovial fibroblasts Inflammatory cytokines IL-6, IL-8
IL-21 Th2 NK Proliferation, activity
Th17 T cells, Th17 Proliferation, activity
Follicular T cells Treg Inhibition
B cells Class switching, ↑IgG, ↓ IgE
DC Regulation, ↓maturation, ↓activation
Hematopoietic tumor Growth stimulation, inhibition, or no effect
IL-22 Th17 Liver Acute phase proteins
Mast cells Th2 ↓ IL-4
Th1 Epithelium Inflammatory cytokines
Th2 Keratinocytes Inflammatory cytokines, migration, β-Defensin
NK Hepatocytes Survival, acute phase proteins
IL-23 DC CD4+ T memory Proliferation
Macrophages Th IFNγ, Il-17, IL-6, IL-1, TNF, ICOS
Keratinocytes DC IL-12, IFNγ
Macrophages IFNγ
Th17 Activation (IL-17)
IL-24 Th2 PBL Proinflammatory, IL-6, TNF
Monocytes (activ) Melanocytes Differentiation, ↓Proliferation
Melanocytes Keratinocytes Proliferation
PBMC (activ) Vasc sm muscle ↓Growth, ↓migration, ↓angiogenesis
Some tumor cells Monocytes TNF, IL-6
Tumors ↓Growth, ↑apoptosis, ↓angiogenesis,
↑radiosens
Endothelium ↓ Angiogenesis
(continued)
160 Cytokines

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
IL-25 Th2 Eosinophils Growth, infiltration, ICAM, L-selectin,
Eosinophils IL-6, IL-8, MCP-1, MIP-1α
Mast cells Th2 Activation (IL-4, IL-5, IL-13)
Microglial cells Th17 Regulation
Basophils Cartilage Catabolism
Fibroblasts (Lung) Inflammatory cytokines
IL-26 T cells Epithelial cells ICAM-1, IL-8, IL-10
NK
IL-27 DC T cells Proliferation, IL-12R, IFNγ
Monocytes (LPS) NK IFNγ
Th1 Differentiation, proliferation, activity
Macrophages Th17 ↓ Differentiation response to TFGβ/IL-6
Granulocytes ↓ ROS response to endotoxin
Macrophages ↓ ROS response to endotoxin
Endothelium ↓ Angiogenesis
IL-28A, B, IL-29 Monocytes Most cells Antiviral, oligoadenylate synthetase, MHCI/II
Macrophages DC Maturation, Treg induction
DC Tumor cells ↓Growth, ↑apoptosis, ↑ Immune response
IL-31 Th2 Th2 ↓ Activation (↓ IL-4, IL-5, IL-13)
Myeloid progenitors Survival
Monocytes Inflammatory cytokines
Epithelial cells Inflammatory cytokines
Skin epithelium Dermatitis
Lung epithelium EGF, VEGF, inflammatory cytokines
Sensory neurons Itching sensation, development
IL-32 NK cells Monocytes/DC TNFα, IL-1β, IL-6, IL-8, PGE2
T cells T cells Apoptosis, TNF
Epithelium
Monocytes
Synoviocyte (RA)
IL-33 Endothelium Th2 Activation (IL-4. IL-5, IL-13)
Fibroblasts Mast cells IL-8, IL-4, IL-5, IL-13, survival
Endothelium Nuclear protein/transcription repressor
Fibroblasts, heart ↓ Collagen
Cardiomyocytes ↓ Hypertrophy
IL-35 nTreg nTreg Proliferation, IL-10
CD4+CD25− Suppression
Th17 cells Suppression, ↓ IL-17
4-1BBL T Cells CD8 T/CD4 Th1 Proliferation/cytokines/↓ AICD/↓ apoptosis
DC CD4 Th2 Suppression/anergy
Monocytes/macro Monocytes/macro Activation/survival
B cells B cells (active) Proliferation/survival
Stromal cells NK/NKT IFNγ/IL-2
Some tumor cells DC IL-12/1l-6
T cells ↓ Activation (by reverse signal)
CD27L B cells B cells Differentiation (+/−), memory/apoptosis/AICD
T cells B cells Activation/ab production (by reverse signal)
NK T cells Stimulate or inhibit, memory/apoptosis/AICD
DC γδT cells Activation/cytotoxicity (reverse signal)
B cell malignancy NK Activation/cytotoxicity (w/IL-2)
Renal cancer cells
Brain tumor
CD30L Macrophages B cells Memory/↓ class switching, Ig prod (by rev signal)
B cells Neutrophils Oxidative burst/IL-8 (by reverse signal)
T cells T cells Proliferation/apoptosis/AICD, IL-6 (by rev signal)
Megakaryocytes Thymocytes Apoptosis/negative selection
Neutrophils NK Apoptosis/AICD
Eosinophils Mast cells Chemokines (rev signal)
(continued)
Walter M. Lewko and Robert K. Oldham 161

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
pre-erythrocytes Eosinophils Apoptosis
Some leukemias Lymphomas Apoptosis/no effect/growth
Some lymphomas
CD40L CD4 B cells Proliferation/class switch/Ig/survival
CD8 DC Activation/B7.1/B7.2/ICAM/IL-12, survival
Mast cells Macrophages Activation/proinflammatory cytokines
Basophils T cells Costimulation/activation/growth/longevity.
Fibroblasts NK Activation/cytotoxicity
Platelets Fibroblasts Activation/proinflammatory cytokines/ICAM/
VCAM
Endothelium ICAM/VCAM/Angiogenesis factors
CD4 IL-4 (by reverse signal)
CD8 Activation/cytotoxicity (by reverse signal)
Platelets RANTES/Inflammation
Some tumors Apoptosis/AP
FASL T cells T cells Apoptosis/AICD/Immune privilege
B cells B cells Apoptosis/AICD/Immune privilege
NK cells Some tumors Apoptosis; chemotherapy response
Some tumors Endothelium Damage (VLS)
DC (Immature) Some tumors Apoptosis
FLT3L Hematopoietic Hematopoietic SC Growth/survival
Non- Progenitor cells Growth/survival
hematopoietic DC Growth/maturation (w/SCF, GM-CSF, IL-4, TNFα)
B cells Growth (w/IL-7, SCF)
NK/LAK Growth of progenitors; response to IL-2
Osteoclasts Differentiation
Neurons Survival (w/NGF)
Neuron SC Growth regulation
Some leukemias Growth
Some tumors Growth (especially neural crest origin)
Interferon γ Th1 Cells in general MHCI/II, FcR, P1 Kinase,
CD8 T cells 2 -5 oligoadenylate synthetase
NK Macrophages Activation, H2O2
NK Cytotoxicity
B cells Altered Ig isotype secreted
Inhibition of IL-4-induced functions
CD8 T cells Differentiation/cytotoxicity (w/IL-2)
Th1 Growth/cytokine production
Th2 ↓ Growth/cytokine production
Th17 ↓ Differentiation
Endothelium ↓ Growth/angiogenesis
Tumor cells ↓ Growth; increased differentiation
↑ MHC expression; ↓ NK sensitivity
LIF Fibroblasts T cells Differentiation
Endothelium Colon Colitis; progression to colon cancer
Macrophages Mammary Development
Epithelium, thymus Osteoclasts Growth/activation
Synoviocytes Endometrium Implantation/trophoblast differentiation
Neurons Survival/↓ maturation
Corticotropes ↓ Growth↑ differentiation, ACTH secretion
Myeloid leukemia Differentiation/↓ growth
Glioma Differentiation/↓ growth
Some carcinomas Growth (breast, colon, renal, prostate)
Embryonic SC ↓ Differentiation
LIGHT T cells T cells Proliferation, IL-2/IFNγ/IL-4/TNFα
Granulocytes DC Costimulation for AP to T cells
Monocytes NK T cell help
DC (immature) Hepatocytes Proliferation
HVEM+ LTβR+ Apoptosis
(continued)
162 Cytokines

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
Some tumors Apoptosis
Lymphotoxin α Th1 B/T/DC etc. Lymphoid organogenesis
(TNFβ) CD8 T cells Endothelial VCAM/ICAM
B cells (early) NK T cells Growth/development
NK Osteoblasts Growth/activation
Astrocytes Osteoclasts Growth/activation
Lymphotoxin β T cells Lymphoid cells Lymphoid organogenesis
B cells T cells Survival
NK cells B cells Survival
DC (Immature)
Novel LN cells B cells Growth/IgM/IgE/IgG
Neurotrophin Spleen cells Macrophages Growth
Neurons Survival/development
Oncostatin M T cells LN cells T cell development/migration
Mono Fibroblasts Growth/wound repair/collagen/fibrosis
Neutrophils Osteoclasts Growth/activation
Certain tumors Vascular sm muscle Growth/wound repair
Synoviocytes Inflammation regulation (+/−)
Some gliomas Differentiation/↓ growth
Sarcoma (Kaposi) Growth
Osteopontin T cells Th Activation (favors Th1)
Mono/Macro DC Activation (favors Th1)
Osteoblasts Osteoclasts Binding/recruit/remodel/↓Hydroxyapatite deposit
Endothelium Osteoblasts Binding/remodeling/↓Hydroxyapatite deposit
Some epithelial Mono/macro Chemotaxis/IL-1, TNF, IL8/↓ NO
Brain Endothelial Survival/angiogenesis/↓ NO
Certain tumors Epithelium, kidney ↓ NO
Melanocytes Survival
OX40L DC CD4 T cells Costimulation/IL-2/IL-4/IL-5/longevity/memory
B cell DC Maturation/TNF/IL-12/IL-1β/IL-6 (by back signal)
Endothelium B cell Growth/differ (by back signal)
T cells Endothelial Inflammation (by back signal)
T cells Leukemia (w/HTLV)
RANKL T cells DC Activation/AP/survival
B cells T, B, DC etc. Lymphoid organogenesis
DC Preosteoclasts Maturation; bone development
Stroma, BM Mono Osteoclast differ (w/GM-CSF, TGFβ)
B cells B development; osteoclast development
SCF Stroma, BM Stem cells Growth
Fibroblasts Progenitor cells Growth
Liver Mast cells Growth/differentiation/survival/fibrosis
Spleen γδ T cells Growth
Certain tumors Tumors Growth (breast, lung, testicular, utererine,
cervical, ovarian, melanoma)
TNFα Mono/macro T cells Growth/development (wIL-1)/IL-2/IFNγ
T cells (active) B cells Activation/Ig production
NK Endothelial NO/cytotoxicity
Endothelial Macrophages NO/cytotox/IL-12/IL-10/apoptosis
Mast cells DC Growth/maturation (w/GM-CSF)
Neutrophils Neutrophils Degranulation/complement R/
Keratinocytes adhesion/ADCC/apoptasis
Adipocytes TIL Cytotoxicity
Pancreatic β LAK Cytotoxicity
Osteoblasts γδT cells Growth
Astrocytes Endothelial VCAM, damage, vascular leak syndrome
Neurons Osteoblasts Mitosis
Adrenal cells Osteoclasts Activation
(continued)
Walter M. Lewko and Robert K. Oldham 163

Table 1. (continued)
Cytokine Cell source Cells influenced Effectb
DC (Immature) Synovial Enzyme secretion
Epithelial cells Fibroblasts Growth
TRAIL CD4 T cells Tumor cells Apoptosis; apoptotic bodies for AP
NK cells DC AP/T cell activation
Monocytes Autoimmune T cells Apoptosis
DC (Immature) Mast cells Apoptosis
Tweak Many cell types Fibroblasts IL-8, IL-6, RANTES, IP-10
Synoviocytes Inflammatory cytokines
Bronch epithelium Inflammatory cytokines
Macrophages IL-6, MCP-1, IL-8, MMP-9
Mesangial cells MCP-1, RANTES, IP-10, VCAM
Astrocytes IL-8, IL-6, ICAM
Keratinocytes RANTES
Liver Proliferation
Endothelium Proliferation, ICAM, E-selectin, IL-8, angiogenesis
Some tumors Proliferation, survival
a
This is a partial listing that emphasizes cancer related activities.
b
Activity increased unless otherwise indicated.

suppressors of cytokine signaling (SOCS) which are also presence of microbial invaders and initiating immune
referred to as cytokine-inducible suppressor proteins response. They do this by specifically binding molecules
[332, 1882]. They act by binding Jak/STAT components, (danger signals) shed from microbes, collectively called
interfering with their binding to the receptor and their pathogen-associated molecular patterns (PAMPs) [1282,
function [333, 492, 1882]. They are tightly regulated and 1654]. Bacterial lipopolysaccharide (LPS) (endotoxin)
required for normal lymphoid development [1882]. is a well-studied PAMP. LPS is a major cell surface
Various cytokines induce SOCS to regulate their own component of Gram-negative bacteria. It binds TLR on
activities (a form of feedback inhibition) [1857] and the macrophages, endothelial cells and dendritic cells where
activities of other cytokines [428]. For example, SOCS it is a potent activator and stimulator of cytokine pro-
may be increased by immune suppressor cytokines such duction [1983, 2081]. Activation may be very vigorous.
as IL-10 [256]. LPS induces toxic shock syndrome [1629]. Knockout
Cytokines may act synergistically. For example, gene studies in mice have shown that TLR4 recognizes
IL-18 and IL-12 are synergistic in the way they induce LPS whereas TLR2 is essential for response to several
IFN-γ secretion. Synergism may be explained in a num- gram positive PAMPs [1586, 1956]. There are at least
ber of ways, but frequently, it is due to the upregulation six human Toll-like receptors. They are integral mem-
of one cytokine’s receptor or its signaling pathway by brane proteins. The intracellular region is homologous
the other cytokine [521, 965, 1724, 2214, 2253]. with the signaling domain of the IL-1 receptor family
[1303]. TLR signaling activates several cells that are
important in immune response [516, 2081]. Immature
dendritic cells contain these receptors. Binding LPS
Toll-Like Receptors induces DC activation, maturation and secretion of
and the Response to LPS cytokines such as IL-12, allowing antigen presentation
to occur. Toll receptors are an important bridge between
There is a family of receptors, molecularly related to the innate and specific immune response systems.
cytokine receptors, called the pattern recognition recep-
tors [520, 1089]. They are better known as the Toll-like
receptors (TLR) for their genetic similarity to an impor-
tant class of Drosophila morphogenesis genes. In addition
The Helper T Cell System
to their role in fruit fly embryogenesis, Toll also has an Mosmann and coworkers proposed a paradigm for the
antifungal immune function in the adult flies [1089]. differentiation of CD4+ helper T (Th) cells, cytokines
These receptors have the important role of sensing the secreted and their roles in regulating of the development
164 Cytokines

of cellular versus humoral immunity [1375, 1376, 1659, Th2 path selection. (a) Antigen dose; generally, low
1709, 1937]. This paradigm has been expanded to antigen dose favors the Th1 path, while higher antigen
include additional cells and newly discovered cytokines. dose favors Th2 cells [798]. (b) Route of antigen admin-
The following is a very brief summary of an involved istration [1993]. (c) Type of antigen presenting cell
system. [1775]. (d) Type of costimulation: during antigen
A model for the differentiation of Th1 and Th2 helper presentation, B7.2 favors Th2 [565].
T cells is shown in Fig. 1. Thp cells are naive precursors Th17 cells induce inflammation. This path responds
to helper cells; IL-2 is the major cytokine they produce. to extracellular microbes. Th17 cells have important
Antigen presentation stimulates their conversion to Th0, roles in autoimmunity. Th17 cells secrete IL-17A,
the intermediate precursor cells. Th0 cells secrete IL-2, IL-17F, IL-21, IL-22, IL-6 and TNF-α. TGF-β and IL-6
IL-4 and IFN-γ [1314, 1376]. Depending on environmen- induce differentiation of Th17 cells; IL-23 maintains
tal conditions, Th0 cells differentiate into Th1, Th2, Th17 and stimulates cytokine production. IL-2 and IL-13
and regulatory T cells. This process is commonly referred inhibit Th17 cells [55, 869].
to as polarization; the cells become polarized to secrete Regulatory T cells (Treg) are defined functionally by
specific cytokines and perform certain functions. their suppressive effects on immune response, inflam-
Th1 cells favor the development of cellular immunity. mation and autoimmunity. Tregs are the subject of
Intracellular microbes (such as viruses) are eliminated intense research; they have important roles in the cause
by this path. Cancer cells are also killed by cellular and prevention of diseases including cancer. Many types
immune response. Th1 responses are important in of cells show regulatory character. Here we will
delayed type hypersensitivity and in certain autoim- mention three major types. Th3 cells secrete the immu-
mune diseases. Th1 cells secrete several cytokines, nosuppressive cytokine TGF-β. Tr1 cells produce
among them INF-γ, IL-2 and TNF-β, which are collec- immunosuppressive IL-10. Natural Treg cells have the
tively referred to as Th1 cytokines. In particular, IFN-γ phenotype CD4+ CD25+ Foxp3+. IL-2 and TGF-β
is considered a key marker for Th1 response. IL-12 and induce nTreg cells. The nTreg mechanism of suppres-
IL-2 favor the Th1 path. IL-4, a product of Th2 cells, sion involves cell contact (cell surface receptors) and
generally inhibits the Th1 path. possibly the secretion of immunosuppressive IL-10 and
Th2 cells favor the development of humoral immu- TGF-β [1714, 2002].
nity. Th2 response also stimulates eosinophil recruit- Polarization is not limited to Th cells. Based on the
ment and macrophage function. This path eliminates cytokines secreted, NK cells [285], cytotoxic CD8
extracellular microbes and large parasites. Th2 responses T cells [251, 1708], macrophages [1313] and dendritic
are important in infectious diseases and in allergy. Th2 cells [1206, 1596, 1647] may develop type 1, type 2
cells secrete IL-4, IL-5, IL-6, IL-9 and IL-13. IL-4 is the and regulatory cell profiles. Imbalance in type cells
hallmark Th2 cytokine. Mouse Th2 cells secrete IL-10. (exaggerated polarization/cytokine secretion) appears
(In humans, both Th1 and Th2 cells produce IL-10). to drive development of diseases including autoimmu-
IL-4 stimulates the Th2 path whereas IFN-γ, a major nity (exaggerated Th1 or Th17) [6, 290], allergy
Th1 cytokine, inhibits it [2, 1746]. In addition to cytokines, (exaggerated Th2) [923, 1101], and cancer (Treg)
there are further influences that fine tune Th1 versus [1590, 2056].

Figure 1. Differentiation of Th1 and Th2 helper T cells.


IL-12 + IFNγ IL-2 TNF
Antigen presentation (AP) stimulates the conversion of
naive precursor Thp cells to immediate precursor Th0 cells. Th1
IL-4 - IL-13 IL-17
Th1 cells favor the development of cellular immunity and
response to intracellular microbes. Th2 cells foster humoral AP
immunity and response to extracellular microbes and para- Thp Th0
sites. The major positive and negative stimulators of each
path are indicated. Cytokines within the boxes are produced IFNγγ - IL-4 IL-5 IL-6
by the indicated cells. IL-10 is a Th2 cytokine for mouse Th2
IL-2 IL-2 IL-4 + IL-9 IL-10 IL-13
cells; in humans, both Th1 and Th2 cells produce IL-10
IL-4 OX40 +
IFNγ B7.2 +
IL-13
Walter M. Lewko and Robert K. Oldham 165

Antigen Presentation; bound proteins [685]. The active, receptor binding form
of most TNF family members is a trimer. These trimers
Dendritic Cells induce receptor clustering and signal transduction [75].
Membrane-bound TNF family members act by direct
T-cells and B-cells do not typically respond to antigens
contact between the cytokine bearing cells and receptor
directly, rather a family of cells referred to as profes-
bearing target cells. Certain membrane-bound cytok-
sional antigen presenting (AP) cells make the presence
ines, upon engaging receptor, may signal back (reverse
of an antigen known to these effectors in a complex and
signaling) inducing effects in the parental cell; where
highly regulated process referred to as antigen presenta-
reverse signaling occurs, both cells are, at the same
tion [1159, 1888]. Dendritic cells (DC), macrophages, B
time, targets and effectors.
cells, Langerhans’ cells, and eosinophils are antigen
TNF family members bind receptors that are related
presenting cells. These cells have the necessary ability
molecularly. They are transmembrane glycoproteins.
to migrate (between tissues and lymphnodes), they con-
Some TNF family receptors have soluble/shed forms
tain the appropriate antigen processing machinery (anti-
that are the result of proteolytic cleavage or alternative
gen is broken down to small peptides in the process) and
mRNA splicing. The amino acid identity between the
surface binding proteins (including MHC to display
human receptors is in the range of 25–35%. The extra-
antigen peptides) to help insure that an effective and
cellular regions characteristically have three to six copies
safe immune response occurs. AP cells also have the
of cysteine-rich pseudo repeats, each containing six
necessary costimulatory molecules such as CD80 (B7.1)
cysteines in a segment of about 40 amino acids. The
and CD86 (B7.2). Among the AP cells, dendritic cells
cytoplasmic region of certain receptors contains a con-
are particularly good at inducing specific T cells with
served “death domain,” involved in apoptosis (e.g.
anticancer and antiviral activity. Different subpopula-
TNFRI, Fas, DR3, DR4, DR5). There are intracellular
tions of AP cells may favor different types of immune
receptor associated proteins such as TRAF (TNFR-
response (e.g. Th1 vs. Th2) [1595, 1596]. Cytokines
associated factors), TRADD (TNFR1-associated death
control the proliferation and development of AP cells.
domain protein), and RIP (receptor interacting protein)
AP cells are also a source of cytokines which influence
which bind and communicate between the TNF receptor
innate as well acquired immunity. Abnormalities in anti-
family members to regulate apoptosis and other cytokine
gen presentation may result in autoimmune disease or,
responses [2137]. Members of the TNFR family share
in the opposite extreme, a failure to mount any immune
these intracellular signaling proteins to activate similar
response at all.
transduction pathways. Certain members of the TNF
receptor family are found in many types of cells (TNFRI,
TNFRII, Fas, Fn14) whereas others are restricted to
Tumor Necrosis Factor Family hematopoietic cells (CD27, CD30, CD40, HVEM, OX40,
of Cytokines 4-1BB) or specific tissues (nerve growth factor receptor)
[75, 2137].
Tumor necrosis factor (TNF) was originally discovered as
a component of blood, produced by host cells in response
to infection and bacterial products such as LPS [150, 151,
152, 250, 1471]. A number of related factors turned up [6, Interleukin-1
150, 657, 685]. Now it is known that TNF (now called Inflammation, Immune Regulation,
TNF-α) is the founding member of a family of at least 19
cytokines and related virus proteins. The members of the
Hematopoiesis, and Wound Repair
TNF family are involved in cell growth, differentiation, IL-1 was originally called lymphocyte activating factor
inflammation, wound repair, costimulation of immune for its effects on mitogen-treated thymus cells [617].
response and the regulation of cell death. It has also been called leukocyte pyrogen and endoge-
There is homology in amino acid sequence among nous pyrogen, for its fever inducing effects [436, 1401].
most TNF family members, though it is not particularly Monocytes, macrophages, keratinocytes, T cells, B cells,
high [685]. Nerve growth factor (NGF) is not homolo- NK cells, eosinophils, dendritic cells, fibroblasts, epithe-
gous but its receptor is related to the TNF receptor. lial cells, endothelial cells, neurons, glial cells and
TNF-β and NGF are soluble cytokines. TNF-α and astrocytes produce IL-1 [551, 1170, 1735, 1972, 2140].
TWEAK are mostly soluble, partly membrane bound. IL-1 is involved in inflammation, hematopoiesis, immune
The remaining TNF family members are membrane- regulation, wound healing and metabolic regulation.
166 Cytokines

It is responsible for, or at least involved in, a number of Then TNFR-associated factor-6 (TRAF-6) binds. Now
inflammatory diseases. IL-1 also appears to influence this multicomponent receptor complex is functional and
cancer growth and metastasis. it induces the activation of transcription factors, in par-
The IL-1 gene family has three main members: IL-1α, ticular NF-κB, which translocates to the nucleus, binds
IL-1β and IL-1 receptor antagonist (IL-1Ra). IL-1α and DNA response elements and induces the production of
IL-1β are synthesized by separate, distantly related mRNA’s responsible for IL-1 effects [243, 354, 1008].
genes [1225, 1500]. The proteins have 26% homology. The second receptor, IL-1RII binds IL-1 but cannot
Both cytokines are synthesized as 31,000 kDa pro-IL-1 signal due to the lack of a binding site for MyD88. There
molecules. They lack the usual signal peptides charac- is a soluble form of RII, which is found in tissue fluids;
teristic of most secreted proteins. it is increased during inflammation [628, 1593, 1938].
Pro-IL-1α and the processed 17,000 mw product are Soluble receptor II may be generated by proteolytic
both active cytokines. Newly synthesized IL-1α accu- cleavage [1506] or by alternative mRNA splicing
mulates within the cytosol. Not much IL-1α is secreted. [1136]. The cellular form of RII binds IL-1α, IL-1β and
Intracellular IL-1 may serve an autocrine function in IL-1Ra; soluble RII binds IL-1α and IL-1β but not
endothelial cells, keratinocytes and fibroblasts. Pro-IL-1α IL-1Ra [65]. RII appears to function as a competitive
is found on the cell surface, associated with lectin; decoy for IL-1 [334, 1446, 1621] and IL-1RAcP [1056]
bound IL-1α has cytokine activity [201]. available to bind IL-1RI.
Pro-IL-1β is inactive. It is processed to a 17,000 mw The biological properties of IL-1 are far reaching and
form, which is functional and the major secreted form of complex; it is often referred to as a two edge sword. On
IL-1. Processing is carried out by the protease, IL-1β one hand IL-1 functions in hematopoiesis, the regula-
converting enzyme (ICE, caspase 1) [68, 160, 272, 299, tion of metabolism, wound healing and the control of
1992]. ICE is involved in the processing of other cytok- infection. On the other hand, IL-1 appears to be involved
ines (IL-16 and IL-18), but not IL-1α, which is cleaved in the debilitating effects of several inflammatory dis-
by another protease. eases and septicemia [846, 1482].
mRNA synthesis and posttranslational processing IL-1 has a remarkable effect on hematopoiesis. In
control the production of IL-1β. Bacterial products early progenitor cells, IL-1 is survival factor; it protects
(LPS), TNF-α, IL-1 itself, and the immunostimulatory stem cells from environmental insults, cytotoxic chemi-
drug OK-432 stimulate IL-1β secretion by blood mono- cals, radiation and the like. IL-1 also primes progenitor
cytes and other cells [782, 971, 435, 437]. Extracellular cells for outgrowth, by placing them in the G0-G1 stage.
ATP triggers activation of ICE and a burst of IL-1β IL-1 by itself does not induce outgrowth [191, 530,
secretion [529, 528, 1557]. ATP is a signal that is 1442, 1501, 1762]. IL-1 increases IL-3, G-CSF, M-CSF
released by activated lymphocytes and from damaged and GM-CSF; together with these cytokines, IL-1
cells [782, 1925]. synergistically stimulates the expansion of specific cell
IL-1Ra is an inhibitor of IL-1. It competes with IL-1 lineages [100, 1329, 1349, 2306, 2309]. Patient treatment
for its receptor [454, 482, 658, 723]. Secretion of Il-1Ra with moderate doses of IL-1 caused elevated neutrophil
generally follows IL-1 production. In patients and in and platelet levels [1842]. Interestingly, prolonged treat-
cultures of human PBMC, IL-1Ra secretion is stimu- ment and higher doses caused an apparent decrease in
lated by IFN-α, IL-4 and to a lesser extent by IFN-γ neutrophils, platelets and erythrocytes. There are two
[1997]. Patients treated with IL-1 had increased serum possible explanations for this decrease: First, IL-1
IL-1Ra and with time the antagonist remained measur- induces TNF and TNF has a depressive effect on
able after IL-1 was no longer detected [1007]. hematopoiesis [594, 845, 898]. Second, IL-1 induces
There are two receptors for IL-1, IL-1RI that is adhesion molecules on capillary endothelial cells.
responsible for activity and IL-1RII that is a regulatory Studies in culture showed that endothelial cells with
decoy receptor. They are distinct membrane bound pro- elevated adhesion molecules bind a number of different
teins, produced by separate genes [310, 1829]. types of cells; these cells may be sequestered in the cap-
The active signaling complex contains several com- illary bed resulting in an apparent decrease in cell num-
ponents. IL-1 binds the IL-1RI. IL-1 receptor accessory bers [406, 1380, 1681].
protein (IL-1RAcP) binds and enhances affinity for IL-1 stimulates dendritic cells. It acts together with
IL-1; it is required for signal transduction [663, 1008, GM-CSF to promote maturation [744]. Dendritic cells
2144]. MyD88 is an adapter protein that joins the com- have an important function as antigen presenting cells in
plex [2145]. IL-1R associated kinase (IL-1RAK) then the development of lymphocytes. T cells and B cells
binds; the kinase is phosphorylated and activated [354]. synthesize IL-1 and have the IL-1 receptor [1163, 1504].
Walter M. Lewko and Robert K. Oldham 167

IL-1 stimulated T cells, induced IL-2 production, IL-2 and IFN-α. IL-1 increases IL-6 levels; blocking IL-1
receptors and cytokine production; IL-2R induction activity lowered IL-6 and depressed fever [327, 616,
appeared to be a key step [940, 1039, 1827]. Immature 2277]. IL-1 also induces anorexia in animals. The mech-
T cells appeared to require IL-1 in order to synthesize anism appears to involve increased brain cyclooxyge-
IL-2; IL-6 acts synergistically with IL-1 in T-cell matu- nase and prostaglandin production [733].
ration and the production of IL-2 [1686]. In more mature IL-1 has several effects on metabolism. It interacts
T cells, IL-1 favored Th2 cells. It stimulates the prolif- closely with a number of hormones, in particular the
eration of Th2 cells and the production of IL-4, IL-5 and glucocorticoids and insulin. IL-1 influences the brain-
IL-6. IL-1 had no apparent effect on Th1 cells. IL-1 also pituitary-adrenal axis. IL-1, injected i.v., increased the
acts as a chemoattractant for T cells and it stimulates release of corticotropin releasing hormone and ACTH
production of IL-8 [822, 1039, 1335]. In B cells, IL-1 as well as endorphins, vasopressin and somatostatin.
acts together with B cell growth and differentiation Glucocorticoid levels increased in response to injected
factors IL-4, IL-6 and IL-2 to stimulate growth and the IL-1 and during stress and trauma [1639, 1812].
production of immunoglobulin. IL-1 increases the levels Glucocorticoids influence metabolism throughout the
of glucocorticoids. Glucocorticoids stimulate B cell body; hydrocortisone feeds back on the production of
IL-1 receptors. Glucocorticoids and IL-1 together stim- IL-1 [144, 148, 410, 1080]. In this way glucocorticoids
ulate antibody production [14]. In natural killer cells, appear to regulate inflammatory response. IL-1 pro-
IL-1 and IL-12 together stimulated the secretion of motes bone resorption and cartilage degradation [424].
IFN-γ in response to infection [824]. It induces the release of collagenase, phospholipase A
The activities of IL-1, TNF and IL-6 are interrelated. and prostaglandin E2 from synovial cells [154, 1326,
IL-1 and TNF have similar biological properties and 2304]. IL-1 altered the production of several liver pro-
their combined effects are usually synergistic [1216, teins; fibrinogen, clotting factors, metallothionein and
2093]. IL-1 induces TNF production [1077, 2173] and complement were increased. Albumin, transferrin and
TNF induces IL-1 production [197, 435]. The receptors lipoprotein lipase were decreased. Negative nitrogen
for IL-1 and TNF share a common accessory protein balance in muscle protein is associated with inflamma-
and signaling path. As part of a regulatory loop, IL-1 tory diseases. IL-1, IL-6, TNF and insulin are involved.
tends to depress TNF receptors [788, 2093]. In mice Pancreatic β islet cells have IL-1 receptors [706]. Both
IL-1 induced IL-6 production. Blocking IL-1 by IL-1Ra IL-1 and IL-6 stimulate production of insulin [2182].
depressed IL-6 levels and inflammation [616, 1090]. IL-1 and IL-6 together with TNF have insulin-like
TNF stimulates both IL-1β and IL-6 [550]. IL-6 regu- effects on metabolism. In rats treated with endotoxin,
lates IL-1 activity by increasing IL-1Ra levels [1749] for example, IL-1 blockade using IL-1Ra spared muscle
and by decreasing IL-1 formation [1742]. protein [2272].
IL-1 stimulated mast cell production of IL-3, IL-4, IL-1 stimulates the growth of several different types
IL-5, IL-6, IL-9 and TNF. Thus, IL-1 may enhance Th2- of cells including keratinocytes, smooth muscle cells,
related events including humoral response and allergic glial cells, mesangial cells and fibroblasts [996, 1126].
inflammation. IL-1 may also influence the natural In this way, IL-1 has a role in wound repair and angio-
response of mast cells to parasites, viral infection [820] genesis. In fibroblasts, IL-1 increased proliferation
and cancer. [1744] and the uptake of glucose [156]. IL-1 also stimu-
The nervous system has receptors for IL-1 and brain lates collagen production by fibroblasts and epithelial
cells are capable of IL-1 production [196, 1072]. IL-1β cells. Tissue fibrosis is associated with inflammatory
is secreted by microglial cells in response to brain disease; it appears to be related to paracrine and auto-
trauma. IL-1 induces production of nerve growth factor crine IL-1 secretion. In cultured fibrotic kidney cells,
[1873] and ciliary neurotrophic factor [742], which are IL-1 blockade using IL-1Ra depressed fibroblast prolif-
involved in nerve cell survival and wound repair. In this eration [1150].
way IL-1 released during brain inflammation may have There are interesting relationships between IL-1 and
a beneficial effect on CNS regeneration [742]. IL-1 endotoxin. IL-1 is one of several cytokines (IL-2, IL-15,
treated patients often suffer from sleepiness, fever and IFN-γ and TNF-α), which stimulate Toll-like receptor
lack of interest in eating. IL-1 activity in the nervous (TLR) levels in macrophages [1247]. TLRs bind micro-
system appears to be involved. IL-1 injections in ani- bial products and stimulate innate immunity and inflam-
mals induced slow wave sleep and depressed rapid eye matory responses to pathogens. Endotoxin and IL-1
movement [1815]. IL-1 induces fever. There are several have similar biological properties including the induction
other cytokines capable of doing so including TNF, IL-6 of IL-1 and TNF levels. Interestingly, TLR and the
168 Cytokines

IL-1R have homologous intracellular signaling domains. possible that IL-1 blockade may be a way to intensify
Activation of these receptors brings about similar proin- IFN response in the treatment of viral disease [1994]
flammatory responses [164]. and possibly cancer. IL-1 is of interest in the treatment
IL-1 is involved in several inflammatory diseases. of cancer [1838]. In culture, IL-1 has a direct inhibitory
In the intestine, IL-1 and TNF are produced by epithe- effect on the growth of several types of human cancer
lial cells and appear to prevent microbial invasion. In cell lines [378, 959, 1498]. In animal studies, intratu-
inflammatory bowel disease, these cytokines were moral treatment with IL-1 induced regression of injected
increased. Somatostatin inhibited basal IL-1β produc- tumors but not distant metastases [1418]. Fibrosarcoma
tion and prevented induction by TNF and bacteria. cells expressing cell surface IL-1α, implanted in mice
These results suggested that somatostatin regulates showed decreased growth and induced protective immu-
IL-1 and may have a role in prevention of bowel inflam- nity [1858]. While preclinical studies showed promise,
mation [314]. In patients with rheumatoid arthritis, little or no benefit has been observed using IL-1 in
synovial tissues produce IL-1 [538] and the levels of patients with melanoma [1879] or renal cancer [1625].
IL-1 are elevated in plasma. IL-1 is a chemotactic fac- The combination of IL-1 with IL-2 was explored in
tor for neutrophils [1732]; IL-1 induces nitric oxide clinical trials. Increases in NK and LAK activity were
synthetase and the production of nitric oxide, which observed and there were some responses in patients
mediates many inflammatory processes [504]. IL-1 also with colon cancer, melanoma and renal cancer [2026].
induces cyclooxygenase II and the production of pros- IL-1 has also been tested in combination with chemo-
taglandins, which have inflammatory effects [1217, therapy to enhance drug effectiveness and decrease
1360]. IL-1 directly stimulated levels of the proteolytic blood cell suppression. IL-1 decreases the activity of
enzymes involved in joint destruction, collagenase, tis- drug metabolizing cytochrome P-450 [619]. In cultured
sue plasminogen activator, and stromelysin [424, 1403]. cells, IL-1 had synergistic effects with certain forms of
In chondrocytes, chronic IL-1 inhibited proliferation chemotherapy [1417, 2046]. Similar synergistic effects
and the production of collagen and proteoglycan [1156, of IL-1 and chemotherapy were seen in animal tumor
1885, 1968]. IL-1 and tumor necrosis factor act mutu- models [897, 1051]. Thus far the results in clinical trials
ally to stimulate joint inflammation [197, 2173]. have been rather disappointing. In ovarian cancer
Clinical and animal studies have shown that therapies patients treated with carboplatin and in osteosarcoma
blocking TNF-α activity were beneficial. In mice with patients treated with etoposide, the addition of IL-1
collagen-induced arthritis, anti-IL-1 antibodies and resulted in very modest increases in antitumor response
antibodies to the IL-1 receptor [607, 901, 2055, 2173] [2072, 2199]. In preclinical studies, IL-1 offered some
reduced the severity of the disease. Blocking both IL-1 protection to progenitor hematopoietic cells undergoing
and TNF-α had an additive effect [2173]. IL-1Ra and irradiation or treatment with cytotoxic drugs [191, 1441,
IL-1 RII are being studied clinically to see if they are of 1495, 1762]. When cancer patients were given IL-1 in
benefit in treating inflammatory diseases. In patients phase 1 clinical trials, granulocytes and platelets were
with rheumatoid arthritis, rhIL-1Ra reduced the pro- stimulated showing that IL-1 could be beneficial for
gression of joint erosion [199]. Unfortunately, rhIL- blood cell recovery [357, 1842]. In children on chemo-
1Ra did not significantly increase survival in patients therapy (Ifosfamide-Carboplatin-Etoposide), unfortu-
with severe sepsis [1499]. nately, treatment with IL-1α produced no significant
In patients with arthritis, exercise has a beneficial hematoprotective benefit [576].
effect on joints. A biochemical rationale was provided in IL-1 produces significant though manageable side
an interestingly cell culture model for physical therapy. effects. Low dose IL-1 causes headache, myalgia, arth-
Cyclic tensile strain on cultured chondrocytes decreased ralgia, sleepiness and anorexia. Higher doses cause a
the effects of IL-1β by interfering with IL-1 receptor rapid loss in blood pressure. These symptoms mimic septic
signaling. Markers for inflammation were decreased shock. Highest doses produced grade IV hypotension and
and the extracellular matrix was restored [2217]. neurological signs [1842]. In summary, IL-1 stimulates T
IFN is used to treat viral hepatitis. However, 60% of cells, B cells and hematopoiesis. Undoubtedly, it has a
patients do not respond well. IL-1 may be a reason for role in the development of immunity. But at present its
this lack of response. IL-1β appears to depress the anti- toxicity and modest antitumor effects preclude approval
viral activity of type I interferon [1994]. When mice for use in cancer patients.
were injected with IL-1β, liver cell antiviral response There is evidence that IL-1 stimulates the growth of
was depressed. IL-1 appeared to interfere with IFN certain types of cancer. In patients with chronic myelog-
signaling at the level of STAT 1 phosphorylation. It is enous leukemia, Il-1β levels were elevated and blocking
Walter M. Lewko and Robert K. Oldham 169

IL-1 depressed cancer cell growth; IL-1 was determined The IL-2 receptor is composed of three subunits:
to be a negative prognostic factor [2148]. IL-1 is an IL2Rα, IL2Rβ and IL2Rγ (also called γC, common γ
autocrine growth stimulator for certain human gastric subunit). The α subunit has high affinity for IL-2. The
cancers [864]. In capillary endothelial cells, IL-1 β and γ chains are involved in signaling [968, 1002,
increased cell surface adhesion molecules and the bind- 1097, 1823]. The three subunit complex exhibits high-
ing of tumor cells [406]. Pretreatment of mice with IL-1 est affinity for IL-2 (Kd 10−10 M). In NK cells, the
increased the metastasis of B16 [2074] and human mel- IL-2R is in the form of a βγ dimer; this two-subunit
anoma cells [620]. When the mice were treated with receptor has moderate affinity (Kd 10−9 M) and requires
IL-1Ra, there were fewer metastases, smaller metasta- higher IL-2 concentrations to produce effects [734,
ses and the animals lived longer [620]. Perhaps thera- 945, 1024, 1508, 1823]. Janus kinase activity is associ-
pies aimed at blocking IL-1 may be beneficial in ated with the γ chain. The receptor complex is activated
controlling the growth and spread of certain cancers. by phosphorylation. A major signal path involves the
STAT transcription factors (signal transducer and
activator of transcription), which bind to the activated
Interleukin-2 receptor complex and are in turn activated by phospho-
rylation. The activated STATs translocate to the nucleus,
Growth And Activation of T, B and NK bind an array of gene promoters and initiate transcrip-
Cells; Activation-Induced Cell Death; tion associated with IL-2 response. Immune suppressive
drugs FK406 and Cyclosporin A act at the level of
Elimination of Self-Reactive T Cells transcription factors, which are activated by IL-2 and
IL-2 was originally described as a factor in the condi- other cytokines [491, 544, 656, 1262, 1263].
tioned medium of mixed lymphocyte cultures that stim- A peptide has been produced which comprises
ulated T cell growth [643, 936]. The following is a brief amino acids 1–30 of human IL-2. It forms a tetra-
review of this very well studied cytokine. The early meric structure that binds IL-2R β dimers. The pep-
work is covered in greater depth in prior editions of this tide is an IL-2 agonist: it induces activation of CD8+
book [1110]. T cells and lymphokine-activated killer (LAK).
IL-2 is a member of the helical cytokine family, Interestingly, it acts synergistically with IL-4, IL-9,
which includes IL-2, IL-4, IL-7 and IL-15 [1963]. IL-2 IL-15 and with IL-2 itself. This peptide may have
is produced mainly by T cells. Activated cytotoxic T therapeutic potential [476, 477].
cells, Th0 and Th1 cells secrete IL-2 but Th2 cells do IL-2 was originally discovered by its capacity to
not [1040, 1798]. stimulate proliferation in T cells. T cells produce IL-2
Initial studies used natural IL-2 prepared from lym- and it stimulates T cell growth. A defect in IL-2 produc-
phoid cells [561, 1305, 1649, 2141]. It is a 133 amino tion appears responsible for a type of severe combined
acid glycoprotein with variable molecular weight due to immunodeficiency disease [2135]. IL-2−/− knockout
the carbohydrate. The gene for IL-2 has since been mice have been developed which lack IL-2. In initial
cloned [1666, 1963]. The recombinant protein made in studies, proliferation response was weak in Con A or
bacteria lacks carbohydrate. Certain forms of recombi- anti-CD3 Mab activated mononuclear leukocytes; other-
nant IL-2 have an amino acid change to facilitate pro- wise the mice appeared rather normal [1754]. Further
duction [210]. Recombinant IL-2 and the natural product studies showed IL-2 −/− mice had increased autoim-
have similar biological activities and stabilities [1663]. mune disease, ulcerative colitis, inflammatory bowel
Both forms of IL-2 have relatively short half-lives of disease, anemia, progressive loss of B cells and altered
1–2 h following intravenous injection [696, 1158, 1160] bone marrow cell profiles [1185, 1710, 1711]. The animals
and 4 h following bolus sc injection [696]. IL-2 loss is suffered from uncontrolled T cell proliferation, appar-
mainly by renal clearance rather than hepatic metabo- ently due to a defect in Fas-induced apoptosis [990,
lism or target cell binding [448]. Intraperitoneal injec- 1092]. Related observations have been made in mice
tion has been used to treat abdominal malignancies. lacking the IL-2 receptor α chain [2168] and β chain
Injection i.p. produces sustained, locally high levels of [1933]. In germfree IL-2 −/− mice, autoimmune disease
IL-2; systemic toxicity is lower. Sometimes, repeat i.p. developed but not colitis, which appeared to require
injections induced fibrosis, likely due to the release of enteric microbial antigen [341]. Interestingly, allograft
secondary cytokines such as PDGF or TNF-α [145, rejection still occurred, not only in IL-2 −/− knockout
1091, 2044]. IL-2 has also been delivered by inhalation mice [1886] but also in IL-2 −/− IL-4 −/− double knock-
[818] or and by mini-osmotic pumps [1458]. out mice [1119]. These results suggested IL-2 is not an
170 Cytokines

absolute requirement for rejection. Other cytokines such IL-2 is used to treat renal cell carcinoma and mela-
as IL-7, IL-12, IL-15 or IL-21 may provide redundancy noma patients [190, 1674, 1675, 2146]. Its effects gen-
in certain IL-2 functions [1886]. Nonetheless, IL-2 erally appear due to T cells and NK cells. Response
appears necessary for the normal regulation of T-cell rates were related to IL-2 dose. Most patients tolerate
growth and involution. In particular, IL-2 has a role in IL-2, but in addition to the usual flu-like discomforts
tolerance by inducing T cell suicide [1706, 1933, 2168], associated with cytokine therapy, high dose IL-2 induced
in activation-induced T cell death [1092, 1626, 2062] hypotension and vascular leak syndrome (VLS) which
and it is involved in the inhibition of T cell memory may be life threatening [106, 1676].
maintenance [1026]. Further, IL-2 induces regulatory T VLS is due to capillary endothelial cell damage.
cells (Treg: Foxp3+ CD4+ CD25+), which are antigen Possible sources of damage include cytotoxic lympho-
specific cells that regulate immune response. In particu- cytes, neutrophils [717], complement [1982], TNF [458,
lar, they suppress effector T cells, which are self-antigen 1607] and inflammatory factors such as histamine, sero-
specific and autoimmunogenic. In many cancer patients, tonin and bradykinin [1211]. Toxicity may be due to
these regulatory T cells are enriched in the tumors and IL-2-induced IL-1. In a phase I trial, cancer patients
blood. Since many tumor antigens are essentially over- were treated with soluble IL-1 receptor, an antagonist,
expressed self antigens, Treg cells may have a role in in an attempt to lessen IL-2 toxicity [1266].
tumor immune escape and poor response to cancer bio- Unfortunately, soluble IL-1R provided no benefit. The
therapy [165, 1309, 1806]. evidence for cytotoxic lymphocyte involvement is rather
IL-2 stimulates cytotoxicity in NK cells [734, 1386, strong. Immune suppressive agents including IL-10
1387], thymocytes [1800] and in cytotoxic T lympho- (discussed later) [1676, 1115] and inhibitors of immune
cytes (CTL) [1799]. Peripheral blood lymphocytes cell outgrowth [1606] depress VLS. IL-2-induced LAK
treated with IL-2 generate LAK cells [176, 444]. cells adhere to endothelial cells and kill them [375].
Cytotoxic NK cells make up the major active compo- Blocking the binding of leukocytes to capillary endothe-
nent of LAK [1507, 1567, 1995, 2133]. NK cells lyse lium [1481] and depletion of NK cells [1547] ameliorate
tumor cells by membrane channel forming perforin, VLS. Finally, IL-2-induced endothelial cell damage is
granzymes and receptor mediated cell death by apopto- significantly lower in mice, which are defective for
sis. IL-2 also enhances antibody dependent cellular perforin or Fas ligand (discussed later), both involved in
cytotoxicity (ADCC) mediated by lymphocytes against lymphocyte-mediated cytotoxicity [1612].
tumor cells [1810]. IL-2 has been used ex vivo to stimulate patient periph-
IL-2 stimulates B cell growth, differentiation and the eral blood lymphocytes to form LAK cells [1668, 1669].
production of Ig. IL-2 acts alone and together with Lymphocytes are collected by cytopheresis and cultured
other B cell stimulating cytokines such as IL-6 [886, with IL-2 for several days. IL-2-treated lymphocytes are
1419, 1872]. IL-2 also activates production of several then reinfused back into the patient. Oldham and co-
cytokines including the interferons, tumor necrosis fac- workers later developed a protocol, which only required
tor α (cachectin) and tumor necrosis factor β (lympho- a very short bedside incubation of lymphocytes with IL-2
toxin) [478, 611, 1016, 1599]. In this way IL-2 may prior to reinfusion [794]. LAK cells are preferentially
induces a cascade of effects influencing immune cytotoxic to neoplastic cells, though lysis of normal capil-
response. lary cells has been observed [76, 375, 1512, 1855]. LAK
Many tumor cell lines express the IL-2 receptor. cells act in a non-MHC-restricted manner [673, 1677].
Some of these cells respond directly to added IL-2 with In fact, the presence of strong MHC on the tumor cell
decreased growth. Interestingly, IL-2 sometimes down surface inhibits LAK. Initial reports indicated that LAK
regulates surface molecules such as ICAM and MHC, appeared effective in metastatic renal cancer and mela-
which are involved in antitumor immune response noma with response rates of 15–25% [474, 541, 1667].
[2234]. Denileukin diftitox (ONTAK) is a recombinant But subsequent reports for patients with renal cancer
cytotoxic chimeric protein composes of active domains questioned whether the cells provided any added benefit
of diphtheria toxin fused with the receptor-binding to the IL-2 given systemically at the time of LAK therapy
domain of IL-2. It binds to cells expressing the IL-2R, is [1669, 1674]. It appears that cancer patients treated with
internalized and kills susceptible cells. ONTAK has IL-2 develop LAK-like cells in vivo [1670].
been approved for use in cutaneous T cell lymphoma IL-2 has also been used ex vivo to stimulate out-
and is being tested in other hematologic malignancies. growth of tumor derived T cells. These expanded T cells
It is also being studied as a means of controlling regula- are called tumor infiltrating lymphocytes (TIL) [1018,
tory T cells in cancer patients [86, 380]. 1671, 2011]. They are also referred to as tumor derived
Walter M. Lewko and Robert K. Oldham 171

activated T cells (TDAC) [1108, 1109, 1207, 1208, The receptor for IL-3 has two subunits. The IL-3Rα
1493, 1494]. In the original work in mice, Rosenberg binds IL-3 [977]. It has homology with the GM-CSFRα
showed these cells were 100x more potent than LAK in [1010]. The β subunit is shared with receptors for IL-5
anticancer activity and TIL were effective against and GM-CSF [726]. The β subunit does not bind
certain LAK-resistant tumor cells [1672]. TIL are part cytokine but as part of each receptor complex it enhances
of the adaptive immune response system; they are cyto- affinity for the cytokine and it is responsible for signaling
toxic in an MHC restricted manner; antigen presenting (Jak2-STAT5) [1315]. IL-3, IL-5 and GM-CSF generate
cells were involved in their development. TIL are grown similar signals [697, 1323]. There is also evidence that
to greater than 1010 cells and then reinfused back into the IL-3Rα chain signals [1314] adding complexity and
the patient. Response rates in melanoma and renal specificity to the response.
cancer are on the order of 10–25% with some durable IL-3 stimulates the growth of most early multipo-
remissions. Combinations with other cytokines are still tential progenitor cells and early committed precursors.
being tested for increased treatment efficacy. IL-3 also stimulates macrophages, granulocytes and
Biochemotherapy, the combination of traditional che- mast cells through their most mature forms. IL-3 is a
motherapy with biological response modifiers, produces major developmental cytokine for basophils [1061,
an unexpectedly high response in melanoma patients. 1230, 2033, 1296]. Only the later stages of the eryth-
The reason for this is not understood. Response rate was roid [857] and megakaryocytic [2171] lines are no
correlated with increased serum IL-6 and IL-10, both longer sensitive. IL-3 thus increases the production of
Th2 cytokines, in patients receiving cisplatin, vinblastine macrophages, granulocytes, erythrocytes, and mega-
and dacarbazine followed by IL-2 and IFN-α-2b [674]. karyocytes. Effects on early erythroid and megakaryo-
However, in spite of response rates exceeding 50%, it cyte growth are rather distinctive to IL-3. IL-3 serves
is not clear that biochemotherapy adds any long term as a primer and costimulator while other cytokines act
survival benefit over IL-2 alone. later to induce differentiation. It stimulates both cell
IL-2 is an effective adjuvant, stimulating immune division and cell survival [407, 835, 2229, 924, 1919].
responses to tumor cells in mice. IL-2 is being tested as It acts synergistically with several cytokines including
a drug and as a product of genetically engineered cells, IL-6 [843], IL-11 [1405], G-CSF [842], thrombopoi-
in tumor cell and dendritic cell cancer vaccines [30, 587, etin [1027], stem cell factor [2031] and Flt3 ligand
588, 1106]. [1791]. In murine cell cultures, IL-3 tended to depress
In addition to cancer therapy, IL-2 has been used to the B lymphoid potential of lymphohemopoietic pro-
treat patients infected with human immunodeficiency genitors [767]. In human cells, IL-3 increased the
virus. CD4+ cell levels were increased with no increase production of B cell progenitors from uncommitted
in plasma virus [1017]. IL-2 has also been used ex vivo CD34+ CD38− cells [353].
to generate antiviral T cells, which were reinfused into Osteoclast development in hematopoietic tissue is
patients [200, 2095]. influenced by colony stimulating factors. IL-3 has been
reported to be stimulatory and regulatory. In RANK
ligand mobilized osteoclast precursors, IL-3 inhibited
osteoclast differentiation, diverting the precursors to
Interleukin-3 macrophages [956].
Natural cytotoxic (NC) cells [1524, 1912] are mast
Hematopoietic Cytokine cell-like morphologically and they participate in immune
IL-3 is a 28,000 mw protein. It is produced mainly by reactions such as tumor rejection and graft versus host
activated T cells [323, 324] but also by mast cells, thy- disease. They differ from standard NK cells in origin,
mic epithelium, keratinocytes, neurons, monocytes, growth and kinetics of target cell lysis. Natural cyto-
neutrophils and eosinophils [835, 857]. In the past, IL-3 toxic cells are IL-3 dependent. Unlike NK cells, they do
has been referred to as CFU stimulating activity, mast not respond to IL-2 [875–876]. Natural cytotoxic cells
cell growth factor, Thy1 inducing factor, multicolony appear to induce lysis by the release of TNF [876].
stimulating factor, P cell stimulating factor, and IL-3 has been studied extensively for possible clinical
hematopoietic growth factor [1327]. The gene for use [479]. It has been tested in combination with other
human IL-3 is on chromosome 5 in close linkage with cytokines such as GM-CSF, M-CSF, and EPO to stimu-
IL-4, IL-5, IL-9 and IL-13 [2227, 2248]. IL-3 is released late hematopoietic recovery of chemotherapy patients
during immune response and serves as a bridge between and bone marrow transplant recipients. IL-3 increased
the immune and hematopoietic systems. platelets, leukocytes and reticulocytes. Side effects due
172 Cytokines

to IL-3 (fever, headache, myalgia) were generally mild dylinositol-3′ kinase [944]. In mononuclear and B cells,
to moderate and manageable [589]. There were some IL-4 receptor levels are increased by IL-4; IFNγ inhibits
benefits but not sufficient to warrant becoming a part of this increase [1853].
standard therapy. PIXY321 is a GM-CSF:IL-3 fusion In addition to the membrane bound receptor, there is
product, which has been tested in cancer patients with a soluble form found in body fluids and in the medium
similar results and toxicities [899]. Unfortunately, most of cultured cells [526, 527]. The soluble receptor is
patients developed neutralizing antibodies suppressing smaller in size but it retains the capacity to bind IL-4
the effects of subsequent PIXY321 treatments [1311]. [1374]. The soluble receptor is not due to proteolysis of
At present, the main clinical use for IL-3 is in the pro- the membrane receptor, but rather differential mRNA
duction of cultured stem cells for patient reinfusion and splicing [122, 1374]. IL-4 stimulates soluble receptor
gene transfer protocols [214, 467, 475, 997]. production, apparently as a way of regulating IL-4
It has been reported that IL-3 may stimulate [90] or activity [307, 1638]. Soluble receptor competes with
inhibit [1789] certain hematopoietic malignancies. the membrane receptor for IL-4 [527, 1212]. The func-
DT(388)IL3 is a diphtheria toxin-IL-3 fusion protein, tion of the soluble receptor appears to be mainly inhibi-
which is being tested in patients with acute myeloge- tory [1212, 1730] but stimulatory [1730, 1871] effects
nous leukemia. It kills malignant progenitors while have also been observed. Further, the soluble receptor
sparing normal progenitors [781]. may also serve as a carrier for IL-4, which protects IL-4
There is interest in the use of particulate tumor anti- from metabolism and excretion, increasing its func-
gens for anticancer vaccination. Particulate antigens tional half-life [526, 457]. Dissociation of IL-4 from
tend to induce cytotoxic CD8+ cells while soluble the soluble receptor would allow it to bind and activate
antigens induce CD4+ helper T cells. IL-3 appeared to the membrane receptor.
be beneficial in this type of vaccination. IL-3 increased IL-4 has a major role in immune response. It has
the number of antigen presenting cells, the percent of direct effects on target cells, which are often synergis-
these cells actually presenting particulate antigen and the tistic with other cytokines. IL-4 also has a major regu-
number of CTLs produced [1594, 2239]. latory function in immunity by its influence on the Th1/
Th2 system [2, 254, 1821]. IL-4 induces precursor Th0
differention to Th2 cells [2]. In part, regulation in the
Th1/Th2 system involves apoptosis, programmed cell
Interleukin-4 death. Glucocorticoids favor apoptosis in thymocytes
and in mature T cells. Several cytokines including IL-4,
B Cell and T Helper Response IL-2 and IL-1 inhibit glucocorticoid-induced apopto-
Interleukin 4 (IL-4) is a 20 kDa glycoprotein. It was sis. IL-4 specifically rescues Th2 cells from death while
originally called B cell stimulatory factor-1 (BCSF-1) IL-2 rescues Th1 cells [1306, 2308].
for its effects on B cell growth and Ig secretion [514, As B cells depend on T cells for help, IL-4 is one of
801, 1544, 1608, 2249]. IL-4 is produced by activated the major cytokines for this purpose. IL-4 acts directly
CD4+ Th2 cells [132, 1036, 1040, 1127, 1375], mast on B cells and it is the main inducer of other Th2 cytok-
cells [189, 204], basophils [1190], eosinophils [1463], ines, most of which also stimulate B cells. IL-4 increases
NK cells [461, 1154], γ/δT cells [2307], CD4+ CD8+ T pre-B cell growth, differentiation and Ig secretion
cells [1086] and NKT cells [1087, 1832, 2252]. The [1015, 1597, 1850, 1851, 1871, 2082]. IL-4 stimulates
gene for IL-4 is located on human chromosome 5 in a class switching and the production IgG1 and IgE [321,
region with other cytokines including IL3, IL-5, IL-9 329, 409, 1552, 1851, 1871]. Among its other effects,
and IL-13. IL-4 increases MHC II on B cells for antigen presenta-
The IL-4 receptor is composed of two chains, IL-4Rα tion [1459].
and γc, the common γ chain found in several receptors In bone marrow IL-4 has a synergistic effect with
including the IL-2R [1372]. The α chain has the IL-4 IL-11 on the induction and growth of hematopoietic
binding site and conveys specificity as an IL-4 receptor. progenitor cells [1407, 1537, 1637]. IL-4 increases the
The γ chain is involved in signaling. There is another growth of monocytes [1974], macrophages [1447] and
configuration of the IL-4 receptor in which IL-13Rα increases macrophage antitumor activity and antigen
replaces the γ chain, in which case the same receptor presentation [239, 351, 2302].
complex binds and responds to both IL-4 and IL-13 IL-4 enhances the growth of activated PBL but had
[736, 1439]. Signaling involves phosphorylation of cer- little effect on unprimed resting cells. Timing of the
tain proteins including the receptor itself and phosphati- cytokine appears to be a factor. While IL-4 stimulated
Walter M. Lewko and Robert K. Oldham 173

PBLs previously activated with IL-2, IL-4 added to This may be related to the observations that long term
cultures simultaneously inhibited proliferation induced IL-4 benefits IL-12 secretion that in turn stimulates
by IL-2 [707, 2021]. cellular immunity [911].
IL-4 tends to stimulate the growth of thymoctyes IL-4 appears to have a role in several inflammatory
[2301] and T cells [2152]. But IL-4 effects are complex diseases. IL-4 together with IL-3 stimulated prolifera-
and in certain circumstances may be inhibitory [2153]. tion of mast cells [1374, 2032, 2033], polymorphonu-
In cultures of TIL, addition of IL-4 in combination with clear leukocytes [518] and fibroblasts [1342]. IL-4 also
IL-2, grew T cells from human tumors with specific stimulates ICAM-1. In allergic airway inflammation,
activity against autologous tumor [933, 934, 1108, 2034]. eosinophils accumulate and release IL-4 [1463]. IL-4
IL-2 was required; TIL could not be generated using increases production of IgE [321, 329, 1552, 1852,
IL-4 only [934]. IL-4 induced the IL-2 receptor in mouse 1872]. IL-4 also induces eotaxin, to further increase
T cells [254]. IL-4 together with IL-2 produced better eosinophil infiltration [2298]. In TNF-activated kerati-
TIL growth and specific antitumor activity in many but nocytes, IL-4 enhances secretion of chemokines, which
not all tumors [534, 933]. In certain TIL cultures, early in turn attract and activate effector cells [19]. While
growth effects of IL-4 were lost or even reversed with IL-4 may have a role promoting inflammation, it also
time [534, 933]. In another study, IL-4 in combination modulates inflammation directly, or by way of the Th
with IL-2 tended to produce more T cells with increased system [1541]. In monocytes IL-4 inhibits production of
antitumor cytotoxicity compared to TIL induced with IL-1, TNF-α and IL-6 while it stimulates IL-1Ra [525,
IL-2 only (8,724). We developed an IL-4 dependent T 1973]. Conversely, IL-1 inhibits the production of IL-4
cell line from a node specimen of a lymphoma patient. [1720]. Mouse autoimmune allergic encephalitis is a
This tumor cell line required IL-4 in addition to IL-2 Th1-related disease. In knockout mice lacking IL-4, the
[1108]. Other reports did not show remarkable benefits disease was exacerbated; treatment with IL-4 had a
of IL-4. In one study using melanoma, the addition of protective effect [507]. There is, therefore, clinical
IL-4 to IL-2 promoted TIL growth in a minority of cases interest in IL-4 for the management of allergy, inflam-
(5/24), but decreased growth in most cases (17/24). mation, autoimmune disease and also cancer.
Likewise, specific lysis was enhanced by IL-4 in a The receptor for IL-4 is expressed in several types of
minority of cases but depressed in the majority (13/19) cancer. In culture, IL-4 has inhibitory effects on certain
[1132]. It may be that IL-4 stimulated those T cells, hematological malignancies [16, 399, 452, 850, 1214,
which had been optimally activated by tumor antigen, 1630]. IL-4 also inhibited the growth of certain solid
but inhibited those cells, which had not been properly tumor cells including breast [2004], stomach [1358,
activated. 1359], colon [2004, 2013] melanoma [790], lung [2012,
Treating PBLs with IL-2 typically induces LAK cells. 364], sarcoma [1602] and renal [791, 1473]. Antitumor
In mice IL-4 could induce LAK in the absence of added effects in culture were generally IL-4 receptor-dependent
IL-2 [1388]. When mouse PBLs were induced with [17, 1359, 1473, 1602].
IL-2, IL-4 tended to enhance LAK activity [1388]. In IL-4 may be considered angiogenic in that normal
human PBLs, LAK may be induced by IL-4 in cancer fibroblasts and endothelial cells are stimulated by IL-4
patients pretreated with IL-2 but not in unprimed cells [574, 1342, 2005]. On the other hand, it has also been
[752, 874, 932, 933]. In previously untreated cells, IL-4 reported IL-4 is antiangiogenic [2083].
added at the same time as IL-2 inhibited induction of In animal studies, IL-4 inhibited certain human head
LAK by IL-2 [203, 707, 752, 932, 1870, 2152]. Priming and neck, glioma and colon cell lines growing as xeno-
with IL-2 in vivo or in culture was necessary for IL-4 grafts in nude mice. No remarkable immune cell infil-
activity. Loss of the IL-2 receptor may be the mecha- tration accompanied depressed growth [2012, 2013]. An
nism by which IL-4 inhibited LAK [1133]. acute lymphoblastic leukemia cell line from a child
Knockout mice have been developed which are defi- grew in nude mice. IL-7 stimulated growth while IL-4,
cient in IL-4. IL-4 has many important and interrelated TNF and IFN were inhibitory [664]. There are other
immune functions, but it is interesting that these mice had reports of inhibited acute lymphoblastic leukemia,
a relatively normal immune profile. Serum IgG1 was low. non-Hodgkin’s lymphoma and multiple myeloma cell
In response to nematode infection, the normal rise in IgE growth [16]. These animal studies showed IL-4 may be
levels failed to occur. Otherwise, T and B cell develop- able to inhibit tumor growth directly in addition to its
ment appeared relatively normal [1032]. More recent immune effects. But the growth inhibition is in most
studies showed that IL-4 deficient knockout mice had cases just that; decreased growth rate; growth proceeds
impaired antitumor cellular immunity (Schuler T. 1999). but at a slower pace.
174 Cytokines

Phase II trials have been carried out using IL-4 in Signaling is through the Jak-STAT pathway. IL-3, IL-5
cancer patients [85, 1157, 1171, 1226, 1971, 2021, and GM-CSF show similar patterns of signaling [1320,
2150]. Unfortunately, response rates were very low and 1323]. There is evidence that the IL-5Rα chain also sig-
toxicities were significant, particularly in the gastroin- nals adding to the uniqueness and complexity of the
testinal tract [1157]. IL-4 may cause cardiotoxicity IL-5R signaling system [1316].
[2020]. In vitro inhibitory effects of IL-4 on B cell There are membrane bound or soluble forms of the
chronic lymphocytic leukemia could not be confirmed receptor, the result of alternative mRNA splicing [1970].
in treated patients [1167] The soluble form appears to have a regulatory role in
Vaccines have been tested using mouse tumor cells eosinophilia in that it binds IL-5 and neutralizes its
genetically engineered to produce IL-4. In mice vacci- activity [205, 421].
nated with IL-4 secreting tumor cells [637, 1979, 2264] IL-5, IL-3 and GM-CSF downregulate the levels of
or IL-4 secreting fibroblast cells mixed with tumor cells IL-5Rα. This happens rather rapidly, within hours. This
[1572], animals rejected the modified tumor cells and regulation occurs at the level of mRNA transcription as
subsequent challenges of parental tumor cells [2264]. the gene is turned off [2105].
Cures of established tumors have also been reported IL-5 stimulates growth and differentiation of eosino-
[30, 636]. These approaches are now being tried in phils [1723, 2222]. It activates eosinophil function
patients [67, 1485]. IL-4 stimulates cell growth and [1151] and potentiates chemotactic responses in eosino-
antigen presentation in dendritic cells, B cells and mac- phils by IL-8 and RANTES [1769]. IL-5 also stimulates
rophages [239, 351, 1974, 2301, 2302]. Cancer patients, B cell growth and the production of IgA and IgM [1722,
treated with daily sc IL-4 and GM-CSF had increased 1935, 2246, 2247, 1953]; as such it is one of the main
numbers of circulating antigen presenting cells [1683]. factors from T cells, which provides help to B cells [96,
In current dendritic cell protocols for tumor vaccination, 969]. IL-5 enhances IL-2 receptors and responsiveness
IL-4 is often used in culture together with GM-CSF to in T cells and NK cells [935, 1953]. IL-5 augments
prepare and grow the dendritic cells for patient treat- IL-2-induced LAK activity in peripheral blood cells
ment [2039]. [58] and promotes, together with other factors, T cell
differentiation and mast cells growth [2246].
Eosinophils have important proinflammatory effects,
Interleukin-5 particularly in asthma [635, 783, 938]. For this reason
there is interest in IL-5 and its antagonists in asthma and
Eosinophil Growth/Differentiation; other inflammatory diseases [1723, 1278].
Inflammation; Augmentation of T, B, NK Though IL-5 influences several cells in immune
response, significant antitumor activity in preclinical
and Mast Cells models has not been demonstrated. No clinical trials
IL-5 is a 12–18 kDa protein, which was originally with IL-5 in cancer have been done.
known as B cell growth factor 2 (BCGF-2), T cell
replacing factor, eosinophil colony stimulating factor,
eosinophil differentiation factor and IgA enhancing
factor. The human gene is located on chromosome 5
Interleukin-6
where it is linked with several other cytokines includ- Hematopoiesis, Thrombopoiesis,
ing IL-3, IL-4, IL-9 and IL-13 [969]. The molecule is a
homodimer held together by disulfide bonds [96, 969].
Inflammation
IL-5 is produced by activated Th2 cells [1935, 1936], IL-6 is the founding member of the IL-6 family of
mast cells, eosinophils [202, 418, 459] and NK cells cytokines which includes leukemia inhibitory factor,
[2121]. In NK cells, IL-5 production is enhanced by oncostatin M, ciliary neurotrophic factor, cardiotro-
IL-4 while IL-10 and IL-12 are inhibitory [2120]. phin-1, novel neurotrophin/B cell stimulating factor-3
The receptor for IL-5 is composed of two subunits, an and IL-11. These cytokines share structure and their
α chain (IL-5Rα) and a β chain, which is identical to the receptors share a common gp130 subunit for signaling
β chain of IL-3R and GM-CSFR. The α chain binds [183, 601, 906, 2273].
IL-5. Its structure is quite distinct from that of the α IL-6 is a 22–27 kDa glycoprotein. It is also referred to
chains of the IL-3R and GM-CSFR [1010, 1398, 1929, as B cell stimulating factor-2, B cell differentiation
1970, 2040]. The β subunit enhances the affinity of the factor, cytotoxic T-cell differentiation factor, hepatocyte
α chain for IL-5 and it is responsible for signaling. stimulating factor, IL-1 inducible 26 kDa protein, and
Walter M. Lewko and Robert K. Oldham 175

interferon β2 [2052]. The gene for IL-6 is located on STAT 3 has different and even opposite functions
chromosome 7p21. Its primary structure has some depending on the cell type and physiological status.
homology with G-CSF [766]. IL-6 is produced by The gp130 subunit is shared with the receptor for
several types of activated cells including fibroblasts oncostatin M and other members of the IL-6R cytokine
[1257], macrophages, B cells [1868], CD4+ Th2 cells family [602, 974, 1138]; the shared subunit appears
[1043], CD8+ T cells [1708], epithelial cells [1023], responsible for the common effects brought about by
endothelial cells [2213], eosinophils [1287], astrocytes these cytokines. Essentially all the cells of the body
[1079], neurons [1234, 1646], synovial cells [1324, produce gp130, whereas IL-6Rα is expressed by IL-6
1940], megakaryocytes [1324], osteoblasts [859], mast target cells including B cells [766] hepatocytes [610],
cells [580], keratinocytes [365], Langerhans’ cells monocytes, CD4 and CD8 T cells [2183], CD34+ stem
[365], neutrophils [496, 1287], colon epithelial cells cells [1946, 2183], neurons [1752], neutrophils [1331]
[907] and certain tumors [743, 1164, 1749]. and osteoblasts [2041]. There is a soluble form of
As with many other cytokines, the regulation of IL-6 IL-6Rα. It is interesting in that unlike most soluble
is complex and disregulation may be involved in disease. cytokine receptors, it is active. The soluble IL-6Rα is
IL-6 levels increase with bacterial and viral infection, generated in two ways, by proteolytic cleavage of cell
inflammation and trauma [2053, 2102]. Wound fluids surface receptor and by alternate splicing of mRNA
contain IL-6 and wound fibroblasts secrete IL-6 in [1391, 1665]. TACE, the protease involved in TNF-α
culture [1238]. IL-6 levels may increase remarkably production, appears to be responsible for proteolytic
after surgery, up to several hundred-fold. Thrombin, shedding of IL-6R [716]. The soluble form binds IL-6
which induces clotting and wound healing, stimulates and mediates IL-6 responses in target cells that express
the production of IL-6 by fibroblasts, epithelium and the gp130 but lack membrane bound IL-6Rα [1197,
other types of cells [1865]. Endotoxin and TNF-induce 1562]. Certain hematopoietic cells [1563], neurons [1234]
the production of IL-6 and other inflammatory cytok- and smooth muscle cells [987] were responsive to IL-6
ines [550]. IL-1 increases expression of IL-6 and IL-8 in only in the presence of the soluble receptor subunit.
skin fibroblasts and arthritic synovial cells [612]; TGF-β IL-6 has a major role in the regulation of hematopoi-
induced the production of IL-6 in several cells including esis, inflammation and immunity. IL-6 −/− knockout
monocytes, keratinocytes and bone marrow stroma. In mice were defective in immune and acute phase protein
plamacytoma cells TNF and IL-1 were both stimulators responses [991]. IL-6 stimulates proliferation of multi-
of IL-6 production [2052]. IL-4 and IFN-γ acted syner- lineage hematopoietic stem cells. Mice that over
gistically to enhance IL-6 production in endothelial expressed both IL-6 and the soluble IL-6 receptor had
cells [803]. In lung fibroblasts, TGF-β induced IL-6 grossly enlarged livers and spleens, and overproduced
synthesis by a transient burst of H2O2 followed by an blood cells [1562].
increase in cellular calcium, which activated the signal IL-6 arrests the growth of cultured M1 cells while
pathway for IL-6 expression [909]. And CD40 engagement inducing differentiation into macrophages. IL-6 stimu-
stimulated IL-6 in B cells and several non-hematopoietic lates B cell differentiation and immunoglobulin pro-
cells; activation of NF-κB was involved in the process duction [766, 1394, 1872, 2064]. IL-6 is a costimulator
[487, 743]. of growth and cytokine production in thymocytes and T
Heparin and heparin sulfate selectively bind IL-6 cells [1155, 2016, 2064]. IL-6 stimulates T cell activation,
and several other cytokines. Bound cytokine, upon dis- differentiation and antitumor activity [1385]. Fibroblasts
sociation, is available to its receptor. Heparin protects and epithelial cells secrete IL-6 in response to blood
the cytokine from proteolysis and may serve to keep clotting [1865]; along with other factors, IL-6 enhances
the cytokine in high local concentration, close to where megakaryocyte colony formation and the production of
it was secreted, available for paracrine effect [1392]. platelets [308, 755]. Natural killer cells are activated by
The receptor for IL-6 is composed of two subunits, IL-6 in peripheral blood lymphocytes [584, 838, 1847].
the IL-6Rα chain and gp130. IL-6 binds to the IL-6Rα IL-6 has direct effects on several types of non-
subunit [2225], which in turn binds, dimerizes and acti- immune cells. IL-6 is a hepatocyte stimulating factor
vates gp130 subunits [1566, 1943]. This leads to the [999]; it induced the production of acute phase proteins
activation of tyrosine kinases, in particular Jak1. These including fibrinogen [258] and the receptor for anaphy-
kinases phosphorylate gp130; gp130 then binds and latoxin C5a [1740]. IL-6 also induced acute phase
activates STAT transcription factors, in particular proteins in intestinal cells [1340]. IL-6 and the other
STAT3. Phosphorylated transcription factors translocate inflammatory cytokines TNF-α and IL-1 induce ACTH
into the nucleus where target genes are activated [1566]. and cortical release [1237]. IL-6 has a part in angiogenesis;
176 Cytokines

it stimulates endothelial cell growth and blood vessel Human herpes virus 8, which is associated with
formation [1378]. IL-6 also increases fibroblast out- Kaposi’s sarcoma, produces a viral IL-6 molecule
growth and collagen production for wound repair [468]. that has 25% homology with human IL-6 [1437]. It
Physiological levels of IL-6 appear to protect neurons has certain effects that are similar to those of cellular
and support nervous system repair [682, 1037]. Mice IL-6 [226]. The viral IL-6 appears to be responsible
that over expressed both IL-6 and its receptor showed for some of the virus’ pathology, including increased
accelerated nerve regeneration after damage [769]. In angiogenesis and hematopoiesis [60]. Interestingly,
bone, IL-6 stimulated osteoclast production and bone the viral IL-6 binds directly to gp130 and exerts its
resorption activity [1220]. IL-6 may be involved in effects on cells independently of the IL-6 receptor,
epithelium-stroma signaling during normal development, for which viral IL-6 appears to have little or no
for example, in breast tissue [831]. affinity [1390].
IL-6 appears to have a role in inflammatory condi- In preclinical studies, IL-6 showed promise as an
tions. The blood of patients with gram-negative septic anticancer agent. In vitro, IL-6 inhibited the growth of
shock contained elevated IL-6, TNF, IL-1 and LIF and certain breast cancer cells, leukemia, lymphoma and
the levels of each were correlated with disease severity other cell lines [296, 1357]. IL-6 increased ICAM-1
[2077, 2118, 2119]. IL-6 and LIF were induced by (CD54) in certain breast cancer and melanoma cell lines
TNF-α [880]. IL-6 and its soluble receptor were ele- [831, 973]. IL-6 inhibited growth in several mouse
vated in inflamed intestinal tissue [676, 1317]. IL-6 was tumor models; these mice developed tumor-specific
one of the proinflammatory cytokines required for the CTLs, but not LAK cells. In mice treated with IL-6,
induction of mouse colitis as a model for human Crohn’s growth and metastasis of Lewis lung [927] and mela-
disease [2223], for experimental antigen-induced rheu- noma [926, 1164, 1165] were inhibited. Resistance may
matoid arthritis [1510] and autoimmune encephalomy- develop in IL-6 sensitive cells. Loss of sensitivity was
elitis [1718]. IL-6 is among the several proinflammatory associated with loss of IL-6 receptors in some but not all
cytokines expressed by synovial cells of arthritic patients cases [1825]. IL-6 is involved in the inflammatory
[1324, 1963]. IL-6 is elevated blood and cerebral spinal response that produces tumor infiltrating lymphocytes
fluid of patients with multiple sclerosis, Parkinson’s and [1385]. In cultures of TIL from human renal cell carci-
Alzheimer’s disease [168, 682, 1434]. Overexpression noma, addition of IL-6 to the standard IL-2-containing
of IL-6 in mouse brain induced acute phase proteins and medium stimulated TIL growth though not tumor cell
neurodegeneration [235]. In scleroderma patients, IL-6 kill; IL-6 alone did not induce TIL proliferation [1082].
was one of the cytokines required for the continued pro- In vaccination studies, IL-6 engineered mouse mam-
duction of autoimmune antibodies [1043]. IL-6 may mary tumor cells offered some protection to subsequent
play a part in cardiac disease. Fibrinogen is considered challenge by unmodified tumor cells.
a cardiovascular risk factor [497]. Fibrinogen levels are IL-6 and other IL-6 family members (IL-11, LIF,
elevated during inflammation; IL-6 is one of the main CNTF, NNT) induce appetite loss and wasting. In mice,
inducers of fibrinogen expression [1620]. colon adenocarcinoma cells produced elevated serum
IL-6 is generally referred to as a proinflammatory IL-6 and cachexia. The drug Suramin interfered with the
cytokine, but it appears to be involved in the resolution binding of IL-6 to its receptor and reversed cachexia in
of inflammation as well. IL-6 attenuates the synthesis of tumor bearing mice [1900, 1901].
proinflammatory cytokines while it has little effect on IL-6 is an autocrine growth factor in certain human
production of cytokines such as IL-10 and TGF-β. In cancer cells including multiple myeloma [115, 724,
IL-6 deficient mice, inflammation, eosinopilia, the 1478], B-cell leukemia [754], cervical carcinoma [503],
secretion of chemokines and Th2 cytokines were renal cell carcinoma and prostate cancer [1005, 1307,
increased. In mice that produced excess IL-6, inflamma- 1955]. In myeloma, growth stimulation appears to be
tion and Th2 cytokine secretion were decreased [2102]. linked with several of the IL-6 family cytokines (OSM,
IL-6 has also been shown to induce the production of LIF, CNTF, IL-6) [689, 690, 2147]. In human melanoma,
IL-1 receptor antagonist and the soluble TNF receptor, there is evidence that with tumor progression, there is a
both of which inhibit inflammation [1998]. In astro- transition from IL-6 paracrine growth inhibition to auto-
cytes, IL-6 inhibited expression of TNF-α and down crine growth stimulation. Serum IL-6 in melanoma
regulated levels of cell adhesion molecules [137, 1480, patients was found to be a prognostic factor for poor
1816]. One mechanism for the anti-inflammatory effects survival and lack of response to IL-2 therapy [1966].
of IL-6 (and the related cytokine IL-11) may involve IL-6 was tested in cancer patients, including renal
inhibition of transcription factor NF-κB [2023]. cancer, in phase I and II trials [2057, 2126, 2136].
Walter M. Lewko and Robert K. Oldham 177

In addition to its potential anticancer activity, there was There is a soluble form of the receptor that is produced
interest in IL-6 for the stimulation of platelet and blood by a specific mRNA [641].
cell levels. Patients treated with IL-6 demonstrated the Three different IL-7 binding affinities have been
usual cytokine flu-like side effects plus reversible anemia, observed [74, 558, 1523, 2017]. The highest affinity
leukosis, thrombocytosis, increased acute phase proteins, receptor is generally found in activated cells and appears
heart problems, elevated bilirubin and confusion indica- to be the one responsible for cell division [73, 1523]. In
tive of neurotoxicity. There was a suggestion of benefit human T cells, response to IL-7 depends on the activa-
in certain studies but toxicity was significant. One report, tion state. In resting cells, IL-7 induces certain cellular
a phase II study in advanced renal cell cancer, showed proteins (CD25, IL2 R) but no proliferation. In activated
lack of efficacy [1758]. Anti-IL-6 monoclonal antibodies cells, IL-7 is a potent inducer of cell division [648, 1365].
have been used to treat patients with myeloma [114, 2065] Immunosuppressive drugs cyclosporin and FK506
and AIDS associated Kaposi’s sarcoma [1610] without inhibit IL-7 stimulated growth, apparently by interfering
any apparent benefit. In a phase I trial involving pediat- with the function of the high affinity receptor [557].
ric patients given IL-6 plus G-CSF after myelosuppres- IL-7 has important influences on several different
sive chemotherapy, there were increased hematological immune cell types. Genetically deficient mice, lacking
responses but with a high incidence of toxicity [188]. IL-7, were extremely lymphopenic [2084]; early lym-
IL-6, IL-10 and TGF-β are cytokines reported to be phocyte development was severely impaired [1548]. In
secreted by tumors, which interfere with dendritic cell mice there is an absolute requirement for IL-7 in the
differentiation and antigen presentation, resulting in development of B and T cells. Interestingly, it is unusual
ineffective antitumor immune response [129]. for the loss of any one cytokine to cause such impair-
ment. This is probably due to redundancy of cytokine
functions, which is not the case for IL-7 in mice [2084].
In humans, IL-7 is not so strictly required for the devel-
Interleukin-7 opment of B cells [1592].
T Cell, B Cell, Macrophage and Dendritic IL-7 induces the growth of early B lineage cells. In B
lymphocytes, IL-7R was found on pre-B cells but not on
Cell Development
mature B cells, consistent with its role in the early stages
IL-7 was first described as a factor produced by mouse of B cell maturation [371, 372, 1918]. Stem cell factor
bone marrow, which stimulated B-lymphocyte progeni- and Flt3 ligand are costimulators [1229, 1424]. IFN-γ
tor cells to multiply and develop [1422, 1423]. IL-7 is a blocks the stimulatory effect of IL-7 on pre-B cells
25 kDa glycoprotein. It is produced by bone marrow [592]. Mice deficient in IL-7 fail to support normal B
stroma [641, 1422, 1423], thymic stroma [1716], kerati- cell development [2084].
nocytes [69, 745, 1245], human intestinal epithelium For prolonged B cell outgrowth, additional IL-7
[2115], B cells [134], follicular dendritic cells and related signals appear to be required, which occur by
vascular cells [1024]. IL-7 is also produced by tumors way of physical contact with stromal cells. IL-7 supports
including certain carcinomas [910] leukemias [569, outgrowth of pre-B and pro-B cells but the pre-pro-B
1578] and lymphomas [973, 1001]. IL-7 is not detected cell population requires, pre-pro-B cell growth-stimulat-
in normal T cells [2263]. Upon secretion, IL-7 binds to ing factor. PPBSF is composed of at least two covalently
integrin in the extracellular matrix; this may be the form associated subunits, one of which is an IL-7 chain. The
utilized by thymocytes [980]. other subunit is a protein that is attached to the stromal
The IL-7 receptor is composed of two subunits, cell surface. Pre-pro-B cells bind PPBSF during stromal
IL-7Rα and γc. The IL-7Rα chain contains the IL-7 cell contact; PPBSF stimulates division and primes the
binding site [640]. The γc is shared between IL-2R, cells for IL-7 [1273]. Thymic stromal lymphopoietin is
IL-4R, IL-7R, IL-9R and IL-15 R [1002]. γc enhances another IL-7-related factor in early B cell development.
receptor affinity of IL-7 and it participates in signaling It was discovered in the conditioned medium of a thy-
[1002, 1461]. The activation response involves Jak1, mus cell line [568]. TSLP and IL-7 have overlapping
pI-3 kinase, STAT3 and STAT5 [556]. Receptors that actions [568]. TSLP binds the IL-7Rα with another
share γc have similar but not identical nuclear effects. receptor subunit; it does not use γc. TSLP induces the
Lack of active γ chain in patients with severe combined transcription factor STAT5 as does IL-7, but TSLP did
immunodeficiency disease results in loss of function in not activate any known Janus kinases.
these five receptors. Loss of IL-7R function is a major Normal T cell development depends on IL-7. Mice
reason for poor immune response in these individuals. lacking IL-7Rα [1205, 1223, 1548] and mice treated
178 Cytokines

with neutralizing antibodies to IL-7 [650] do not pro- be administered to patients to expand CD4+ and CD8+
duce normal T cells. γδ T cells are missing entirely in lymphocytes. This occurs with a decrease in the per-
IL-7Rα (−/−) mice [728, 1548]. αβ T cells are present, centage of regulatory CD4+ T cells [1673].
but they are not responsive [1548]. IL-7 is a growth/ Several preclinical studies suggested IL-7 may be
differentiation factor for fetal pre-T cells, thymocytes useful in the treatment of cancer. Lung metastases of
[143, 735, 1492, 2080, 2153] and mature CD4+ and Renca renal cancer cells were reduced in mice treated
CD8+ T lymphocytes [73, 143, 293, 347, 564, 648, with rhuIL-7; the mice showed increases in T cells,
1148, 1365, 2243]. An important effect of IL-7 in both CD8/CD4 ratio, B cells, macrophages and NK cells
T and B cells appears to be its stimulation of the bcl-2 [1001]. Nude mice with human colon cancer xenografts
antiapoptotic pathway, for the maintenance of lympho- lived longer when treated with rhuIL-7 plus human T
cyte viability [15, 1222, 2085]. IL-7 was the only cells compared to mice treated with either alone. The
cytokine among 16 factors tested capable of inducing antitumor activity appeared due to interferon induced
rearrangement (diversity) in the T cell receptor V(D)J in CD8+ cells, not cytolysis. Interestingly, injected
region. IL-7 activated expression of the RAG1 and interferon was not that effective suggesting local con-
RAG2 genes involved in gene rearrangement [306, tinuous release was better than systemic interferon
1383]. In activated human peripheral blood T cells, IL-7 therapy [1402].
induced the secretion of several cytokines including IL-7 appears to be a good adjuvant during tumor vac-
IL-2, IL-3, IL-4, IL-6, IFN-γ and GM-CSF [442, 443]. cination. Tumorigenesis was decreased in mice inocu-
IL-7 also increased antitumor cytotoxicity in T cells [22, lated with IL-7 gene engineered plasmacytoma cells
143, 749, 776, 1180] and this cytotoxicity may persist [775], glioma cells [59] and fibrosarcoma cells [1261].
long term in culture without frequent antigenic restimu- Animals that rejected engineered tumor cells developed
lation [1182]. immunity to subsequent injections of the parental cells
IL-7 acts together with several cytokines in the devel- [61, 1261]. The tumors were infiltrated with T cells.
opment of T cells [2139]. Flt3 ligand and stem cell fac- Complement receptor rich macrophages, eosinophils
tor [1350, 1364] are co-stimulators with IL-7 for early T and basophils were also increased [776, 1261]. IL-7 was
cells. IL-1 and GM-CSF induced outgrowth of mouse one of several cytokines which upregulated intercellular
thymocytes; IL-7 was required [735]. IL-2 acts together adhesion molecule-1 (ICAM-1) on human melanoma
with IL-7 to stimulate antigen-induced effector cells cells. This protein is involved in immune recognition
from memory CD8+ T cells. IL-7 may act alone or with and anticancer action [973].
IL-2 [125, 1283]. Together the effect is synergistic Tumor infiltrating lymphocytes have been used to
[1283]. IL-7 stimulates IL-2 receptor (CD25) levels in treat cancer patients. Interleukin-2 is used to activate
resting T cells [73]; this may explain the synergy. and grow TIL in culture. IL-2 generally stimulates good
Among its many other functions, IL-7 induced prolif- initial growth and tumor cell kill, but antitumor activity
eration and antitumor activity in human blood mono- is often difficult to maintain over the long culture time
cytes and macrophages [24, 871]. In treated mice, IL-7 needed to grow sufficient cells for therapy, especially
mobilized myeloid progenitor cells from bone marrow when tumor cells are not available as a source of antigen
to peripheral sites [686]. It supported eosinophil pro- to restimulate the T cells. There are reports of difficulty
genitors in human bone marrow cell culture [2071]. maintaining CD4+ helper cells in IL-2-induced cultures.
IL-7 induced lymphokine activated killer cells [1181, IL-7 has been examined to determine whether it pro-
1736, 1896]. While IL-2-LAK exhibited better mela- vides any benefit in culture. IL-7 alone stimulated the
noma cell kill, IL-7 LAK killed with a different pattern growth of TIL cultures from certain renal cancer and
of cytokine release [1431, 1432], without TNF secretion enhanced IL-2-induced growth in others [1182]. Growth
and without toxicity towards normal cells [1731]. and antitumor activity persisted during long term culture.
IL-7 appears to have a part in antigen presentation. IL-7 Antigen restimulation was not required to maintain
was a growth factor for mouse dendritic epidermal T cells antitumor activity. Others have reported IL-7 was not
[1245, 1246]. Dendritic cells produce IL-7 [1022]. beneficial at the initiation of TIL culture but rather IL-7
Keratinocytes that surround the dendritic cells in skin pro- stimulated growth and cytokine secretion in cultures
duce IL-7 and TNF-α [1245, 1246]. Together these cytok- that were already responding to IL-2 [1820].
ines stimulated dendritic cell growth. IL-7 also increased There are reports that IL-7 stimulates certain types
levels of the co-stimulatory protein B7 on B cells [414]. of cancer. IL-7 increased the growth of several leu-
Lymphopenia can occur in patients with HIV infec- kemia cell lines [664, 1242, 1351, 1491, 1531, 2017].
tion and chemotherapy. It has been shown that IL-7 may Sezary lymphoma cell lines responded to IL-7 and
Walter M. Lewko and Robert K. Oldham 179

IL-2 with increased and in some cases synergistic endothelial cells and keratinocytes [950]. Like other
growth [374, 554]. The effect of IL-7 may be auto- chemokine receptors, the IL-8 receptors are members of
crine or paracrine [554, 664]. Abnormal cytokine the rhodopsin superfamily; these proteins characteristi-
secretion by tumor cells may be responsible for cally contain seven transmembrane domains. Chemokine
immune system abnormalities common in CLL receptors are linked to phospholipase C through G
patients [569]. Transgenic mice have been developed proteins. Receptor activation results in increased diacyl-
with increased IL-7 expression in lymphoid tissues. glycerol, inositol triphosphate and increased intracellular
These animals had remarkable skin T cell infiltrates, calcium released from cellular stores.
the result of the growth stimulating effects of IL-7. IL-8 was first described as a macrophage-derived
These transgenic mice also produced T and B cell chemoattractant for neutrophils [1384, 1755, 2096]. IL-8
lymphomas. This model system shows that the IL-7 is also a chemoattractant for T cells [98]. It has little effect
locus could behave as an oncogene. on B cells. In dogs injected with human IL-8, the site
There have been preliminary studies using IL-7- became infiltrated, mainly with neutrophils [1987]. IL-8
transfected autologous tumor cells as vaccines. Phase I stimulates neutrophil production of superoxide anion,
trials in patients with melanoma showed that IL-7 gene- degranulation with the release of hydrolytic enzymes
transfected, irradiated cells administered subcutane- such as elastase, and migration through endothelium of
ously were safe and produced tumor-specific CTL capillaries [101, 252, 815, 1565]. Mast cell-dependent
responses. One minor clinical response was reported recruitment of neutrophils appears to be mediated by IL-8
[1338, 1339]. Another study, a phase I/II trial in ten [1130]. IL-8 also induces migration in IL-2 activated
patients with metastatic carcinoma, used autologous peripheral blood NK cells [1774]. In rabbits, IL-8 block-
tumor cells transfected with IL-7, GM-CSF and double ing antibodies had an inhibitory effect on inflammation,
stem-loop immunomodulating oligodeoxyribonucle- especially in the lung [1780]. IL-8 also stimulates angio-
otides (d-SLIM). One CR, one PR and one MR were genesis [1346]. IL-8 is an attractant to antigen presenting
observed [2179]. cells such as macrophages and eosinophils [408, 1769,
1777]. As such, IL-8 may have an important role in the
initiation of immune response as well as the recruitment
of effector cells that carry out the response.
Interleukin-8 Fibrin formation is associated with trauma, inflam-
mation, wound healing and cancer. The addition of
Chemotaxis, Angiogenesis fibrin to cultured endothelial cells induced the release of
IL-8 is a potent chemotactic and proinflammatory IL-8 [1603]. The physical act of blood clotting may
cytokine [1250, 1384]. It is a member of the CXC stimulate cytokine release and cell migration. Besides
(ELR+) chemokine family (see below). IL-8 is a rela- IL-8 there are several additional proteins and peptides
tively small, 6,000–8,000 mw glycoprotein [1755, 2096, that are chemotactic to immune cells; macrophage
2257]. It is secreted by a variety of cell types including inflammatory protein α (MIP), IL-1α, and RANTES
macrophages [1755], endothelial cells [623, 1455, 1603, (see below) to mention a few.
1756, 1905], neutrophils [255, 1949, 2130], epithelial IL-8 appears to have a role in inflammatory diseases.
cells [123, 913, 961, 1878], fibroblasts [1063], keratino- Arthritic synovial fibroblasts constitutively oversecrete
cytes [1063, 2281], mast cells [1337] and eosinophils IL-8; IL-1β, itself elevated in synovial fluid, stimulates
[2261]. Certain tumor cells also produce IL-8 including secretion further. Specific inhibitor studies showed that
melanoma [1737], squamous cell carcinoma [2281] NF-κB, an important signal for inflammation genes,
colon adenocarcinoma [1757]. IL-8 secretion is con- was involved in both spontaneous and IL-1-induced
trolled by various factors such as IL-1 [1063, 1757], expression of IL-8 [612].
IL-3 [2121], IL-5 [1769], GM-CSF [1264, 2121], TNF IL-8 or other chemokines released within tumors may
[1063, 1904], vitamin D [2281], lipopolysaccharide have anticancer effects by recruitment of macrophages,
[1757] and fibrin [1603]. Mast cell secretion of IL-8 was granulocytes and lymphocytes. But there is also evi-
specifically stimulated by stromal cell factor-1 [1130]. dence that IL-8 contributes to tumor development and
There are two receptors for IL-8, CXCR1 and CXCR2 spread. In melanoma, IL-8 has been shown to be an
[5, 787, 1074, 1371, 1400]. These receptors bind IL-8 autocrine growth factor [1737, 1833]. IL-8 stimulated
with high affinity and they also bind other members of tumor cell movement. Metastasis of melanoma cells in
the chemokine family [1074]. There are receptors for nude mice was correlated with the tumor’s capacity to
IL-8 on neutrophils, T cells, mast cells, macrophages, produce IL-8 [1833]. Developing tumors require
180 Cytokines

adequate blood supply and may benefit from IL-8- IgE in several Th cell clones. These are mast cell-like
induced angiogenesis [994, 1905]. IL-8 secretion cor- characteristics that IL-9 induced in Th cells [1161]. In
related with vascularity in human gastric cancer [976]. cultures of mast cells, IL-9 enhanced survival; IL-9
In a nude mouse model for human tumor progression, together with IL-3 and IL-4 stimulated mast cell growth
IL-8 was one of several angiogenesis factors that corre- and the secretion of IL-6 [819]. Proliferation of mast
lated with progression in ovarian cancer [2250]. Reduced cells was one of the remarkable features of IL-9 trans-
IL-8 expression or blocking antibodies administered to genic mice [631]. In B cells, IL-9 potentiated IL-4-
nude mice decreased growth, invasiveness and angio- induced immunoglobulin secretion [461]. IL-9 increased
genesis in melanoma, breast cancer and transitional cell resistance to nematode infections [515].
carcinoma cells [812, 966, 1299]. Therefore, IL-8 may Mice that oversecreted IL-9 showed increased allergen-
act directly to stimulate tumor cells. Alternatively, IL-8 induced inflammation and airway hyperresponsiveness
may activate neighboring normal cells to produce factors [1279, 1977]. There is genetic evidence that IL-9 is
that tumors utilize for growth and metastasis. involved in human asthma [451, 1450, 1460].
Related to cancer, IL-9 has been shown to stimulate
growth and depress apoptosis in mouse lymphoma cells
[1633, 2079]. Transgenic mice overexpressing IL-9
Interleukin-9 tended to develop lymphomas [1632]. HTLV-transformed
Mast Cell, T Cell, Lymphoma Growth T cells produce IL-9 [947]. IL-9 was an autocrine growth
factor for cultured Hodgkin’s lymphoma and Reed-
Factor; Inflammatory; Hematopoietic
Sternberg cells [684]. It is possible that antagonists of
IL-9 is a 32–39 kDa, 144 amino acid protein. Its gene is IL-9 may be useful therapeutically.
located on chromosome 5 in a region near genes for
GM-CSF, IL-3, IL-4 and IL-5. IL-9 is structurally
related to IL-2, IL-4, IL-7 and IL-15 and some of their
activities are similar. IL-9 was discovered as a factor in Interleukin-10
the medium of HTLV-transformed T cells that stimu-
lated growth in a human leukemia cell line [2229]. IL-9
Regulator of Immune Response
has also been referred to as P40 [2063, 2170], mast cell IL-10 was first discovered in cultures of Th2 cells as a
growth enhancing activity [1332] and T cell growth fac- factor that inhibited IFN-γ production by activated Th1
tor III [1748]. It is produced by activated Th2 cells cells [535]. It is a 17,000 mw protein with relatively lit-
[1634], by naive CD4+ cells [1747], and by mast cells tle glycosylation [1177]. IL-10 is produced by several
[820]. In CD4+ cells, IL-9 secretion is stimulated by types of cells including regulatory T cells (Tr1 and
IL-1, IL-2, IL-4 and TGF-β and inhibited by IFN-γ nTreg) [2002, 2204], CD4+ Th2 cells [297, 535, 2262],
[1745, 1746]. In mast cells, IL-1, IL-10 and kit ligand thymocytes [1198], B cells [629, 1470], monocytes
increase IL-9 secretion [820, 1899]. [423, 548], mast cells [1990], eosinophils [1416], and
The receptor for IL-9 contains IL-9Rα and γc [968]. keratinocytes [494]. In CD4+ T cells, activation
The signaling pathway involves Jak3, Jak1, the adaptor induces several cytokines including IL-2 as an early
protein IRS-1 [2245] and the transcription factors response and IL-10 as a relatively late regulatory
STAT1, STAT2, and STAT5 [412, 116]. response [332, 2262]. Blocking endogenous IL-2 pre-
IL-9 usually acts together with other cytokines. With vented the increase in IL-10 [332]. IL-4, IL-7, IL-15 and
IL-3 or GM-CSF, IL-9 induced the growth of hematopoi- IL-12 were costimulators of IL-10 production. IL-10
etic progenitor cells [784]. With erythropoietin, IL-9 feeds back on the production of IL-2 and its own synthe-
supported erythroid colony (BFU-E) formation [447, sis by deactivating T cells [332]. In macrophages, activa-
2170]. In mice, IL-9 with IL-2 stimulated fetal thymo- tion induced several cytokines including an early
cyte proliferation [1917]. IL-9 increased growth in T response for TNF-α (3.5 h) and a much later peak in
cell lines and activated helper T cells [800]. Interestingly, IL-10 (48 h) [388, 423, 2116]. Cortisol (immunosup-
quality of growth induced in certain Th cell lines by pressive) stimulated plasma IL-10 levels in treated nor-
IL-9 was different from that of IL-2 in that IL-9 induced mal volunteers [376]. IL-10 feeds back on its own
growth was hardly affected by glucocorticoids while production by deactivating macrophages and TNF-α
IL-2-induced growth could be inhibited [1162]. IL-9 secretion [388, 423, 2167].
also increased granzyme B (protease involved in apop- The IL-10R structurally resembles the IFN receptor
tosis and cytotoxicity) and high affinity receptors for [773, 1014, 1869]. Signaling in monocytes and T cells
Walter M. Lewko and Robert K. Oldham 181

involves tyk2, Jak 1, STAT1α, STAT3 and a different IL-2 acted together synergistically, as IL-2R levels were
STAT3-like protein in monocytes, not observed in T increased [547, 867]. IL-10 enhanced class II MHC
cells [533]. expression [629] and stimulated the production of IgG,
IL-10 is a key regulator of immune response. A major IgA and IgM [400, 867]. In B cells, IL-10 had opposite
effect of IL-10 is the suppression of Th1-dependent cel- effects on apoptosis that appeared to depend on the acti-
lular immunity and the promotion of Th2-dependent vation state of the cells. IL-10 depressed apoptosis in
humoral immunity [1347]. IL-10 deactivates a number germinal center B cells [1107] but it enhanced apoptosis
of macrophage activities. It inhibits macrophage pro- in chronic lymphocytic leukemia B cells [546]. In
duction of IL-12 and other cytokines that stimulate T Staphylococcus aureus-activated B cells, IL-10 added 3
cells [173, 369, 388, 536]. Further, IL-10 depresses days later inhibited apoptosis; when IL-10 was added at
macrophage production of peroxide [173] and nitrogen the time of S. aureus, apoptosis was stimulated [866]. In
oxide [600]. In activated monocytes, IL-10 also inhibits mast cells IL-10 acts together with IL-3 and IL-4, to
the production of M-CSF. stimulate growth [1990].
IL-10 regulates neutrophil activity. It inhibits endotoxin- IL-10-deficient knockout mice exhibited a number of
induced release of proinflammatory cytokines [920]. IL-10 problems, in particular the mice developed enterocolitis
depresses neutrophil production of MIP-1α, MIP-1β and due to unregulated immune response to microbes [1031].
IL-8 [920] and enhances the release of IL-1Ra [257]. IL-10 protects mice against the effects of endotoxin
IL-10 inhibits antigen presentation and T cell response shock [613, 802].
[537]. In target cells, IL-10 down-regulates MHC class I Epstein Barr Virus genes code for vIL-10 (BCRF1).
[1243]. In monocytes, IL-10 depresses costimulatory The protein is highly homologous with hIL-10 [806,
and adhesion molecules involved in antigen presentation 1348, 2076]. It appears that at some point in evolution-
[2167]. IL-10 also inhibits dendritic cell-induced pro- ary time, EBV picked up this gene from a mammalian
duction of IFNγ by Th1 and CD8+ T cells [1188]. It cell source. viral IL-10 is expressed soon after infection,
decreases antigen presentation by macrophages to T within 2–3 h, whereas host IL-10 appears 20–30 h after
cells, in particular, to Th1 cells [388]. IL-10 also infection. viral IL-10 expression interferes with antigen
depresses T cell growth directly [1942]. In mitogen stim- presentation and the antiviral immune response [423].
ulated T cells, IL-10 inhibits cell division and production IL-10 functions during pregnancy. The uterus pro-
of IL-2 and IFN-γ [1942]. In many ways IL-10 resembles vides a site of immune privilege for the development
IL-4, but interestingly, IFN-γ levels are stimulated by of a fetus. Successful pregnancy is said to be a Th2
IL-4. This is the key difference in activities between IL-4 phenomenon [2127]. In an abortion-prone mouse
and IL-10 which otherwise act similarly as they tend to model system, a defect in IL-10 production appeared
suppress cell mediated immune response. to be responsible for fetal loss; treating the mice with
In NK cells, IL-10 depresses INF-γ secretion and IL-10 prevented fetal resorption [313]. The pregnancy
cytotoxicity induced by IL-12 and TNF-α [2027]. In hormone progesterone induces the production of
fibroblasts, IL-10 depresses the production of collagen, IL-10 [709]. In human trophoblasts, IL-10 is an inhib-
enhances the production of collagenase and stromelysin itor of matrix metallopoteinase-9 production, a pro-
[1628]. In macrophages, it inhibits production of colla- tease believed to have a role in parturition [1682].
genase and stimulates the production of tissue inhibitor IL-10 production in the placenta was shown to
of metalloproteinases (TIMP) [1052]. As already noted, decrease at term. Lower IL-10 levels would allow the
glucocorticoids, which are immunosuppressive, stimu- expression of MMP-9 during parturition and a shift
late IL-10 levels in blood [376]. The overall effect of from Th2 to Th1 immunity [709].
IL-10 is antiinflammatory and it tends to limit tissue IL-10 may have a role in human diseases involving
damage associated with inflammation. inflammation and inappropriate immune response.
IL-10 has been described as a growth and differentia- In volunteers treated with a single i.v. dose of IL-10, T
tion factor for CD8 T cells [297, 1198] and activated B cell levels were decreased. In endotoxin treated volun-
cells [1687]. In T cells, IL-10 induced the IL-2R and teers, pretreatment with IL-10 lowered fever and sev-
thereby enhanced IL-2-dependent proliferation [1198, eral proinflammatory cytokines [300, 1525]. IL-10
331]. IL-10 is a chemotactic factor for CD8+ T cells (the depresses lung granulocyte number and capacity for
first described) but it is an inhibitor of IL-8-induced degranulation [1525]. IL-10 inhibits collagen produc-
migration for CD4+ cells [893, 895]. In a model for dia- tion in fibroblasts. In patients with chronic hepatitis C,
betes, IL-10 expression by islet cells caused the local IL-10 normalized serum ALT (marker for hepatic
accumulation of T cells [2181]. In B cells, IL-10 and inflammation), improved liver histology and reduced
182 Cytokines

fibrosis [1440]. Most patients with psoriasis benefited cells expressing either IL-10 of IL-12 alone yielded little
from treatment with IL-10 [77, 1627]. In Crohn’s dis- or no response. CD8 and CD4 cells were both involved
ease, responses to IL-10 were observed in 23.5% and tumor specific antibodies were induced [677].
patients. In another study, patients treated post surgi-
cally to prevent recurrence of Crohn’s disease did not
show any benefit [336]. IL-10 treatments were well
tolerated; observed toxicities, were moderate and
Interleukin-11
reversible. In another approach, blocking antibodies to Hematopoietic Progenitor/
IL-10 were used to treat a small group of SLE patients. Thrombocytosis Stimulator;
The patients were treated for 21 days; five/six patients
benefitted from the antibody therapy [1142]. The
Regulator of Inflammation
immunosuppressive and antiinflammatory properties IL-11 was discovered as a factor produced by a bone
of IL-10 may be of value in tissue transplantation marrow stromal cell line, which supported the growth
[125]. But it should be noted, certain mouse studies of an IL-6-dependent plasmacytoma cell line [1543]. It
did not show any IL-10 benefit [1022]; in some cases is a 22 kDa glycoprotein [455, 456]. IL-11 is a member
IL-10 exacerbated graft vs. host disease [163]. of the IL-6 cytokine family. It is produced by stromal
Although IL-10 is generally antiinflammatory, it has cells in hematopoietic tissues [1543], fibroblasts [486,
certain proinflammatory effects, which may compli- 1653, 2290], epithelial cells [485], chondrocytes and
cate its use in therapy. For example, one study showed synovial cells [1200] and by airway smooth muscle
that IL-10 potentiated IFN-γ release in LPS-treated cells [484]. IL-11 is induced by IL-1, TGF-β, hista-
volunteers [1066]. mine, eosinophil major basic protein and by certain
IL-10 may be elevated in cancer patients, naturally viruses [484, 485, 481]. The receptor for IL-11 shares
due to immune response or due to IL-10 production by the gp130 subunit with receptors for the other IL-6
tumors. Increased IL-10 could be involved in tumor family members [1138, 1944].
growth and immune escape [882, 1173, 1574]. It has IL-11 stimulates the growth and differentiation of
been shown that Epstein-Barr viral IL-10 is an autocrine hematopoietic progenitor cells and megakaryocytes
growth factor for certain B cell lymphomas [121, 957]. [1404, 1405]. It influences very early stem cells and more
IL-10 is also a growth factor for human myeloma cells, committed precursor cells. In mouse cells, IL-11 acted
apparently by induction of a gp130 R dependent (e.g. synergistically with IL-3 and IL-4 to stimulate growth of
LIF) process [689]. IL-10 may interfere with antigen primitive blast colony forming cells [1504, 1405]. IL-11
presentation. Using antigen-pulsed DCs, repetitive vac- stimulates B cells and the production of immunoglobulin
cination induced CD4+ cells, which produced IL-4 and [1543, 2244]. IL-11 has a remarkable effect on platelet
IL-10 [282]. IL-10 appeared to induce T cell anergy production and this is its main clinical use [456].
[282, 1173, 1934]. In tumor cells, IL-10 decreased IL-11 inhibited the differentiation of fibroblast lines
expression of HLA class I, which would decrease recog- into adipocytes; IL-6 and TNF-α had similar effects
nition by effector cells [1243]. In Hodgkin’s disease, [937]. IL-11 inhibited the production of IL-12 by mac-
elevated IL-10 was determined to be an independent rophages [1104]. IL-11 is involved in bone metabolism;
prognostic factor for treatment failure [174, 1727]. It it is said to be critical for osteoclast development [625].
thus appears IL-10 may have a role in tumor progression. Also IL-11 is among the several cytokines capable of
It should be mentioned that during immunotherapy, inducing acute phase protein release from hepatic and
IL-10 levels might rise [1996]. This could be a reason nonhepatic cells [1340].
for lack of response. IL-11 regulates Th2 responses and the production of
One might not expect IL-10 to be useful in a tumor inflammatory cytokines [2022, 2296]. IL-11 is secreted
vaccine. However, interesting results were obtained in during respiratory tract infections, airway inflammation,
mice vaccinated with mammary adenocarcinoma cells and asthma [481, 1229, 2103] and certain other inflam-
engineered to produce IL-10 [30, 624]. Some mice matory responses such as Lyme disease [53]. IL-11 also
developed long term immunity. In another study, trans- ameliorates inflammatory bowel disease and oral
genic mice overexpressing IL-10 in antigen presenting mucositis [958]. IL-11 protects and restores damaged,
cells rejected an immunogenic melanocytoma [678]. In inflamed tissue by the secretion of protease inhibitors
another study, mice received tumor cells expressing both [1202]. In an ovalbumin allergy model, lung overex-
IL-10 and IL-12. 50–70% showed remission of metasta- pression of transgenic IL-11 decreased antigen-induced
ses to lung, colon and breast tumors. Vaccination with eosinophilia, inflammation and endothelial VCAM-1 in
Walter M. Lewko and Robert K. Oldham 183

lung tissue. Th2 cytokines IL-4, IL-5 and IL-13 were [680]. These chains are produced from two distinct
diminished [2102]. genes, on separate chromosomes [680, 1822]. The α
IL-11 also has protective effects during radiation- subunit has homology with IL-6 and G-CSF [1293]. The
induced thoracic injury [1623] and intestinal damage β subunit is unusual in that its structure is more like that
[455]. IL-11 protects against immune complex lung of a receptor than a cytokine [603]. Expression of these
injury [1095] and oxygen-induced lung damage [2123]. two genes does not appear to be coordinated. Curiously,
In the care of cancer patients, IL-11 augments bone many cells in the body produce the α subunit without
marrow recovery and platelet production. It is approved making β [368]. But both chains must be transcribed
for use in patients with non-myeloid malignancies to within the same cell for the production of active IL-12
prevent thrombocytopenia and to reduce the need for [2185]. Regulation of IL-12 secretion generally occurs
platelet transfusion following chemotherapy. It has been at the level of β chain synthesis [368] though this wasn’t
shown in breast cancer patients, for example, that treat- the case in DCs prepared from newborns where low
ment with rhIL-11 increased bone marrow megakaryo- IL-12 production appeared to be due to lack of the α
cytes [1503]. Blood platelet counts were increased and chain. Low DC IL-12 may be one reason why newborns
patients experienced less thrombocytopenia [645]. In have relatively poor cellular immune response [646].
patients with thrombocytopenia requiring platelet trans- Mouse cells secrete an IL-12 β-β homodimer that
fusions, treatment with rh-IL-11 significantly decreased acts as a physiological antagonist of IL-12 [1252, 595,
the need for transfusions with subsequent chemotherapy 731]. It is not clear whether the β-β homodimer has a
[1978]. RhIL-11 is well tolerated by adults and children. function in humans.
Adverse events are generally mild or moderate and IL-12 is produced by macrophages, dendritic cells,
reversible [1843]. neutrophils, microglial cells, keratinocytes and trans-
Il-11 may have a role in progression of breast cancer. formed B cells [368, 1189, 804, 1608]. IL-12 is not typi-
IL-11 and its receptors were higher in tumors than normal cally secreted by tumors except certain cancers of B
tissue and higher in node positive tumors than node cells origin [2185]. IL-12 production in macrophages is
negative tumors. Higher expression of IL-11 in tumors stimulated by bacteria, viruses, parasites [368, 804,
was linked to poorer survival of patients [708]. IL-11 2185] and by contact with T cells through CD40–CD40L
also induces the formation of osteoclasts, which appear interaction [1817]. IL-12 production is enhanced by
to be involved in the metastasis of breast cancer to bone IFN-γ (Th1 cytokine) and inhibited by IL-10, IL-4 (Th2
[918, 1353, 1830]. cytokines), TGF-β and IFN-α/β [2108].
IL-12 receptor contains two chains, IL-12Rβ1 and
IL-12β2. They have homology with the β chain (gp130)
Interleukin-12 of the IL-6R family [318]. Both subunits are required
for IL-12 function. β1 is responsible for binding. β2 is
Immune Stimulation, Inflammation, the subunit that signals [2200, 2201]. IL-12R is
Hematopoiesis, Regulator of Innate and expressed mainly on NK cells and T cells [416]. It is not
expressed on Th2 cells [1939]. In PBMC, IL-12R was
Adaptive Immunity upregulated by activation with PHA or IL-2 [416].
IL-12 is a key cytokine with far reaching influences. It IL-12R signaling involves Tyk2, Jak2, STAT3 and
serves as a link between the innate and adaptive immune STAT4 and p38 MAPK [97, 326, 873, 2282, 2303].
response systems [993, 2025, 2186]. IL-12 is a well IL-12 promotes the development of cellular immune
studied cytokine. It was reviewed in some detail in a response. It does this by stimulating the production of
previous edition [1110]. The following is a brief over- IFN-γ and Th1-related cytokines in PBL, T cells and
view with a discussion of certain new reports related to NK cells [284, 317, 824, 993, 1028, 1283, 1284, 1554,
cancer. 2202]. IL-12 also stimulates hematopoiesis. It acts on
IL-12 was discovered in B cell lines as a factor, which progenitor cells together with growth factors such as
acted with IL-2 to stimulate proliferation, IFN-γ secre- GM-CSF [130, 1431, 1432, 1433]. IL-12 increases DC
tion and cytotoxicity in NK cells and T cells [596, 737, production of cytokines (GM-CSF, IL-1β, IL-6, IL-12,
1149, 1560, 2054]. It has been referred to as natural TNFα and IFN-γ) and antigen presentation [675, 1028,
killer cell stimulating factor [993], T cell stimulating 1412]. IL-12, together with IL-2, stimulates the growth
factor-1 [649], and cytotoxic lymphocyte maturation and differentiation of B cells [885]. IL-12 induces LAK
factor [1891]. IL-12 is a 70,000 mw glycoprotein; it is cells when added to cultures of PBLs for 3–5 days. LAK
composed of two chains, α (also called p35) and β (p40) cells are also induced by IL-2. At least part of the IL-2
184 Cytokines

effect on LAK appeared due to IL-12 for antibodies to IL-12-induced, CD8+ mediated immunity to mouse
IL-12 decreased the response [597, 1431]. neuroblastoma [1559]. In mice with brain tumors, IL-12
In anti CD3 activated TIL, IL-12 stimulated growth induced a T cell response, decreased tumor size, and
and tumor cell lysis [50]. The effect of IL-12 was not prolonged survival time [1690]. These studies suggest
prevented by antibodies to IL-2. When IL-12 was that systemic IL-12 may be of benefit in the treatment of
added to cultures with a low, suboptimal dose of IL-2, CNS cancers.
there was an additive effect on growth and cytotoxic- Several studies have shown that IL-12 inhibits angio-
ity [50]. Growth induced by IL-12 alone was tran- genesis in tumors [249, 349, 460, 606, 1898, 1964]. The
sient. Maintenance of growth after 72 h required IL-2. process involves a complex interaction between several
IL-2 appears to be more potent than IL-12. It remains types of cells. IL-12 does not appear to act directly on
to be seen whether IL-12-treated TIL cells have added endothelial cells [460, 1898]. Rather IL-12 induces
clinical benefit. IFN-γ, which in turn induces IP-10 and Mig; these two
IL-12 appeared to be involved in inflammatory dis- chemokines act directly on endothelial cells, to induce
eases such as multiple sclerosis, diabetes, and arthritis vascular damage, clotting and tumor necrosis [52, 64,
[2025]. However, more recent studies suggest IL-23, 350, 1874, 1898, 1964]. Vasostatin is another antiangio-
which is proinflammatory and structurally related to genic factor. In nude mouse studies, treatment with
IL-12, may be responsible for certain effects attributed vasostatin or IL-12 inhibited tumor growth. Together
to IL-12, specifically in experimental allergic encephalitis. they effectively blocked Burkitt’s lymphoma, colon
Further, IL-12 may be more regulatory, suppressing carcinoma, and ovarian carcinoma [2230].
TNF and other proinflammatory cytokines [2279]. IL-12 engineered cells show promise as anticancer
IL-12 secretion by antigen presenting cells may be vaccines. IL-12 transfected tumor cells [264, 1152] or
responsible for differences in immune response between fibroblasts [2299] inhibited the growth of some estab-
females and males [2161]. Females tend to have more lished tumors and induced the rejection of subsequent
vigorous humoral and cellular immunity. Females also tumor implants. In a model using an MHC I negative
have a higher incidence of autoimmune disease. An small cell lung cancer growing in nude mice (no capac-
interesting study showed that activated AP cells from ity for CTL response), cells engineered to produce both
SJL female mice secreted IL-12 but not IL-10 and IL-15 and IL-12 did not grow; coinjected wild type
favored Th1 response. These female mice suffered a tumor was also rejected. This study showed that IL-12
high incidence of experimental allergic encephalomy- could stimulate innate antitumor immunity and that
elitis. In males, AP cells produced IL-10 but not IL-12 macrophages appeared to be involved [427].
and had a much lower susceptibility to the disease. Dendritic cells transfected by bombardment with
Castration or estrogen treatment increased IL-12, DNA-coated gold particles containing genes for tumor
decreased IL-10 and increased the incidence of this antigens express antigen and presented it. Co-transfection
autoimmune disease [2161]. of these dendritic cells with IL-12 or IFN-α consistently
Anticancer activity of IL-12 may be related to any of enhanced the induction of specific CTLs in vaccinated
a number of direct or extended effects that this cytokine mice [2039].
has on Th1 differentiation, CTLs, dendritic cells, mac- IL-2 and IL-12 interact; receptors are mutually stim-
rophages, NK cells, NK T cells, and vascular endothe- ulated [416] and they have additive or synergistic
lial cells [1848]. IL-12 induces several cytokines; IFN-γ effects on NK cells [394], T cells [596, 1284] and mac-
is a major down stream mediator of IL-12 antitumor rophages [2157]. Administration of IL-12 with pulse
activity [1898, 2297]. As a single agent, IL-12 inhibited IL-2 induced complete regression of established mam-
a number of mouse model tumors [216, 1425]. Mice mary carcinoma in treated mice [2158]. IL-12 has also
bearing B16 F10 melanoma, M5076 sarcoma and Renca been reported to potentiate the effects of tumor cell
renal carcinoma benefitted from treatment with IL-12. vaccines engineered to secrete IL-2 for colon cancer
The antitumor effect did not require NK cells but did [2048] and glioma [981].
depend on T cells, specifically CD8+ T cells [216]. Mice Time-release microcarriers have been used to deliver
with sarcomas, treated with IL-12, showed decreased a single dose of adjuvant IL-12 by intratumoral injection.
tumor growth, increased longevity and in some cases IL-12 alone and especially with GM-CSF, activated T
complete regressions. Interferon γ was required for the effector/memory cells, killed regulatory T cells and
IL-12 effect and it appeared to be mediated by CD4+ increased CD8+ effector cells. Elimination of suppression
and CD8+ T cells [1425]. Downstream the chemokine and development of cytotoxic T lymphocytes eradicated
IP-10 (interferon inducible protein) was responsible for metastatic tumors [960].
Walter M. Lewko and Robert K. Oldham 185

The preclinical studies with IL-12 were quite The receptor for IL-13 has two subunits, IL-4Rα and
encouraging. This cytokine had many effects similar to IL-13Rα1 [741, 758]. This complex also binds and
those of IL-2 and appeared to be less toxic. For these responds to IL-4. Signaling involves Jak1 and Tyk2 [943,
reasons and others, IL-2 and IL-12 may prove to be 944]. In addition, there is IL-13Rα2, which may be solu-
effective in combination or sequential biotherapy. ble or membrane bound [244, 445, 1940]. The soluble
Similarly, IL-12 gene insertion may prove an effective form appears to be a non-signaling decoy receptor [2197];
strategy for increasing tumor immunogenicity. Finally, the membrane bound form may have certain functions
ex vivo expansion of T cells with IL-12 in addition to [302, 531].
IL-2 might be rendered more specific and more cytolyt- IL-13 appears to have remarkable effects on T cells;
ically effective. Studies from our laboratory and others Th2 cells are stimulated and Th17 cells are inhibited.
indicate that IL-12 is effective in stimulating cytolytic But the receptor for IL-13 is not found on T cells; these
populations of tumor derived T cells and may be of effects of IL-13 must be indirect. In DC for example,
benefit in the development of T-cell based therapy. IL-13 inhibits expression of IL-6, a key factor necessary
There have been several reports on phase I/II studies for Th17 differentiation [985, 1276, 2310].
using IL-12. Patients with cutaneous T cell lymphoma, In monocytes, IL-13 is anti-inflammatory and pro-al-
injected with rhIL-12, had a response rate (CR + PR) of lergy. IL-13 decreased production of several pro inflam-
50% [1661]. IL-12 has also been tested in patients with matory cytokines including IL-1α, IL-1β, IL-6, IL-8,
renal cancer [84, 635, 1379, 1484, 1588, 1650], mela- IL-10, IL-12, GM-CSF, G-CSF, MIP-1α, TNF-α, GROα
noma [84, 635, 1075, 1367, 1650, 1928] and ovarian can- and prostaglandin E2 [422, 1315, 1598, 2198]. IL-13
cer [829, 1096]. IL-12 has been used in combination with also inhibited antibody-dependent cell-mediated cyto-
Herceptin in breast cancer patients [1534] and with IFN- toxicity [422]. IL-13 caused remarkable changes in
α2b in with renal cancer and melanoma patients [832]. monocyte phenotype and adherence. It stimulated levels
Vaccination studies have been carried out using IL-12 as of complement receptors CD11b and CD11c. IL-13 also
an adjuvant with peptide-pulsed antigen presenting cells stimulated MHC class II levels. IL-13 increased CD23
[581] and using transfected, IL-12-secreting autologous (an IgE receptor). IL-13 also increased CD49e and
tumor cells [1928]. Patients have also been injected intra- CD29, which together form VLA5 (integrin α5β1, a
tumorally with transfected, IL-12-secreting fibroblasts fibronectin receptor) [162]. IL-13 depressed levels of
[917]. Collectively, these studies showed that IL-12 CD64, CD32 and CD16 (IgG receptors) [422]. IL-13
induced immune response; NK cell and specific CTL decreased CD14 (LPS receptor) [162, 339]. IL-13 pro-
activities were increased and serum cytokine levels were tected animals from the lethal effects of LPS-endotoxemia
elevated, in particular, IFN-γ. Toxicities were significant [1382]. IL-13 increased production of IL-1 receptor
but manageable. Lymphopenia was the most common antagonist [422, 1408]. IL-13 is also a potent suppressor
problem. Clinical activity was observed but the response of nitric oxide production in activated macrophages, epi-
rates were disappointingly low (generally less than 10%) thelial cells and mesangial cells [139, 172, 286, 1733].
and not durable. In order to improve the response rate, IL-13 does this by regulating levels of nitric oxide syn-
IL-12 may have to be administered in combination with thase and substrate arginine available for production
other cytokines, IL-2 for instance [635]. [286, 1733]. Further, IL-13 inhibited the production of
HIV-1 virus in cultured human macrophages [1343].
In B cells, IL-13 increased proliferation, the synthesis of
immunoglobulin, and IgE class switching [328, 398, 1275,
Interleukin-13 1597]. IL-13 also enhanced the expression of MHCII and
CD23 (an IgE receptor) [1277]. Essentially all of these
Regulation of Inflammation; Allergy effects of IL-13 in B cells are similar to those of IL-4.
IL-13 is a Th2 cytokine that has a role in parasite infec- IL-13 has various additional activities. In neutrophils,
tions, allergy and the regulation of inflammation. IL-13 induced the secretion of interleukin-1 receptor
Recombinant IL-13 has a molecular weight of about antagonist [337]. IL-13 also stimulated the production of
14,000. It is related to IL-4 [1274, 2310]. Activated Th2 IL-6 in human keratinocytes [415] and in human micro-
CD4+ T cells, CD8+ T cells [908, 1275, 1315, 1597], glial cells [1773]. In NK cells, IL-13 induced the produc-
mast cells [225], eosinophils [2180], basophils, dendritic tion of IFN-γ and cytotoxicity [1315]. IL-13 is a chemotactic
cells and certain B cell lines produce IL-13. The gene of factor for human osteoblasts [1131]. In endothelial cells,
IL-13 is located on human chromosome 5 together with IL-13 stimulates production of VCAM enhancing T cell
genes for IL-3, IL-4, IL-5 and IL-9 [1271]. adhesion [620]. IL-13 is a chemoattractant for monocytes
186 Cytokines

[1200] and stimulates production of monocyte chemoat- memory [40, 1479]. Mice that were transgenic for IL-14
tractant protein-1 in vascular endothelial cells [633]. developed autoimmunity and B cell lymphomas, which
IL-13 is involved in inflammatory diseases. IL-13 reg- resembled systemic lupus in patients [1803]. IL-14 is
ulates rheumatoid arthritis. Cultured explants of synovial secreted by B cell malignancies and may have a role in
tissue from patients contained elevated levels of IL-1β, the development of these diseases [552].
TNF-α and PGE2. Transfection with IL-13 depressed the
production of these inflammatory factors [2198]. While
IL-13 is generally antiinflammatory, it does have certain
proinflammatory effects. IL-13 has a central role in the
Interleukin 15
development of allergic asthma [681, 2175]. IL-13 is a NK Cell Activity, Maintenance
Th2 cytokine. It fosters humoral immune response and
of T Cell Memory
the production of IgE. Bronchoalveolar lavage cells from
asthmatic patients challenged with ragweed allergen pro- IL-15 was discovered as a factor in culture media of
duced high levels of IL-13 [813]. Antigen stimulated monkey kidney epithelium that supported IL-2 depen-
CD4+ cells of patients with allergic rhinitis produced dent growth in a T cell line. The same factor was identi-
more IL-13 compared to normal controls. fied in a human bone marrow stroma cell line [647].
There are some reports on IL-13, relating to cancer. IL-15 was also discovered independently, as the factor
Renal cell carcinomas produce IL-13 and IL-13R IL-T in a T cell leukemia cell line [108, 228].
[1474]. Addition of IL-13 to the medium of cultured IL-15 is a 14–15 kDa, 114 amino acid protein. It a
cells inhibited growth up to 50% [1475]. Human glioma member of the four α helix bundle cytokine family to
cells contained unusually high levels of the IL-13R. which IL-2 belongs [647]. Many of its activities are
Tumor growth was inhibited by an IL-13-exotoxin con- similar to those of IL-2. While IL-2 is produced mainly
jugate [393, 830]. by activated T cells, IL-15 is produced by a rather wide
IL-13 is angiogenic. It stimulated the formation of variety of cells types including peripheral blood mono-
vessel-like structures from endothelial cells in collagen nuclear cells, epithelial cells, fibroblasts, placenta, skel-
culture and in vivo it stimulated outgrowth of vessels in etal muscle, heart, lung, liver, kidney [647], and
the rat cornea assay [574, 703]. The mechanism appeared keratinocytes [1334]. IL-15 was not detected in acti-
to involve the production of VCAM, which in its soluble vated T cells [647]. While IL-2 synthesis is regulated at
form is angiogenic. the level of transcription and mRNA stability, IL-15 is
IL-4 is used together with GM-CSF for in vitro pro- regulated at the translational level [107] which is less
duction of dendritic cells in human vaccination studies. common but has been shown to occur for certain cytok-
IL-13 substitutes for IL-4 in the production of dendritic ines (e.g. IL-1β, TNF-α, TGF-β3, TGF-β1, GM-CSF).
cells [33]. It remains to be seen whether IL-13 may be of A pool of mRNA, readily available for translational
advantage in the development of anticancer vaccines. activation, may allow rapid production of IL-15 when
required, as in response to an intracellular infectious
agent [107].
The IL-15R is composed of three subunits: IL-15Rα,
Interleukin 14 IL-2Rβ and γC. The β and γc subunits are shared with
IL-2 receptors; IL-2 and IL-15 have similar signaling
B Cell Development and Memory patterns and cytokine activities [626]. And although the
IL-14 was discovered in the conditioned media of IL-15Rα chain binds IL-15 specifically, it is structurally
Burkitt’s lymphoma cell lines. It was originally referred related to IL-2Rα [46, 627]. IL-15R is expressed in a
to as high molecular weight B cell growth factor (HMW- greater variety of cell types than the IL-2R [46, 627].
BCGF) [39–41]. It is a rather large glycosylated protein Interestingly, mast cells respond to IL-15; they have a
with a variable mw of 53–65 kDa, depending on carbo- different IL-15 receptor and signal transduction path-
hydrate content [42]. IL-14 is produced by PHA stimu- way [1945].
lated CD8+ T cells, CD4+ T cells (Th1 and Th2), NKT There is a soluble form of the IL-15Rα chain. It is
cells, follicular dendritic cells [342, 1704] and B cell released from cells by proteolysis. It binds IL-15 with
lymphomas [552, 553]. Its mRNA has been detected in high affinity and it acts as an IL-15 antagonist [1369]
unstimulated vascular endothelial cells [1455]. There is evidence that IL-15 can exist in a mem-
IL-14 stimulates B cell growth and differentiation brane-bound form, which is capable of reverse/bidi-
[39, 41]. It also appears to induce and maintain B cell rectional signaling when engaged by its receptor.
Walter M. Lewko and Robert K. Oldham 187

This membrane bound IL-15 was upregulated in [1026]. On the other hand, IL-15 has anti-apoptotic
monocytes by IFN-γ [219]. effects [223] and increases the survival of CD8+ mem-
IL-15 stimulates proliferation and differentiation of ory cells [1026, 2284]. In mast cells, IL-2 has little or no
NK cells, B cells, and T cells. Knockout mice lacking effect while IL-15 stimulates growth and response to
IL-15Rα were deficient in NK cells, NK-T cells, CD8+ IL-3 and stem cell factor [1945, 2091].
T cells and γδ T cells [1145, 1457, 2172]. In NK cells, Human T cell lymphotropic virus codes for tax protein;
IL-15 stimulated differentiation and activation [249, when T cells were infected, tax expression increased the
1145, 1381, 1477, 2172]. IL-15 upregulated expression production of IL-15 [94, 95] and IL-15Rα [1227]. IL-15
of NKGD2 receptors, which are involved in antitumor appears to be responsible for the abnormal proliferation of
activity [1930]. IL-15 is also chemotactic for NK cells T cells associated with infection by this virus [95].
[29]. In B cells, IL-15 stimulated proliferation and secre- The anti-apoptotic effects of IL-15 may have a role in
tion of IgM, IgG, and IgA [71]. In activated cytotoxic T psoriasis, a chronic proliferative inflammatory skin
lymphocytes, it increased growth and cytotoxicity [617]. disease. Keratinocytes produce IL-15 and the IL-15 R.
IL-15 stimulated the growth of an IL-2 dependent CTL Compared with normal epidermis, biopsies of psoriatic
cell line [228]. IL-15 has been shown to rescue tolerant skin lesions were high in IL-15 and IL-15 binding
CD8+ T cells for use in adoptive anticancer therapy capacity [1693].
[1975]. IL-15 also increased growth and activity of γδ T Cellular immunity is responsible for allograft rejec-
cells [591, 1457]. IL-15, like IL-2, induced formation of tion. A study showed that blocking the effects of IL-15
LAK cells in cultures of peripheral blood lymphocytes using soluble IL-15Rα chain prolonged the survival of
[228, 617]. In primary cultures of tumor infiltrating lym- heart grafts. This indicated that IL-15 has a role in graft
phocytes, IL-15 replaced IL-2, inducing outgrowth and rejection and that IL-15 antagonists may be therapeuti-
cytotoxicity of tumor derived activated T cells (TDAC) cally useful in the treatment of graft recipients [1845].
[1111]. In TDAC cultures that were initially induced In a nude mouse xenograft tumor model, MHC I nega-
with IL-2 and dependent on IL-2 for growth, IL-15 could tive small cell lung cancer cells, engineered to secrete
replace IL-2 for the maintenance of growth [1111]. With IL-15, grew more slowly and had a slightly reduced take
increased awareness of the importance of IL-2 in Treg rate; tumors were infiltrated with NK cells and the deple-
immunosuppressive function, there has been consider- tion of NK prevented the antitumor effect. Interestingly,
ation for replacing IL-2, in the development of CTL for when engineered to secrete both IL-12 and IL-15, the
immunotherapy, with IL-15 alone or in combination with tumors were completely rejected, as were coinjected
IL-21 [56, 2275]. wild type tumors. Tumors were infiltrated with mac-
IL-15 is synergistic with IL-12 in the induction of rophages, NK cells and granulocytes. Activated mac-
mouse Th1 clones [92]; IL-15 appears to have a strong rophages appeared to be the major effector cells [426]. It
pro-Th1 tendency. In transgenic mice, overexpression has also been shown that in NK cells, IL-15 potentiates
of IL-15 augmented Th1 response to infection with IL-12-induced secretion of IFN-γ, MIP-1, and IL-10
intracellular bacterium [1457]. Increased IL-15 is asso- [521]. These studies show the importance of natural
ciated with inflammatory diseases including rheumatoid immunity and the synergistic action of IL-15 and IL-12
arthritis [1268, 1692], pulmonary sarcoidosis [12], mul- in the process. And in adoptive immunotherapy,
tiple sclerosis [982] and inflammatory bowel disease Certain tumors express IL-15R and are candidates for
[972, 1715]. On the other hand, IL-15 has been shown cytokine-based therapies. An antibody (Mikbeta1) to
to inhibit allergy. IL-15 overproduction in transgenic the IL-2/IL-15β subunit has been tested in Phase I and
mice had a negative effect on the development of aller- found not beneficial in T cell large granular lymphocyte
gic asthma, a Th2 disease; IL-15 depressed pulmonary leukemia [1363].
eosinophilia and the production of Th2 cytokines [860].
But it should be noted IL-15 may have some pro Th2
effects; in mice primed with dust mite allergen, IL-15
stimulated IL-5 production by allergen-specific human
Interleukin-16
Th2 clones and resulting eosinophil activation [1354]. Chemoattractive; Proinflammatory;
While similar in function, there are differences
Immunoregulatory; Anti-HIV
between IL-2 and IL-15. IL-2 has a role in tolerance by
inducing T cell suicide [1710, 1933, 2168], in activa- IL-16 was originally described in 1982 as the lympho-
tion-induced T cell death [1092, 1626, 2062] and it is cyte chemoattractant factor (LCF) secreted by mitogen-
involved in the inhibition of T cell memory maintenance stimulated peripheral blood mononuclear cells [268,
188 Cytokines

270, 360]. IL-16 is produced mainly by CD8+ T cells. It and inhibits chemokine signaling. In migrating lympho-
is also produced by CD4+ T cells [2205], B cells [921, cytes, IL-16 induces protein kinase C; PKC inhibitors
1797], fibroblasts [1770], eosinophils [1129], mast cells block migration induced by CD4 engagement [1532]. In
[1697], dendritic cells [922] and epithelial cells [70]. macrophages, signaling involved the phosphorylation
IL-16 is also secreted by brain tissue; it may have a role of two MAP kinase family members: the stress activated
in the interaction between the immune and nervous protein kinase/Jun N-terminal kinase (SAPK/JNK) and
systems [1038]. p38 MAP kinase. These two pathways are commonly
IL-16 is synthesized as a proprotein. Nascent IL-16 activated by environmental stress signals such as UV
lacks the usual signal peptide found on most secreted irradiation and by certain other proinflammatory cytok-
proteins [102]. This lack of a signal peptide is also a ines such as IL-1 and TNF-α [1021]. In T cells, the
characteristic of IL-1 [89], IL-18 [1488] and FGF [3, binding of IL-16 to CD4 resulted in activation of the src
884]. Pro-IL-16 is cleaved to its active form by caspase-3, family tyrosine kinase p56 lck, which is associated with
an enzyme in the same family as caspase 1 (ICE) which CD4 [1700]. In NK cells that expressed CD4, ligation
is responsible for the activation and secretion of IL-1β induced cytokine secretion and cell migration [141].
and IL-18 [2286]. The data favoring CD4 as the receptor are strong. But
Regulation of IL-16 release depends on the cell type. there is evidence in knockout mice genetically lacking
CD8+ T cells produce IL-16 constitutively and store it CD4, that IL-16 was in fact still capable of inducing
in the active form; histamine, serotonin, mitogen and cytokine production and lymphocyte migration [1239].
antigen stimulate IL-16 release [271, 1047, 1048]. In a mast cell line that lacks CD4, IL-16 mediated
CD4+ cells, on the other hand, store inactive pro-IL-16; chemotaxis and signaling in a manner that could be
T-cell activation increases caspase-3 activity, pro-IL-16 blocked by antibodies to CD9. This suggested that CD9,
processing and the secretion of active IL-16 [2203]. In a cell surface protein known to be involved in migration
bronchial epithelial cells, histamine, IL-1β and TNF-α and cell adhesion, serves as an alternative receptor for
stimulate secretion. Dexamethasone inhibits secretion; IL-16 [1604].
regulation of IL-16 may be part of the glucocorticoid IL-16 influences several cell types. The major effect
antiinflammatory effect [70]. In fibroblasts, IL-16 is chemoattraction but it also has effects on immune
mRNA is produced constitutively. Inflammatory cytok- response and HIV replication. IL-16 responsive cells
ines such IL-1β induce the release of active IL-16 by a are typically CD4 positive [269, 271]. IL-16 sensitive
caspase-3 dependent mechanism [1770]. cells include CD4+ T lymphocytes [270, 357, 359,
The amino acid sequence for IL-16 is distinct; it has 1532], monocytes [357, 739, 1240], dendritic cells
little or no homology with other known cytokines. The [739, 921, 922, 411], eosinophils [1618], mast cells
monomer of IL-16 appears to be inactive; it aggregates [1604] and, interestingly, brain cells that are also
to form a 56,000 mw tetramer, which is the active CD4+ [1038]. In resting T-cells, IL-16 induced sig-
cytokine [102, 359, 360, 2286]. The C-terminal region is naling and activation with migration and increased
particularly well conserved between species and appears IL-2 receptor levels [357, 358, 360, 1532, 1700,
to be the region most critical for cell binding and activity 1981]. Several chemotactic agents induce migration
[102, 941]. Interestingly, the N-terminal prodomain in both CD4+ and CD8+ T cells; IL-16 is specific for
cleaved from pro-IL-16 by caspase-3 has a cellular func- CD4+ cells [2271]. It modulates and desensitizes
tion. It translocates into the nucleus and induces G0/G1 certain chemokine receptors, thereby orchestrating T
arrest, regulating cell division [269, 1631, 2287]. cell recruitment [1613]. Further, IL-16 appears to
CD4 appears to be the primary receptor for IL-16. prime CD4+ cells for IL-2 and IL-15-induced growth
Cells that respond to IL-16 invariably express CD4. [357, 1532].
Anti-CD4 Fab fragments inhibit signaling by IL-16 IL-16 has several roles during antigen presentation.
[270, 358, 359, 360, 361, 1618]. Transfection of CD4 In monocytes and macrophages, IL-16 induced migra-
cDNA into CD4−cells enabled the cells to respond to tion and the secretion of several cytokines including
IL-16 [358, 361]. The absence of CD4 in a mutant clone IL-1β, IL-6, IL-15 and TNF-α [1240]. In DC’s and mac-
eliminated IL-16 response [1191]. IL-16 binds cell sur- rophages, IL-16 stimulated levels of CD25 (IL-2R) and
face CD4 and induces signaling [1021, 1700]. A peptide the costimulatory molecules CD80 and CD83 [739].
of IL-16, based on the proposed CD4-binding sequence, Further, IL-16 may act together with thrombopoietin
blocked IL-16-induced chemotaxis in cultured spleno- during DC development to induce tolerogenic dendritic
cytes and decreased airway hyperresponsiveness in cells capable of inducing anergy in T cells [411]. And in
peptide-treated mice [385]. HIV-1 (gp120) binds CD4 lymphnodes, resident B cells and DC’s produce IL-16
Walter M. Lewko and Robert K. Oldham 189

involved in the trafficking of Th cells and inward lupus erythematosus, asthma, inflammatory bowel
migration of additional dendritic cells [921, 922]. disease and Crohn’s disease [1076, 362, 750, 942, 1236,
Delayed type hypersensitivity is mediated by antigen 1049]. In related animal models, blocking antibodies to
specific Th cells and involves cell movement. The role IL-16 ameliorated these diseases. On the other hand,
of IL-16 in DTH was studied in mice [2255]. IL-16 was IL-16 appeared to regulate inflammatory cytokines in a
expressed in DTH tissues but not in controls. Extracts rheumatoid synovitis mouse model [986]. The synovial
of DTH tissue exhibited chemoattractant activity and infiltrate contained activated CD4+ T cells secreting the
IL-16 neutralizing antibodies inhibited this activity. proinflammatory cytokines IL-1β, IFN-γ, and TNF-α.
When mice were pre-treated with antibodies, swelling, CD8+ T cells produced a factor, which lowered the
leukocyte infiltration and chemokine levels associated inflammatory cytokines. Anti IL-16 antibodies blocked
with DTH were depressed. These results suggested the effects of this factor while treatment with rIL-16
IL-16 had an important role in the recruitment of leuko- mimicked the factor. It appeared that CD8+ T cells
cytes and secretion of cytokines associated with DTH might have anti-inflammatory effects, which are at least
reactions [2255]. in part mediated by IL-16 [986].
While IL-16 stimulates immunity it also has regula- IL-16 has not been extensively studied in cancer.
tory effects. CD4 is part of the TCR/CD3 complex Nonetheless, its involvement in the activation and
responsible for antigen-induced activation of T cells. migration of several cell types including CD4+ T cells,
HIV-1 gp120 and antibodies that bind CD4 tend to inter- dendritic cells and macrophages suggests it may have a
fere with activation [1476]. IL-16 also has immunosup- role in antigen presentation, the development of tumor
pressive effects. It inhibited mixed lymphocyte reactions, infiltrating lymphocytes and anticancer vaccination. Its
deactivated T cells and lowered IL-2 secretion [1686]. antiviral effects may block HIV-related cancers.
Other studies have shown that skin cells transfected
with IL-16 were immunosuppressive suggesting that
IL-16 might be used to prevent graft rejection [572].
IL-16 induces migration and it also appears to regu-
Interleukin-17A
late migration. When IL-16 bound CD4, there was a Proinflammatory, Hematopoietic,
selective loss of chemokine receptor 5 activity. The two
Neutrophil Development
receptors were mutually inactivating. When MIP-1β
bound CCR-5, T cell migration induced by IL-16 was The IL-17 cytokine family has six members. IL-17A,
inhibited. HIV-1 gp120 also appears to inhibit chemokine the first discovered, is a 155 amino acid, 20 kDa glyco-
receptor signaling [1235]. protein [555, 2232]. IL-17 family members have little or
IL-16 is one of several factors secreted by CD8+ T no homology with cytokines outside the family [555,
cells, which inhibit viruses. (Others antiviral factors 2232]. The gene is on chromosome 2q31. IL-17A was
include MIP-1α, MIP-1β and RANTES). Specifically, originally referred to as CTL-associated antigen-8
IL-16 interferes with HIV-1 replication in T cells [43, (CTLA-8) [1688]. Herpesvirus saimiri encodes a similar
103, 1191, 1734, 2294, 2295], macrophages and den- protein [2231]. IL-17 is produced by CD4 T cells, CD8
dritic cells [2028]. The mechanism of inhibition appears T cells and γδ T cells [1, 951, 555, 1144, 2232]. Bacterial
to involve the interaction of IL-16 with CD4. A factor is endotoxin, IL1β, IL-6, IL-18, IL-23 and TGF-β stimu-
produced which binds the core enhancer DNA, inhibits late IL-17A production [852, 1325, 712].
HIV-1 promoter activity and as a result blocks viral IL-17A is produced primarily by CD4 T-cells that are
replication [1191]. IL-16 also blocks HIV-1 uptake by now referred to as Th17 cells [718]. These cells along
the cells [2028]. Decreased virus entry may be due to with Th1, Th2 and Treg cells are major controllers of
IL-16-induced CD-4 downregulation [739]. In a clinical immune response. It is now believed that Th17 cells are
study, HIV-1 infected patients were followed over an 8 responsible for many autoimmune diseases that were
year period. During the asymptomatic phase, serum originally classified as Th1 diseases. The discovery of
IL-16 levels were maintained or increased. With disease Th17 cells solved a problem in the observation that mice
progression, there was a drop in IL-16 [43]. These lacking the Th1 cytokine IFNγ suffered more, not less,
results support a natural role and potential therapeutic autoimmunity than controls. The development of Th17
benefit of IL-16 in the control of HIV infection. lineage appears to have two phases. In the first phase,
IL-16 is involved in the development of several Th0 cells differentiate into Th17 cells in response to
inflammatory diseases. IL-16 levels were elevated in TGFβ and IL-6 (or IL-1β; a proinflammatory signal is
fluid and tissue samples from patients with systemic needed) and then there is an expansion/survival phase,
190 Cytokines

which is stimulated by IL-23. Th17 differentiation is dendritic cells [57]. This may have a role in cancer
inhibited by IL-2 and IFNγ (type 1 cytokines), IL-4 and vaccination. It has also been shown that Chinese hamster
IL-13 (type 2 cytokines), and IL-35 (Treg cytokine) ovary cells engineered to produce IL-17 were less inva-
[149, 718, 985, 1025, 1218, 1451, 1536, 2070]. sive and metastatic [764]. There is also evidence that
Receptors for IL-17 have been found in most tissues IL-17-transfected tumor cell vaccines can stimulate the
tested. There are at least five members of the IL-17 receptor generation of tumor specific cytotoxic T cells [133]. In a
family. These receptors have little or no homology to study on B16 melanoma in mice, tumor antigen specific
other known cytokine receptors [2231, 2233, 1370]. CD4 Th cells that had been polarized in culture, were
IL-17 functions in host defenses against infection. adoptively transferred into mice with growing tumors;
Bacterial products induce IL-17 secretion [852]. IL-17 interestingly, the Th17 cells had superior antitumor activ-
is required for defence against several types of infec- ity compared to Th1 cells, the type traditionally associ-
tion, for example, bacterial pneumonia [2236]. IL-17 is ated with anticancer activity [1395]. However, there are
involved in hematopoiesis. It appears to promote recovery reports that by its proinflammatory properties, IL-17
in response to radiation damage [1961]. IL-17 is may stimulate development of cancer. It has been shown
involved in inflammation. It stimulates production of that IL-17 increased the growth of cervical tumors trans-
molecules such as proinflammatory cytokines, chemok- planted in nude mice [1967]. In prostate, normal tissues
ines, CSFs, prostaglandin, MMPs, NOS and antimicro- produced little IL-17, but most benign prostate hyperpla-
bial peptides in a variety of cells including macrophages, sia specimens and prostate cancers showed high IL-17
fibroblasts, epithelial cells, keratinocytes, endothelial expression, mainly in T-cells but also in epithelial cells
cells, mesothelial cells and dendritic cells [231, 513, [1887]. In lung cancer cell lines, IL-17 had no direct
555, 1768, 878, 890, 1980, 2178]. IL-17 has particularly effect on tumor cell growth but had a remarkable stimu-
remarkable effects on neutrophil production and tissue latory effect on secretion of several angiogenic CXC
infiltration. IL-17-treated fibroblasts release CSFs that chemokines. In SCID mice, lung cancer cells engineered
stimulate neutrophil differentiation [555]. In transgenic to produce IL-17 grew larger and were more vascular
mice that produced excess IL-17, there was enhanced than controls suggesting a role for IL-17-induced
hematopoiesis in general and granulopoiesis in particu- chemokines and angiogenesis in tumor growth [1467].
lar. Peripheral WBC counts increased fivefold and neu-
trophils increased tenfold [1766]. In gastric mucosa,
infection by H. pylori stimulated production of IL-17.
IL-8 levels were increased and the tissue became infil- Interleukin-17B
trated. Antibodies to IL-17 inhibited IL-8 secretion and
antibodies to IL-8 blocked PMN leukocyte migration
Proinflammatory Cytokine
[1174]. Similar responses occurred in inflamed intesti- IL-17B has 27% amino acid identity with IL-17A. The
nal and bronchial epithelial cells [93, 1046]. gene is on chromosome 5q32–34 [1113]. IL-17B mRNA
Dysregulation of IL-17 appears to have a role in the is produced by cells in the pancreas, small intestine, and
pathology of allergic and autoimmune inflammatory dis- stomach but not by activated T cells.
eases [18, 275, 276, 279, 1011, 1174]. In mice with col- IL-17B does not bind the IL-17 receptor. Rather it spe-
lagen-induced arthritis, IL-1 and IL-17 levels were cifically binds the protein IL-17Rh1 (IL-17 receptor
elevated; blocking antibodies for IL-17 alleviated joint homolog 1). At least one additional IL-17 family mem-
destruction whereas IL-1 blockade had no effect, sug- ber, IL-25, also binds IL-17Rh1 [1073, 1807]. The mRNA
gesting IL-17 was the more direct cause of arthritic dam- for this receptor has been detected in pancreas, kidney,
age [1169, 1414]. In patients with arthritis, synovial fluid thyroid, liver, brain and intestines. Receptor levels in
IL-17 stimulated formation of osteoclasts, which caused intestine were increased during inflammation [1807].
tissue destruction [1011]. IL-17 also increased the pro- In a monocyte cell line, IL-17B stimulated produc-
duction of gelatinase in human macrophages [879], tion of IL-1β and TNF-α while IL-17 had little effect. In
aggrecanase and metalloproteinase in chondrocytes activated fibroblasts, IL-17 stimulated production of
[230] and nitric oxide in osteoarthritic cartilage [88]. IL-6 while IL-17B had no effect [1113]. These results
IL-17 has potential in cancer therapy. IL-17 stimulates showed that though the two cytokines are structurally
bone marrow stromal cells to produce G-CSF, SCF and related, they have distinct activities. In mice, IP injec-
IL-8 for the production and mobilization of hematopoi- tion of rIL-17B induced influx of neutrophils. This was
etic stem cells [1767]. This may be useful for stem probably not a direct effect but due to stimulated release
cell transplantation. IL-17 promotes the maturation of of chemotactic factors [1807].
Walter M. Lewko and Robert K. Oldham 191

Interleukin-17C mast cells produce IL-17F. It was not expressed by CD8


T cells or B cells [1881]. IL-17F is a homodimer; het-
Proinflammatory Cytokine erodimers with IL17A chain occur in mouse Th17 cells.
Potency was intermediate of the two homodimeric
IL-17C has approximately 27% homology with IL-17. forms in a chemokine release assay [1122].
The distribution of IL-17C mRNA appears to be rather The receptor for IL-17F is the protein IL-17RC. This
restricted. It was found in prostate and fetal kidney. receptor is related to IL-17R and IL-17A also binds it
Activated T cells did not produce it. The gene for [1030].
IL-17C is on chromosome 16q24. The receptor used IL-17F has a remarkable effect on vascular endothe-
by IL-17C is not known; it does not appear to bind the lial cells. It stimulated the production of endothelial
IL-17 receptor [1113]. IL-2, TGF-β and monocyte chemoattractant protein-1.
In a monocyte cell line, IL-17C stimulated produc- In an in vitro assay for angiogenesis, IL-17F inhibited
tion of IL-1β and TNF-α while IL-17 had no effect. In the formation of tubular, vessel-like structures [1881].
activated fibroblasts, IL-17 stimulated IL-6 secretion IL-17F may be acting directly or through the induction
while IL-17C had no effect. These results showed that of cytokines such as TGF-β, which inhibit angiogenesis
while IL-17 and IL-17C are related, they have distinct [1219, 1881]. IL-17F also stimulates the production of
activities [1113]. Expression of IL-17C in mouse lung IL-6, IL-8 and GM-CSF in bronchial epithelium and it
by adenovirus-IL-17C infection promoted neutrophilia regulates cartilage matrix turnover [833, 929].
and inflammatory gene expression such as IL-6 and IL-17F is involved in asthma. In mice, expression of
IFNγ [825]. IL-17F in lung by adenovirus-IL-17F infection promoted
neutrophilia and the expression of inflammatory genes
such as IL-6 and IFNγ [825]. In humans, expression of
Interleukin-17D IL-17F (but not IL-17) was increased in lung biopsies of
asthma patients after allergen challenge. When added to
Proinflammatory, Inhibitor cultured bronchial epithelial cells, IL-17F stimulated the
of Hematopoiesis production of IL-6 and IL-8 and cell surface ICAM-1; it
recruits neutrophils into bronchial airways [930].
IL-17D has 202 amino acids and as such it is the largest
Polymorphism in the IL-17F gene appears to influence
member of the IL-17 family. IL-17D is most homolo-
susceptibility to asthma. Japanese subjects with arg sub-
gous to IL-17B with 27% identity. The gene is on chro-
stituted at position 161 were protected from the disease;
mosome 13p11. IL-17D mRNA is expressed in a number
this variant blocked the induction of IL-8 by wild type
of tissues including skeletal muscle, brain, adipose,
IL-17F [931].
heart, lung and pancreas. It was poorly expressed on
There are no reports on cancer for this relatively new
activated CD4 T cells, CD8 T cells, monocytes and
cytokine, though IL-17F is of interest for its proinflam-
activated B cells [1880].
matory and antiangiogenic effects.
In cultured endothelial cells, Il-17D stimulated pro-
duction of IL-6, IL-8 and GM-CSF. IL-17D had a sup-
pressive effect of myeloid colony formation in agar
gel assays [1880]. Interleukin-18
Interferon γ, NK and Th1 Response,
Regulation of Inflammation
Interleukin-17E: see IL-18 was discovered in mice with severe hepatitis
Interleukin-25 induced by Propiobacterium acnes and the endotoxin
LPS [1417]. IL-18 was originally called interferon-γ
Interleukin-17F inducing factor (IGIF) for its remarkable stimulation
Proinflammatory, Asthma, Inhibitor IFNγ secretion. It is synthesized as a 192 amino acid pro-
protein, which is processed to the 156 amino acid active
of Angiogenesis form [2045]. IL-18 is similar to IL-1β in that its nascent
Interleukin-17F is also referred to as ML-1 [930]. It has protein lacks the signal peptide usually found on secreted
high homology with IL-17. The gene is on chromosome proteins [120, 1489] and proteolytic activation of the
6p. Activated CD4 T cells, monocytes, basophils and proprotein is carried out by caspase-1 (ICE) [688].
192 Cytokines

Activation is necessary for secretion; caspase-1-deficient 1300, 1652, 1894, 2253], B cells [2253], NK cells [1954,
macrophages do not secrete IL-18 as usual when treated 2009], macrophages [1393, 1741] and in DCs [573].
with LPS [512, 618, 688, 1779]. IL-18 stimulates activated Th1 cells to grow and
IL-18 is produced by macrophages, dendritic cells produce IFN-γ [1489]. It is well known that Th1 cell
[1489, 1600] and Kupffer cells [1779, 2045]; levels are differentiation requires IL-12. Il-18 acts synergistically
increased by microbial infection or LPS treatment. IL-18 with IL-12 [1300]. IL-12 initiates differentiation; IL-18
is also produced by adrenal cortex (in the area of cortisol intensifies the response. IL-2 is a costimulator of Th1
production) [340], by skin keratinocytes [1895] and by differentiation and growth [998]. IL-18 and IL-12 both
airway epithelium [234]. In adrenal cortex, expression increase IL-2 receptor levels. IL-18 stimulates Th1
was increased by cold stress as a possible link between cells but it did not appear to influence Th2 cell growth
the environment and immune response [340]. Treating or production of IL-4 and IL-10 [998]. With Th2 dif-
mice with contact allergens stimulated IL-18 expression ferentiation, IL-18 receptor expression is lost and with
in keratinocytes [1895]. In airway epithelial cells, IL-18 it capacity to respond to IL-18 [2214].
levels were related to Th1 status [234]. Although IL-18 and IL-12 have related activities,
IL-18BP is a binding protein found in circulation, there are differences. IL-12 initiates differentiation of
which has relatively high affinity for this cytokine. As a Th1 cells; IL-18 does not initiate but enhances Th1 dif-
feedback loop, IL-18 induces IFNγ and interferon ferentiation [1487, 1652]. IL-18 induced Th1 cells to
induces IL-18BP. It competes with the receptor for free produce IL-2; IL-12 did not [998]. In mouse activated
IL-18 and in this way regulates IL-18 activity [963, PBLs, IL-18 stimulated GM-CSF production and
1465, 1640]. A relative lack of IL-18BP occurs in depressed IL-10; IL-12 did not [2046].
patients with hemophagocytic syndrome, a disease with IL-18 is generally considered a proTh1 cytokine, but
elevated free IL-18 and characterized by a strong Th1 IL-18 does stimulate Th2-related responses [995, 1033,
response [1260]. 1199, 1684, 2162]. B cells contain receptors for IL-18
The IL-18R contains two subunits [180, 965, 2014]. and IL-12. These two cytokines synergize in the produc-
The α subunit binds IL-18 and the β subunit performs tion of immunoglobulin and IFN-γ [13]. IL-18 also
signal transduction. Although the β subunit does not stimulates IL-4 secretion in NKT cells [1085, 2251,
bind IL-18, as part of the receptor complex it enhances 2254] and IL-13 secretion by NK cells [797].
affinity for IL-18 [965]. Interestingly, the IL-18Rα sub- Overexpression of IL-18 by B cells and T cells in trans-
unit was originally discovered and named IL-1 receptor genic mice induced high levels of both Th1 and Th2
related protein (IL-1Rrp) for its molecular similarity to cytokines [796].
the IL-1 receptor, though this protein did not actually IL-18 also acts synergistically with interferon-α/β in
bind IL-1 [439, 1488, 1538]. At the time, it was referred the secretion of IFN-γ by T cells. Virus infection induced
to as an orphan receptor for lack of a known ligand; later macrophages to produce IFN-α/β and IL-18 (but not
it was determined to be the receptor for IL-18 [795, IL-12). Conditioned media from these macrophages
1986, 2014]. The IL-18Rβ subunit is also a member of induced IFN-γ in T-cells. Neutralizing antibody to IFN-
the IL-1R family. It is related to the IL-1R accessory α/β blocked secretion of IFN-γ; antibody to IL-12 had
protein. When IL-18 binds, it activates a pattern of cell no effect. These studies showed that the type I interfer-
signaling which is similar to that of IL-1 [392, 1000]. ons acted synergistically with IL-18 in the production of
In knockout mice genetically defective for the IL-18 immune interferon [1725]. IFN-α increased the levels
receptor, development of Th1 cells was impaired. NK of an adapter molecule (MyD88), which facilitated
cells had decreased cytotoxicity and IFN-γ production IL-18 receptor signaling [1724].
[796]. Overexpression of IL-18 in transgenic mice IL-18 has remarkable effects on NK cells. IL-12
resulted in high levels of IgE, IgG1, IL-4, IL-5, IL-13 and and IL-2 are also well known stimulators of NK cells.
IFN-γ, reflecting both Th1 and Th2 cell activation [796]. IL-18 in combination with IL-12 or IL-2 induced
IL-18 and IL-12 have similar effects on several types much higher NK cell proliferation, IFN-γ secretion
of cells. These effects are often synergistic. In certain and antitumor activity than IL-12 or IL-2 induced
cases, IL-12 appeared to be required for IL-18 response. when added alone or together [2009]. These results
This seems to be due to the ability of IL-12 to increase suggested an obligatory role for IL-18 in NK prolif-
IL-18 receptor levels. IL-12 upregulates both α and β eration and function [2009].
subunits. Increased IL-18 receptor levels enhanced the Cell killing by Fas-mediated apoptosis is stimulated
capacity for response [521, 963, 1724, 2214, 2253]. IL-18 by IL-18. IL-18 upregulates Fas ligand (discussed
and IL-12 synergistically induce IFN-γ in T cells [998, below) in NK cells [2035] and in helper T-cells [381].
Walter M. Lewko and Robert K. Oldham 193

In NK cells, IL-18 acts together with IL-2 and IL-12. It has been suggested that IL-18 may be involved in
The IL-18 treated NK cells were positive for perforin cytotoxic T cell development by acting as a bridge
but killed target cells exclusively by FAS engagement between the innate and adoptive immune systems
[2035]. By its ability to stimulate interferon-γ secretion, [1962]. IL-18 stimulates IFN-γ levels and activates NK
IL-18 also stimulates Fas levels in many target cells. cells (Fas L) causing tumor cell apoptosis. This provides
IL-18 has a role in the induction and regulation of apoptotic bodies for dendritic cell antigen presentation
inflammation. Neutrophils respond to IL-18 with and the induction of specific CTLs [1962].
increased production of cytokines, CD11b (comple- IL-18 is being tested in cancer patients. In a phase I
ment receptor) and granule release. In a mouse model, trial, IL-18 was administered to 28 patients, mainly
IL-18 neutralizing antibodies suppressed footpad renal cancer and melanoma. Increases in serum IFNγ,
inflammation caused by carragean injection [1102]. GM-CSF and other indications of cell activation were
PBLs treated with IL-18 produce pro-inflammatory observed. There were two partial responses; further
TNF-α and in turn IL-1β and the chemokines IL-8 and studies are warranted [1651].
MIP-1α [1601]. IL-18 also brings about regulation of It should be mentioned that while IL-18 has antican-
inflammation. In T cells and NK cells, IL-18 together cer activity, certain cancers may be able to exploit the
with IL-2 stimulated the production of IL-13, an inhibitor IL-18 system. For example, IL-18 binding protein is
of inflammation [797]. naturally secreted in response to IFNγ and it downregu-
IL-18 appears to be involved in rheumatoid arthritis lates IL-18 activity. Many tumors secrete IL-18 BP. In
[652, 1103]. Elevated levels of IL-18 are found in this way the anticancer effect of IL-18 may be inhibited
arthritic synovial fluid and serum [652]. In a collagen- by the tumor [1545]. In certain cancers, it has been
induced arthritis model, mice lacking IL-18 had a greatly reported that IL-18 stimulates growth, angiogenesis,
reduced incidence and severity of arthritis compared adhesion to endothelium and invasion [246, 962, 1535,
with control mice. T cells from IL-18 deficient mice had 2075]. In these tumors IL-18 may contribute to progres-
a reduced capacity for antigen-induced cell division and sion. Further, IL-18 may have a role in immune escape.
secretion of proinflammatory cytokines [2131]. IL-18 IL-18 upregulates Fas ligand in B16F10 melanoma
has also been implicated in other inflammatory diseases cells. It is possible that tumor cell-Fas ligand may induce
including Crohn’s disease of the bowel [1577] and in apoptotic death of effector cells resulting in immune
sarcoidosis [662]. suppression [311].
IL-18 appears to have a role as both a mediator and
regulator of angiogenesis. In culture, IL-18 stimulated
vascular endothelial cells to migrate and form capil-
lary-like structures. In animals, implants containing Interleukin-19
small amounts of IL-18 stimulated the growth of new
vessels [1535]. As a regulator, it has been shown that
Inflammation
IL-18 suppressed angiogenesis and it did this by IL-19 was discovered in LPS-stimulated monocytes
blocking bFGF-stimulated cell division in endothelial [582]. It is a member of the IL-10 cytokine family
cells [240]. (IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, IL-28, IL-29).
IL-18 has anticancer activity in many tumor models. The gene for IL-19 is located together with IL-10 on
Elevated IFNγ and NK cells are generally required, but chromosome 1q31–32. A homodimer appears to be the
depending on the system CD4+ or CD8+ T cells may be active form [582]. LPS and GM-CSF stimulate mono-
involved [1301, 1302, 1509]. Combinations of IL-2 plus cytes to produce IL-19. In LPS activated monocytes,
IL-18 [1854, 2156] and IL-12 plus IL-18 [1117, 1509, IL-4 and IL-13 each enhanced [584] and IFNγ depressed
1624] generally have synergistic effects. These studies IL-19 secretion [583]. Il-19 is also produced by kerati-
have been extended to an IL-2/IL-18 fusion protein [4], nocytes of psoriasis lesions [1660].
which produce NK and T cell responses with lower toxicity. IL-19 binds and signals through the IL-20 receptor
Additionally, IL-18 is being tested, with some success, in (IL-20Rα/IL20Rβ). IL-19 does not bind the IL-10
cancer vaccines [840, 1228, 1852, 1969, 2210, 2224]. receptor. IL-20R is expressed in skin, lungs, reproduc-
Further, it has been reported that NKT (generally consid- tive organs and various glands. Receptor levels are in
ered to be a regulatory and immunosuppressive cell type) inflamed tissues [462, 583, 1540].
may have anticancer activity. NKT cells isolated and Monocytes treated with IL-19 secrete IL-6, TNF-α,
cultured with IL-12 plus IL-18 produced activated cells and reactive oxygen species [1125]. In T cells, IL-19
that inhibited tumors when transferred to mice [118]. appears to favor Th2 differentiation and IL-4 secretion.
194 Cytokines

[583]. In PBMCs IL-19 induces IL-10 secretion and its IL-8 are released. However, in studies of lung cancer,
own production. IL-19 production is in turn down- IL-20 inhibited COX-2 and prostaglandin E2 produc-
regulated by IL-10 [902]. tion in epithelial and endothelial cells, and it inhibited
IL-19 appears to be involved in asthma [1124] and PMA-induced angiogenesis [805, 746].
psoriasis [1114, 1660]. Asthmatic patients had twice the
serum levels of IL-19 compared with controls; levels of
IL-4 and IL-13 were also elevated. In culture, activated
T cells treated with IL-19 secreted Th2 cytokines IL4,
Interleukin-21
IL-5, IL-10 and IL-13 [1124]. NK, T and B Cell Development,
Immunomodulation
IL-21 is a pleiotropic cytokine, which is structurally
Interleukin-20 related to IL-2, IL-4, and IL-15 [78, 1520, 1540]. IL-21
Inflammation, Skin Development is produced mainly by Th2, Th17 and follicular T cells
[316]. The receptor for IL-21 has two subunits, IL-21Rα
and Immunity and IL-2Rγ. IL-21Rα is related to IL-2Rβ and IL-4Rα;
IL-20 is a member of the IL-10 family. The gene for IL-2Rγ is the subunit common to several IL-2 family
IL-20 is located on chromosome 1q31–32 together with receptors. The IL-21 receptor is expressed on T, B, and
the genes for IL-10, IL-19 and IL-24 [582]. IL-20 is pro- NK cells [78, 699, 1520].
duced by keratinocytes [1641], monocytes [2187] and T cells are stimulated and regulated by IL-21. Typical
glial cells [799]. Th2 responses (e.g. to schistosomes, airway inflamma-
IL-20 binds two receptors (IL-20Rα/IL20Rβ) and tion) depend on IL-21 [570, 1561]. IL-21 also supports
(IL-22Rα/IL20Rβ) [169, 1540]. IL-20R levels are ele- the clonal expansion of virus and tumor antigen spe-
vated in psoriatic skin [169]. IL-20 signals by STAT3 cific effector CD8+ T cells [2059, 1120, 1121]. IL-21
activation [462, 463]. sustains the expression of CD28, an important costimu-
IL-20 appears to regulate keratinocyte involvement latory receptor [34, 1134]. It increases or decreases
in skin inflammation [1641]. Overexpression of IL-20 IFN-γ production depending on conditions [1903, 1932,
in transgenic mice resulted in aberrant differentiation 2208]. But IL-21 also regulates CD8+ T cell expansion
of epidermis, keratinocyte proliferation and abnormal when required by inducing apoptosis [113]. In mice,
skin resembling psoriasis. These mice also lack adipose plasmid DNA vaccines expressing HIV-1 env glycopro-
tissue and their T lymphocytes show high apoptosis; tein showed increased T cell response when combined
they die soon after birth [169]. IL-20 stimulates the with IL-21 [177]. At least in part, increased generation
growth of CD34+ multipotential progenitors in a unique of CD8+ cytotoxic T cells may be the due to a suppres-
way, without imparting any tendency towards a partic- sive effect IL-21 has on Foxp3 regulatory T cells (Li
ular developmental pathway, erythroid, granulocyte- 08) or the response of Th cells to Tregs [1550].
macrophage or megakaryocyte [1139]. IL-21 is involved in NK cell expansion, function and
In humans, IL-20 appears to be involved in psoriasis regulation. In human bone marrow cultures, IL-21 stim-
and skin inflammation [2128]. In lesions from patients, ulated NK cell proliferation and maturation [1539]. In
IL-20 and its receptor were elevated. IL-20-treated T cultured mouse cells, IL-21 increased NK function even
cells produced increased levels of keratinocyte growth as it depressed growth [919]. IL-21 appears to be able to
factor [2128]. modulate cytotoxicity towards certain target cells. In
Glial cells can produce IL-20 and LPS stimulates human NK, IL-21 depressed the NKG2D receptors
release. This suggests that IL-20 may have a role in while enhancing others [227]. When NK cells were
inflammation of the brain [799]. In arthritis patients, syn- treated with IL-21, human breast cancer cells coated
ovial fluid contains elevated IL-20; synovial fibroblasts with Trastuzumab were more effectively killed by
treated with IL-20 secreted IL-6 and IL-8. In rat model, ADCC [1655].
IL-20 blockade decreased severity of arthritis. It appears IL-21 influences development of B cells and Ig pro-
that IL-20 is involved in joint inflammation [809]. duction. IL-21 induces differentiation of naive and
There is evidence for IL-20 being pro-angiogenic and memory B cells into antibody-secreting plasma cells
anti-angiogenic. Endothelial cells contain IL-20 recep- [501, 1521]. Depending on the conditions, IL-21 may
tors. When treated with IL-20 these cells proliferate, increase or decrease growth or induce apoptosis [1540,
migrate and form tubular structures; bFGF, VEGF and 1285]. IL-21 regulates Ig class switching. It is a switch
Walter M. Lewko and Robert K. Oldham 195

factor for IgG1 and IgG3; IL-21 depresses IgE [1522, There is a soluble IL-22 binding protein, which antago-
1551, 1931, 2196]. nizes IL-22 activity. The IL-22BP has homology with
IL-21 inhibits dendritic cell activation and maturation IL-22R, but it lacks the transmembrane cell-binding region.
[192, 1902]. It is part of a negative feedback loop In effect, the IL-22BP is a decoy receptor [463, 1013].
between dendritic cells and T cells in which activated IL-22 levels rise in LPS-treated animals. IL-22 appears
dendritic cells stimulate T cells, inducing them to pro- to function during inflammation and immune response.
duce cytokines including IL-21 which in turn inhibits IL-22 mainly targets non-circulating cells involved in
dendritic cells [1902]. innate immunity including epithelium, keratinocytes,
IL-21 is involved in the Th system and regulation and hepatocytes. In keratinocytes, IL-22 induces hyper-
of inflammation. IL-21 is produced by Th17 cells and plasia, migration, antimicrobial proteins and production
in an autocrine manner IL-21 sustains expression of of proinflammatory molecules [178, 2191]. In liver cells,
the Th17 transcription factor RORγT and secretion of IL-22 increases the production of acute phase proteins
IL-17 [2129]. [466]; it also induces LPS binding protein, which neu-
IL-21 has been studied in a number of different exper- tralizes this proinflammatory and Th17-inducing micro-
imental tumors. It has been shown to have anticancer bial product [2189]. IL-22 also has a mild inhibitory
activity related to T cells [338, 427, 577, 1361, 1420], B effect on IL-4 production by Th2 cells [2212].
cells [1835], NK cells [338, 577, 1420, 1849, 1951, IL-22 levels are elevated and it appears to be involved
2101] and NKT cells [1849]. Several of these studies in certain inflammatory diseases including rheumatoid
used IL-21 as an adjuvant in vaccines. Addition of IL-15 arthritis [847], psoriasis [1186, 2191, 2291], inflam-
to IL-21 improved the response [177, 1420, 2275]. matory bowel disease and Crohn’s disease [49]. IL-22
Regulatory T cells may limit vaccine immunogenicity. In does stimulate inflammatory molecules, and may cause
one mouse study, use of IL-21 secreting tumor cells pathology but it also appears to be involved in resolu-
together with anti-CD25 antibodies to block regulatory T tion of inflammation and tissue regeneration. In mice,
cells increased cure rate from 17% to 70% [338]. IL-22 has been shown to ameliorate experimental
IL-21 has direct effects on the growth of some hema- autoimmune myocarditis [287] and ulcerative colitis
tologic malignancies. In B cell chronic lymphocytic leu- [1922]. In pancreatic acinar cells, IL-22 induces pan-
kemia, IL-21 is able to induce apoptosis in CpG-treated creatitis-associated protein [11]. IL-22 has a protective
[883] or CD40-stimulated cells [387]. On the other effect during liver injury. IL-22 induced antiapoptotic
hand, IL-21 is a growth factor for certain adult T-cell factors in hepatocytes and stimulated growth suggest-
leukemia [2042] and melanoma cells [198]. ing a role for IL-22 in liver cell survival and tissue
IL-21 is in clinical studies. Melanoma and renal cell repair [1611].
carcinoma patients, treated with IL-21 in phase I, tolerated With regard to cancer, there is a report that IL-22
the cytokine reasonably well and showed evidence of anti- inhibited the growth of EMT6 breast cancer in treated
cancer activity [383, 1187]. IL-21 is also being used to mice by cell cycle arrest [2125].
develop more effective CD8+ T cells for adoptive immu-
notherapy [760]
Interleukin-23
Interleukin-22 T cell Stimulation, Inflammation, Regulator
of Adaptive and Innate Immunity
Inflammation IL-23 is composed of two subunits. The p19 subunit is
IL-22 is a member of the IL-10 cytokine family. It was related to p35 of IL-12. And p40 is identical to p40 of
originally referred to as IL-10-related T cell-derived IL-12. Activated dendritic cells, macrophages and kerati-
inducible factor (IL-TIF). Activated Th17, Th1 and Th2 nocytes produce IL-23 [1502, 1533, 1575]. The IL-23
cells produce IL-22 [464, 465, 1123, 1186, 2188, 2212]. receptor is composed of IL-23 Rα and IL-12Rβ1. IL-23
IL-22 is also expressed by IL-9 stimulated mast cells activates Jak-STAT signaling characteristic of the IL-12Rβ1
[464] and by activated NK cells [2190] subunit [1573]. The IL-23 receptor is expressed on DC,
The IL-22 receptor consists of two chains, IL22Rα macrophages and T cells [1533].
and IL-10Rβ. Each chain separated binds IL-22 but both The effects of IL-23 are similar to but distinct form
chains, associated, are necessary for activity. Signaling IL-12. In mouse cells, IL-23 induces proliferation of mem-
is through STAT 1, 3 and 5 [1012, 2212]. ory T cells (CD4+ CD45RB low). IL-12 does not [1502].
196 Cytokines

IL-23 does not stimulate IFN in mouse T cells. In human T called mda-7 (melanoma differentiation antigen-7). Its
cells, IL-23, like IL-12, increases IFN-γ production, though counterpart in mice is called FISP [1738, 1790]. In
to a lesser degree than IL-12. IL-23 also stimulates growth human cancer, Mda-7 is a tumor suppressor; expression
in human memory CD45RO T cells [1502]. of the gene in melanoma cells inhibited proliferation
T cells express ICOS (inducible costimulator) protein, and induced apoptosis [889]. Loss of mda-7 expression
which is a CD28 family member. ICOS is important for was associated with disease progression [488]. Later,
enhancement of Th and CD8+ T cell responses. In human when the cytokine nature of mda-7 became clear, it was
T cells, ICOS is increased by IL-23 and IL-12; it is inhib- designated IL-24 [261, 278, 810, 1731]. IL-24 is a mem-
ited by IL-4 [2122]. This may provide a powerful way ber of the IL-10 family [261]. The gene is located in the
for IL-23 and IL-12 to amplify T cell responses. IL-10 cluster on chromosome 1q31–32 [810]. Activated
IL-23 induces inflammation [1502]. IL-23 appears to T cells and macrophages produce IL-24. It is also
be involved in encephalomyelitis, arthritis and inflam- expressed by many tumors [1582, 2187]. Expression is
matory bowel disease [298, 363, 1399, 2242, 2279]. stimulated by IL-1, IL-2 and several other cytokines but
Transgenic mice overexpressing IL-23 p19, had severe not by Th2-related cytokines [1582]. IL-24 signals
multiorgan inflammation, impaired growth, infertility through two receptors IL-20R1/IL-20R2 and IL-22R1/
and early death. Blood contained increased levels of IL-20R2 [2104]. IL-24 is a pro-inflammatory cytokine;
neutrophils, IL-1α, TNF and acute phase proteins [2132]. it stimulates the production of IL-6 and TNF [261].
TGF-β and IL6 induce Th17 differentiation; IL-23 stim- Overexpression of MDA-7 in tumor cells, by adenovi-
ulates and maintains the Th17 cells [10, 1058]. The rea- rus-IL-24 gene transfer, suppresses a variety of cancer
son for heightened inflammation and pathology may be types including breast, cervical, colorectal, glioma, lung,
explained by the lack of regulatory IL-10 in IL-23- mesothelioma, ovarian, pancreas, prostate and sarcoma
differentiated T cells. When induced with TGF-β and [242, 277, 278, 810, 1071, 1201, 1712, 1731, 1914, 1915,
IL6 only, T cells secrete IL-10 in addition to IL-17 and 1916, 2289]. Susceptible tumors exhibit decreased growth
the other proinflammatory cytokines [1267]. and increased apoptotic death; IL-24 also inhibits angio-
Microbial products induce macrophages and DCs to genesis [295, 1617, 1731]. Radiation resistance may be
secrete IL-23. IL-23 is required for neutrophil produc- reversed by IL-24 treatment [1914, 1915, 2218]. Conversely,
tion. Mice that were p40-deficient lacked neutrophils; IL-24 resistance, as due to overexpression of antiapoptotic
treatment with IL-23 (but not IL-12) restored neutrophil survival factors, may be reversed by irradiation [1914].
levels [1841]. There is much interest in IL-24 for the treatment of
In dendritic cells, IL-23 stimulated IL-12 production, cancer [540, 854, 1070]. IL-24 depresses growth,
as did IL-12 itself. Further, IL-23 treated DC’s promoted induces apoptosis, inhibits angiogenesis and increases
immunogenic antigen presentation of an otherwise tole- radiosensitivity in a variety of tumors, with little toxic-
rogenic peptide, which IL-12 did not do [128]. ity towards normal cells. There is particular interest in
IL-23 has been shown to be an effective adjuvant in the use of mda-7 for gene therapy [854]. Adenovirus-IL24
anticancer vaccines for colon carcinoma [1143, 1795, infected cells which produce IL-24 are inhibited;
2112], melanoma [1143, 1497, 1516], and glioma bystander tumor cells are also affected [1913]. Several
[2267]. Prior depletion of regulatory T cells enhanced experimental adenovirus constructs have been devel-
antitumor response. IL-23 induced IFNγ; CD8+ T cells oped to optimize the process. INGN 241 has success-
mediated antitumor immunity [1497]. There is also fully completed phase I trials [366, 2010].
evidence for non-T cell mediated antitumor immune
effects of IL-23 in human pancreatic and esophageal
tumors in nude mice [1794, 2043].
Some human tumors have elevated IL-23 levels. There
Interleukin-25
is evidence that the proinflammatory nature of IL-23, Proinflammatory; Regulation of Th17
compared with IL-12, may actually be responsible for the
tumor growth and progression in certain cancers [1057]. IL-25 is a member of the IL-17 family. It is also referred to
as IL-17E. Th2 cells, eosinophils, mast cells, macrophages
and microglial cells produce IL-25 [844, 915, 985, 1807,
2109]. IL-25 binds IL-17Rh1, the same receptor used by
Interleukin-24 IL-17B [1073, 1807]. There are high levels of this receptor
IL-24 was discovered in melanoma cells that had been in kidney and moderate levels in other organs. IL-25 stimu-
treated to induce differention, using IFNβ plus mezerein lates production of type 2 cytokines that protect against
(a protein kinase c activator) [889]. IL-24 was originally intestinal helminths [508, 1518, 1519].
Walter M. Lewko and Robert K. Oldham 197

Transgenic mice that overproduce IL-25 had increased cells and skin keratinocytes, IL-26 may have a role in
expression of Th2 cytokines IL-4, IL-5, IL-10 and mucosal and cutaneous immunology [792].
IL-13, G-CSF and the Th1 cytokines IFN-γ and TNF-α
as well. The TG mice had eosinophilia, elevated IgE and
IgG1, and immune cell infiltration in many organs.
These mice also suffered epithelial hyperplasia, hyper-
Interleukin-27
trophy in several organs and jaundice [1526]. Adaptive and Innate Immune Response;
IL25 appears to be involved in several inflammatory
Regulation of Inflammation/Th17
diseases. In mice, overexpression of IL-25 in lung by
adenovirus-IL-25 infection induced airway hyperreac- IL-27 is a member of the IL-12 family. This cytokine has
tivity with Th2-like responses including IL-4, IL-5, two subunits, p28 and EBI3 (Epstein-Barr virus induced
IL-13 and eotaxin production, eosinophil infiltration, gene 3). Activated dendritic cells and monocytes produce
mucus production [825]. In eosinophils, IL-25 IL-27. Microbial and host factors stimulate production.
enhanced survival, increased expression of surface The IL-27 receptor is WSX-1. The major targets of IL-27
adhesion molecules [301] and stimulated production are CD4+ T cells and NK cells [1713]. In naive T cells,
of proinflammatory IL-6, IL-8, MIP-1α, and MCP-1 IL-27 supports proliferation and increases IL-12R levels.
[2195]. Lung tissue from asthmatic patients and atopic IL-27 stimulates IFNγ production in NK cells and in
skin lesions show elevated expression of IL-25 and its activated T cells; in naïve T cells, IL-27 induces T-bet,
receptor [2109]. IL-25 induces catabolic activity in IL-12R and Th1 differentiation [747, 2258]. Il-27 regu-
articular cartilage and may be involved in arthritis lates inflammation. In mice where the IL-27/IL-27R
[230]. It appears that during inflammation, Th2 cells system was defective, infection resulted in severe
and eosinophils/basophils may collaboratively rein- chronic inflammation. IL-17 inflammatory effects were
force one another to cause the disease state [2109]. exaggerated [115, 1663, 1911, 2176]. IL-27 regulates
While IL-25 seems to cause inflammation, IL-25 also inflammation at several levels. It stimulates regulatory
appears to regulate it. For example, IL-25 controls Th17 cells to secrete IL-10 [115]. IL-27 also appears to act
inflammation. IL-25 −/− mice are much more sensitive directly on Th cells to suppress Th17 cell development
than normal mice to the debilitating effects of experi- [1911, 2258]. IL-27 also acts directly on activated gran-
mental autoimmune encephalitis. In the CNS, micro- ulocytes and macrophages to suppress production of
glial cells produce IL-25, which in turn induces IL-13 in reactive oxygen [2176].
Th2 cells. IL-13 downregulates DC activity and lowers IL-27 has anticancer activity. Expression of IL-27 in
the cytokines IL-1β and IL-6, which drive Th17 differ- colon cancer cells has been shown to be immunogenic
entiation. In this way, IL-25 secretion in brain may be [309, 772]. Tumors secreting IL-27 were rejected. Mice
responsible for alleviation of damage associated with developed tumor specific protective immunity. Interferon
IL-17 in experimental encephalitis [985]. γ, CD8+ T cells and NK cells appeared to be involved
[309, 772]. Poorly immunogenic B16F1 melanoma
cells, engineered to secrete IL-27, were also inhibited.
Interleukin-26 This growth inhibition was due to NK cells; added IL-12
was synergistic [1497]. IL-27 has also been found to be
Epithelial Immune Response antiangiogenic [1811].

IL-26 is a member of the IL-10 family. It is also


referred to as AK155 [532, 988]. IL-26 was discov-
ered as an overexpressed protein in cultured T cells Interleukin-28A, Interleukin-28B,
transformed by herpesvirus saimiri [792, 988]. The
gene for IL-26 is located on chromosome 12q15. T Interleukin-29
cells, in particular activated memory T cells, and NK
cells produce IL-26 [2187]. Secreted IL-26 binds hep-
Type III Interferons; Antiviral, Antitumor
arin where it is stored in an inactive form until it is IL-28A (IFN-λ2), IL-28B (IFN-λ3) and IL-29 (IFN-λ1)
released [792]. are three recently discovered cytokines in a new class of
The receptor for IL-26 is IL20R1/IL-10R2 [792, interferons, referred to as the lambda or Type III inter-
1802]. Epithelium is a major target for IL-26. Treatment ferons. They are produced by activated (e.g. viral RNA-
with IL-26 enhances expression of ICAM-1 and secre- treated) dendritic cells, monocytes and macrophages.
tion of IL-8 and IL-10. By targeting colon epithelial IFNα and TNFα enhance the λ interferon response to
198 Cytokines

viruses by increasing expression of toll-like receptors IL-4, IL-5 and IL-13 were elevated. IL-31 signaling
and signaling molecules. The λ interferons are related to appears to limit the type 2 inflammatory process [1558].
the type I interferons and to IL-10.
The receptor used by these cytokines is IL-28Rα-
IL-10Rβ. The λ interferons activate typical IFN-
inducible genes including MHC I and II, oligoadenylate
Interleukin-32
synthetase and MxA. λ interferons inhibit replication of Inflammation
certain viruses in vitro and stimulate antiviral immune
response in treated mice [54, 1241, 1288, 1514]. IL-32 was discovered as the transcript NK4 in activated
IFN-λs are involved in the exquisite cell interac- immune cells. Many types of cells produce IL-33 when
tions involved in immune regulation. Monocytes, exposed to inflammatory conditions. IL-33 has no
macrophages and dendritic cells produce IFN-λs and homology with other known cytokines. IL-32 has six
these cells are influenced by them, IFN-λ secretion isoforms. Activated NK cells, T cells, and IFNγ-treated
increases with LPS-induced dendritic cell differentia- epithelium and monocytes produce IL-32 [438, 900,
tion and maturation. Keratinocytes may be activated; 964]. IL-32 induces inflammation by way of p38 MAP
Th1/Th2 ratios and cytotoxic T cell activities may be kinase and NF-κB signaling [1444].
enhanced [2190]. And it has also been shown that IL-32 induces monocytes to produce TNFα, IL-8,
IFN-λ-treated dendritic cells induce proliferation of PGE2 and other inflammatory factors. It signals
Treg cells, which eventually suppress effector T cell through NFκB and MAP kinase [692]. IL-32 syner-
activity [1292]. gizes with the microbe sensing NOD receptor system
IFN-λs also have anticancer activity. In cultured neu- for the induction of IL-1 and IL-6 by muramyl dipep-
roendocrine carcinoma cells, IL-28A and IL-29 inhib- tide. But IL-32 had no such stimulating effect on the
ited cell proliferation and induced apoptosis (2300). TLR system [1443].
IFN-λ secreting tumor cells implanted in animals stimu- In addition to inflammation, IL-32 also influences
lated NK and CD8 T cells immune response, resulting specific immunity. IL-32 induced differentiation to
in decreased growth and metastasis in melanoma, fibro- macrophages from blood monocytes and, interestingly,
sarcoma and colon tumor models [1065, 1466, 1728]. from dendritic cells [1444]. Apoptotic T cells were high
in IL-32. Forced expression of IL-32 induced apopto-
sis. This suggested that IL-32 is involved in AICD. This
effect of IL-32 on T cells may be by a non-secreted,
Interleukin-31 intracrine mechanism [630]
There is evidence IL-32 is involved in several dis-
Pruritis, Allergic Inflammation eases including rheumatoid arthritis, psoriasis, Crohn’s
IL-31 is a cytokine that is produced by T cells, in disease and chronic obstructive pulmonary disease
particular, Th2 cells. IL-31 receptor is composed of two [438]. In tissue biopsies from patients with rheumatoid
chains, IL-31 Rα and oncostatin M Rβ. The IL-31R is arthritis compared with osteoarthritis, IL-32 was
found in activated monocytes, epithelial cells and in increased and levels correlated with disease severity
sensory neuron bodies associated with skin. [900]. Of 171 genes tested, four genes were overex-
IL-31 appears to be responsible for itch sensation. pressed in RA fibroblasts, and the gene for IL-32 was
Transgenic mice overexpressing IL-31 develop itching most pronounced [229].
skin lesions analogous to human atopic dermatitis. IL-31
is elevated in mouse and human dermatitis lesions, but
not in human psoriatic skin lesions. Pruritis in a mouse
model appeared to be related to IL-31 secretion [431, Interleukin-33
1438, 1859, 1952].
IL-31 is involved in asthma. In lung epithelium, IL-31
Inflammation, Allergy
induced EGF, VEGF and inflammatory chemokines IL-33 is an IL-1 family member. It is produced by
[855] and inhibited cell proliferation [291]. vascular endothelium and certain fibroblasts. IL-31 is
IL-31 also has regulatory functions. Mice that are synthesized as a precursor and cleaved by caspase-1,
IL31R−/−have exaggerated Th2 responses; when injected in a manner similar to IL-1 processing.
with schistosome eggs, these mice developed severe The IL-33 receptor is the IL-1 receptor-related protein
lung inflammation, granulomas and fibrosis. Levels of ST2. It signals through NF-κB and MAP kinases.
Walter M. Lewko and Robert K. Oldham 199

IL-33 enhances expression of Th2 cytokines IL-4, NK cells [1761, 2160]. It is also found on epithelial,
IL-5 and IL-13. In mast cells, IL-33 stimulates matura- endothelial, and neural cells [184, 1622].
tion, survival and the production of IL-8 and Th2 cytok- In T cells, the engagement of 4-1BB induces prolif-
ines. In treated mice, IL-33 induces severe mucosal eration, cytokine secretion, cell survival and the sup-
inflammation [28, 434, 839, 1750]. IL-33 and Th2 pression of AICD. 4-1BB stimulates memory response
cytokines were required for expulsion of intestinal to second antigen challenges [147, 238, 317, 389, 639,
nematodes [821]. In mice, Il-33 has been shown to pro- 827, 1585, 1764, 1948]. 4-1BB is one of several costim-
tect against the development of atherosclerosis [1310]. ulatory signaling systems, which may supplement,
In endothelial cells, IL-33 appears to also have an modify and, to a degree, replace costimulation through
intracellular role, apart from its receptor. It translocates the B7/CD28 system [213, 389, 390, 391, 715, 828,
into the nucleus where it binds chromatin and as a 833]. 4-1BB is stimulatory but it is also regulatory; it
nuclear protein it represses transcription [247]. preferentially induces proliferation of CD8 T cells over
CD4 T cells [1818]. 4-1BB signaling tends to suppress
Th2 responses and humoral immunity [789, 1319]. In
this way, 4-1BB signaling can ameliorate Th2-related
Interleukin-35 allergy and autoimmunity [312, 1927].
In NK and NKT cells, stimulation of 4-1BB induces
Treg Cytokine, Regulation of Th17 activation and the secretion of cytokines. Mice lacking
IL-35 is a heterodimeric cytokine that is composed of 4-1BB have depressed NK numbers and activity [1289,
IL-12α and IL-27β chains. nTreg cells (CD4+ CD25+ 2078]. NK cells are well known as cytotoxic effector
Foxp3+) produce IL-35. The secretion of this cytokine cells but they also appear to help CTL differentiation.
is upregulated when Treg cells are activated by contact When stimulated with antibodies to 4-1BB, NK cells
with effector T cells. produce IL-2 and IFNγ, which increase the growth and
IL-35 appears to have a major role in regulation for cytotoxicity of antigen-specific T cells [1289, 2160].
IL-27β−/− Treg cells show loss of function. For example, 4-1BB ligation likewise stimulates dendritic cells to
they do not control homeostatic proliferation as wild secrete IL-6 and IL-12, which enhance antigen-specific
type cells do; they do not cure inflammatory bowel T cell response [2159]. In this way 4-1BB costimulates
disease. Recombinant IL-35 has been shown to induce T cells directly and indirectly by its cytokine stimulat-
Treg cell proliferation and production of IL-10. It sup- ing effects in several types of cells.
pressed the expansion of CD4+ CD25−effector T cells On binding its receptor, 4-1BB ligand is capable of
and it inhibited differentiation and IL-17 production by reverse signaling. In T cells, reverse signaling sup-
Th17 cells. And in treated mice, rIL-35 decreased the presses immune response and induces apoptosis [1764].
severity of collagen-induced arthritis [335, 1451]. In monocytes, reverse signaling induces IL-6, IL-8,
TNF-α and ICAM-1 but inhibits the production of
IL-10 [1059].
Several studies show that 4-1BB stimulation serves
4-1BB Iigand as an adjuvant in vaccines against cancer [693, 862,
1029, 1116, 1233, 1290, 1312, 1333, 1718, 2138, 2211,
Costimulation of T Cells, T Cell Memory 2215, 2280] and viruses [146, 390, 1958]. For example,
4-1BB ligand (4-1BBL) is a 30 kDa transmembrane in mice with melanoma, survival was shorter in mice
glycoprotein. T cells, stromal cells of thymus and spleen, that lacked 4-1BB compared to normal 4-1BB+ mice.
and antigen presenting cells including monocytes, B Treatment of normal mice with agonistic antibodies to
cells and dendritic cells express this ligand [23, 389, 4-1BB further increased survival [906]. Agonistic anti-
639, 1059, 1584]. The receptor is 4-1BB (CD137). It is bodies to 4-1BB enhanced the efficacy of a tumor lysate-
a 30–35 kDa transmembrane protein, which is related to dendritic cell based vaccination [1866].
the TNFR [1585, 1761, 1763]. The human gene is 4-1BB ligand has been detected in several carcinoma
located on chromosome 1p36 in a cluster of related cells. These tumors cells costimulate T cells and IFNγ
genes [1763]. There is a soluble form of this receptor, production. Reverse signaling stimulated tumor cell IL-8
the product of alternative mRNA splicing [1787]. 4-1BB production and depressed growth [1717]. Several mouse
was originally discovered as an activation-induced studies showed the benefit of injected mabs to 4-1BB
antigen on T cells [23, 639, 1044, 1045, 1765]. 4-1BB is suggesting it may be another receptor for which antibodies
also produced by B cells, dendritic cells, monocytes and may be useful in the treatment of human cancer.
200 Cytokines

CD27 ligand cells. When vaccine tumor cells are transfected with
CD27L, there is enhanced immune response [348, 1153,
Costimulation of T and B Cells; B Cell 1453]. CD27 ligation supports the clonal outgrowth of
Differention; Apoptosis tumor-specific T cells [638, 762]. Further, the CD27
system appears to be important in the production of T
CD27 ligand (CD27L, CD70) is a cell surface glyco- cells for adoptive cell immunotherapy [811, 1589]. IL-2
protein. It is a member of the TNF family [638]. induces growth of tumor-specific T cells and part of its
Activated B cells [1093], T cells [8, 1505], NK cells stimulatory effect is due to upregulation of T cell
[2226], dendritic cells [224] and certain B cell malig- CD27L. Blocking antibodies to CD27L depressed
nancies [1619, 928] produce CD-27L. It is expressed IL-2-induced growth. It appears that CD27 signaling
preferentially on CD45RO memory CD4 T cells [8]. In mediates IL-2-induced T cell proliferation [811, 1589].
dendritic cells, CD27L is induced by CD40L or by TLR It should be noted, CD27L [928, 1093] and CD27
stimulation [224]. Upon binding its receptor, CD27L [2061] are found on B cell malignancies and these mol-
may back-signal to increase cytotoxicity in γδ T cells ecules can have a role in tumor progression. Also,
[1505], antibody production in B cells [1094] and CD27L has been observed on certain non-hematological
growth of certain B cell malignancies [928]. tumors including renal cell carcinoma [429] and brain
CD27 is the receptor for CD27L [187, 638, 763]. tumors [732, 2177]. Tumor cell-CD27L causes apopto-
CD27 is a 50 kDa transmembrane glycoprotein. It is a sis in T and B cells and might have a role in immune
member of the TNFR family [233, 762, 2060]. CD27 is escape [429, 2177].
expressed on naive and memory T cells [8, 155, 1924,
2060], germinal center and circulating memory B cells
[984, 1253, 2015], B cell leukemias [2061] and on NK
cells [1923]. CD27 is considered a memory marker for T CD30 ligand
cells [2015] and B cells [985]. Activation of T cells T Cell Costimulation, Selection; Apoptosis;
markedly increases CD27 and induces the release of a
soluble form of the receptor, which can be found in blood
Regulation of Ig Class Switching
and urine. The soluble form appears to be a proteolytic CD30 ligand (CD153) is a 40 kDa transmembrane
product [761, 762, 1146]. Elevated levels are character- glycoprotein in the TNF family. It is found on mac-
istic of infection, autoimmunity and B cell malignancies. rophages [1840], B cells [598, 2259, 2260], CD4 and
Soluble recombinant CD27 is an antagonist [87]. CD8 T cells [2259], megakaryocytes, neutrophils, eryth-
The CD27–CD27L system is costimulatory for the rocyte precursors [598], eosinophils [1570], mast cells
induction of T cells. Stimulating antibodies to CD27 [541] and many though not all leukemias [599]. On
induce T cell activation, proliferation and cytotoxicity binding its receptor, CD30L has the capacity to signal
[66, 638, 992, 1924, 2060]. Naive (CD45+) T cells and back into its parental cell [2165]. Reverse signaling in
CD45RO memory cells respond to CD27 ligation [8, neutrophils stimulates an oxidative burst and IL-8 pro-
992]. In NK cells, CD27 expression is upregulated by duction. In T cells, reverse signaling stimulates prolif-
IL-2. CD27 ligation stimulates NK activation and eration and IL-6 production [2165].
cytotoxicity [1923]. CD30 is the receptor for CD30L. It is a 105–120 kDa
For memory B cells, CD27 appears to be part of the transmembrane protein and a member of the TNFR
switch between memory and activated plasma cells. family [473, 605, 1840]. The intracellular segment con-
engagement inhibits differentiation of B cells into tains two binding sites for TNFR-associated factors
Ig-secreting plasma cells [1616]. It favors the develop- (TRAFs 1, 2, 3) which function in signaling [601]. The
ment of memory B cells with high antigen affinity CD30 antigen was originally discovered as Ki-1, a
[1615]. marker for Reed Sternberg cells in Hodgkin’s lymphoma
CD27 engagement induces apoptosis in activated T and [1759]. CD30 is found on activated T, B, and NK cells
B cells. The cytoplasmic segment of CD27 is rather short. [35, 186, 489, 2088]. CD30 expression is transient; in T
Unlike TNF and Fas, CD27 does not have a death domain. cells it depends on activation and CD28 signaling [621].
However, this receptor does associate with a cytoplasmic CD30 is a marker for activated, cytokine secreting
protein, Siva, which has a death domain segment and Siva helper T cells (Th0, Th1 and Th2) [704]. In particular,
mediates CD27-induced apoptosis [1591]. CD30 expression is associated with Th2 response
In the treatment of cancer, there is interest in the [1255]; CD30 is stimulated by IL-4 and inhibited by
CD27 system for its ability to stimulate T, B, and NK IFN-γ [621]. CD30 is expressed on subsets of activated
Walter M. Lewko and Robert K. Oldham 201

CD45RO+ memory T cells [35]. CD30+ cells produced Sternberg cells [1336]. CD30L also appears to have a
IFN-γ and IL-5 while CD30− cells produced more IL-2. role in cutaneous inflammation. In atopic dermatitis and
CD30+ T cells showed enhanced ability to provide help psoriatic skin lesions, mast cells were the predominant
for B cells producing Ig [35]. CD30L-positive cell type. On binding CD30, back
Soluble CD30 is shed from cells and found in blood. signaling in these mast cells induced the secretion of
CD30 shedding is increased by activation of CD30 and IL-8 and other inflammatory chemokines. This mast cell
by treating cells with PMA. The protease responsible activation occurred in an IgE-independent manner [539].
is TACE, the enzyme that processes and releases Eosinophils express CD30L and they stimulate the
TNF-α [711]. Elevated levels of sCD30 are found in growth of co-cultured CD30-Hodgkin’s lymphoma cells
the blood of patients with immune disorders such as [1570]. Eosinophils also express CD30; treatment with
lupus erythematosis and rheumatoid arthritis and in agonistic Mabs to CD30 induced eosinophil apoptosis
patients with colon cancer and Hodgkin’s disease. In [142].
Hodgkin’s lymphoma patients, sCD30 correlates with A relatively small subset of normal lymphocytes
poor prognosis [2256]. Changes in levels may be used express CD30 but it is overexpressed in several diseases
to monitor a disease [711, 868, 904]. making it an attractive target for therapy. In T cells CD30
The specific biological effects of the CD30L–CD30 is increased in autoimmune disease and in allergy [614].
system are difficult to discern. Expression of the receptor The CD30 antigen is also found on virally transformed
is transient and certain effects appear to be opposed. and HIV-infected lymphocytes [729]. It is highly expressed
CD30 engagement is costimulatory for the growth and in Hodgkin’s disease, anaplastic large cell lymphoma
differentiation of specific T cells [621, 683, 1840]. But and certain other malignant lymphoid disorders [1759].
the CD30L–CD30 system also inhibits function and In many lymphoid cell lines, CD30 ligation induces
induces apoptosis [1407, 1840]. CD30-deficient mice, apoptosis but there are cases in which CD30 has no effect
have impaired negative selection of thymocytes sug- or stimulates growth [683, 1081, 1185]. Where CD30
gesting a fault in apoptosis [36]. CD30 transgenic mice ligation induces apoptosis in Hodgkin’s lymphoma cells,
overexpressing CD30 in the thymus showed enhanced the presence of soluble CD30 was found to interfere. This
thymic negative selection [303]. Moreover, CD30 may explain why sCD30 correlates with poor prognosis
signaling induced TCR-dependent apoptosis in a T cell in these patients [2259]. Antibodies are being developed
hybridoma cell line [186]. Lack of CD30 expression is which bind specifically to cellular CD30 epitopes that are
also associated with the development of autoimmune T lacking on the shed receptor [1411].
cells in experimental diabetes [280, 729, 1041]. On the In patients with Hodgkin’s disease, bispecific
other hand, in NOD mice that are genetically prone to (Anti-CD16/CD30) antibodies have been tested with
diabetes, neutralizing antibodies to CD30L interfered some success in a phase I/II trial. This construct was
with the development of the disease [280]. In a cyto- designed to activate NK cells and target the tumor cells
toxic large granular lymphocyte cell line, CD30 activa- [721]. SGN-30 is a humanized monoclonal antibody
tion depressed effector functions including FasL, that inhibits Hodgkin’s disease tumors in nude mice
perforin and granzyme production while inducing the [2088]; it is currently undergoing clinical trials.
effector cell’s own apoptotic program [1407]. CD30
thus appears be able to coordinate gene expression in
NK and T cells to control effector selection, growth and
differentiation and then downregulate cytotoxic func-
CD40 ligand
tions, inhibit growth and induce effector cell death. Costimulatory, Proinflammatory;
T cells influence B cell antibody production and
B, and T Cell Stimulation
class switching by way of the CD30L–CD30 and
CD40L–CD40 systems. CD40 ligation stimulates Ig CD40 ligand (CD154) is a 35 kDa membrane protein. It
class switching. CD30L reverse signaling inhibits class is expressed by CD4 T cells [1691] and by CD8 cells
switching at the level of CD40 signaling [273, 274]. [1707]. The levels of this powerful cytokine are rather
CD30 is also required for the development of B cell well controlled. It is produced during CD4 cell activa-
memory responses [593]. tion and down regulated upon binding its receptor [109,
Mast cells expressing CD30L appear to have a role in 260, 2058, 2240, 2241]. A soluble form of CD40L is
Hodgkin’s disease. Mast cells were the predominant released from CD4 T cells upon activation [653].
CD30L-positive cell type in tumors and these cells were Soluble CD40L is biologically active and will substitute
capable of stimulating growth of Hodgkin and Reed for cell-bound CD40L [955, 1055].
202 Cytokines

CD40 is the receptor for CD40L. It is a 50 kDa mem- CD40 activation exacerbated the disease while block-
brane protein, constitutively expressed, mainly on B ing CD40 depressed it. Development of autoimmunity
cells [266], and to a lesser extent on dendritic cells, is complex and sometimes paradoxical. The absence of
macrophages [21], T cells [511], fibroblasts [2241], thy- CD40–CD40L interactions might induce autoimmune
mic epithelial cells [585] and endothelial cells [786]. disease [1034]. Patients with rare X-linked mutations
CD40 is also present on some tumor cells. The CD40/ in the CD40L gene develop hyper-IgM syndrome,
CD40L system stimulates immune response, inflamma- show defects in thymus-dependent immune responses,
tion and apoptosis depending on the cell type and the antibody production and germinal center formation.
conditions. These patients suffer recurrent infection, increased
In B cells, CD40 signaling causes activation, matura- cancer and autoimmune disease [441, 727].
tion and survival [400, 946, 1294]. The triggering of B The CD40 system regulates apoptosis, positively or
cells through CD40 is critical for the induction of Ig negatively, depending on the target cells. In dendritic
production [72]. In the absence of CD40, B cells were cells, for example, CD40 ligation on immature DCs
tolerogenic [221, 952]. Binding Th2 cell CD40L induces induced survival and maturation, whereas CD40 liga-
reverse signaling and the secretion of IL-4 that in turn tion on LPS-matured DCs induced apoptosis [405].
stimulates B cells [167]. Where CD40 induces apoptosis, it up-regulates Fas.
The CD40 system provides help during CTL devel- Where CD40 inhibits apoptosis, it increases levels of
opment. CD40L on CD4 T cell interacts with CD40 on the survival protein bcl-X [858, 2113].
AP cells (DC, B cell, virus-cell or tumor cell) to induce The CD40–CD40L system has some remarkable
antigen processing, presentation and cytokine secretion antitumor effects. In normal B cells, CD40 ligation
[136, 756, 1645]. CD40 engagement also increases stimulated growth. In neoplastic B cells CD40 ligation
DC survival, costimulatory molecules B7.1 and B7.2, tended to inhibit growth and induce apoptosis [575].
ICAM-1, and cytokines such as IL-12, which stimulate Certain solid tumors express CD40. When this is the
T cells and NK cells [136, 159, 263, 267, 1645, 1753]. case, CD40L might act directly on the tumor cells to
The CD40L–CD40 system is necessary for a good induce death by apoptosis [756, 765, 2086]. CD40 liga-
immune response. CD40L-deficient mice were defective tion stimulates chemokine release. In human cervical
in antiviral immunity and memory CTL response [181]. carcinoma cells, CD40L induced macrophage chemoat-
Mice defective in the CD40 system were very suscepti- tractant protein-1 (MCP-1) and IFN-γ-inducible protein
ble to infection with Leishmania [236, 912, 1861]. 10 (IP-10). The addition of IFN-γ had a synergistic
Treating mice with stimulatory anti-CD40 Mabs pro- effect [32]. These chemokines are involved in the
duced a good T cell immune response to an otherwise recruitment of effector T cells. IP-10 is also angiostatic
weak Listeria monocytogenes immunogen [1658]. CD40 [1788]. As mentioned, CD40 ligation activates antigen-
activation also appears to be required for allograft rejec- presenting cells and secretion of IL-12 by DCs. Good
tion; blockade of the CD40 system generally prevents cytotoxic T cell responses were observed in animals
rejection and induces tolerance [470, 651, 863, 1064]. treated with stimulating anti-CD40 mabs, or immu-
CD40L is a proinflammatory cytokine. Engagement nized with tumor cells engineered to produce CD40L
of CD40 induces the production of inflammatory cytok- [430, 433, 446, 567, 695, 740, 1415, 1863]. CD40
ines in fibroblasts and macrophages [21, 2241], and the ligation enhanced the outgrowth and longevity of T
upregulation of surface adhesion molecules such as cells [1256]. CD40–CD40L interactions were required
ICAM and VCAM on fibroblasts and endothelial cells for protective anticancer immunity by vaccination
[786, 1062, 2241]. Treating animals with soluble CD40L [1196, 695]. When CD40 signaling was blocked, no
intranasally induced lung inflammation with infiltrating systemic antitumor immunity developed [1195].
neutrophils and macrophages. IFN-γ intensifies CD40L- CD40 signaling stimulates NK activity and related
induced inflammation [2163]. Platelets contain both antitumor effects [245, 1415, 2038]. P815 mastocytoma
CD40 and its ligand and may be involved in CD40- cells engineered to produce CD40L were promptly
related inflammation and immune responses [377, 953]. rejected when injected into mice. NK cells mediated the
CD40 on lung fibroblasts appears to have a role in anticancer effect. Production of IL-12 was required but
pulmonary fibrosis [2241]. effector CTL cells were not [1415]. In another study,
CD40–CD40L interaction is also required for a mice were treated with stimulatory anti-CD40 mabs to
number of experimental autoimmune diseases. In activate signaling [2038]. Three different types of
animal models for myasthenia gravis [848], multiple tumors were tested; growth and metastasis were inhibited.
sclerosis [615, 667], arthritis [471] and diabetes [105], NK cells were required. The effect of CD40 was not
Walter M. Lewko and Robert K. Oldham 203

direct but probably through APCs that produced NK cell appears to be the only ligand for CXCR4 [1409].
stimulating IL-12 [2038]. ELR CXC chemokines tend to activate migration in
A phase I trial of rhuCD40L in patients with solid neutrophils, the non-ELR CXC chemokines tend to
tumors and NHL, encouraging preliminary results were activate lymphocytes.
seen including one long-term complete response [2087]. 2. CC chemokines (β chemokines): the first two con-
In a phase I vaccine trial, patients with CLL were treated served cysteines are together. There are nine known
with CD40L-producing autologous tumor cells. receptors for these chemokines termed CCR1, CCR2,
Leukemia cell counts decreased and lymph node size etc. Well studied CC chemokines include MIP-1α
was reduced again suggesting that CD40L may be thera- (macrophage inflammatory protein-1α), MIP-1β,
peutically useful [2154]. MCP (Monocyte chemoattractant proteins) 1, 2, 3,
RANTES and eotaxin. CC chemokines are chemot-
actic for most types of leukocytes (T, B, DC, NK,
Chemokines eosinophils, basophils, DC), but not for neutrophils.
[1656].
Cell Activation, Migration, 3. C chemokines: two of the four conserved cysteines
and Recruitment; Inflammation; are missing, the first and the third. Lymphotactin is a
C chemokine. NK cells produce it. Lymphotactin is
Angiogenesis specifically attractive for lymphocytes [949].
Chemokines include a rather large number of proteins, 4. CX3C chemokines: the first two conserved cysteines
which have a variety of activities but function primarily have three intervening non-conserved amino acids.
as chemoattractants and activators of leukocytes [1656]. There is a specific receptor Cx3CR-1 for these cy-
They induce cell migration, recruitment and infiltration tokines [849]. Fractalkine (neurotactin) is a Cx3C
into sites of inflammation, homing to sites of immune chemokine. It has two active forms; one is a mem-
cell differentiation and passage across cellular barriers. brane bound protein. The other is a soluble protein
They are involved in T cell activation and the polariza- released from the membrane by the protease TACE
tion of Th1/Th2 cells [687, 1656]. Chemokines may [2030]. Fractalkine is expressed in neurons, mi-
also act on non-hematopoietic cells. They facilitate cell croglial cells, fibroblasts and endothelial cells. It is
movement during inflammation, wound repair and upregulated during inflammation. Fractalkine is a
morphogenesis. Chemokines are also involved in chemoattractant for T cells, NK cells and monocytes
pathology, tumor growth and metastasis. HIV viruses and it induces adhesion [119, 1530]. In the brain
use certain chemokine receptors (CXCR4) for entry fractalkine appears to be antiapoptotic, a survival
into cells [524]. factor for microglial cells [171].
Chemokines are relatively small 7–14 kDa proteins. ELR+ CXC chemokines are angiogenic; they stimulate
Most chemokines contain four conserved cysteines in endothelial cell migration and tube formation; these
the amino terminal region. Chemokines are classified include IL-8, GRO (growth-related oncogene)-α,
based on the sequence in the region of these cysteines. GRO-β, GRO-γ, GCP-2 and ENA-78 (epithelial neutro-
1. CXC chemokines (α chemokines): the first two phil-activating protein 78) [1906]. The receptor CXCR2
conserved cysteines are separated by a single non- is responsible for ELR+ angiogenesis [5]. Non-ELR
conserved amino acid. CXC chemokines may be CXC chemokines tend to be inhibitors of angiogenesis.
classified further based on the presence or absence IP-10 and Mig are angiostatic [1204, 1906]. Both cytok-
of Glu-Leu-Arg (ELR) just preceding the first con- ines bind receptor CXCR3 [2142]. The anticancer activ-
served cysteine. There are at least five receptors for ities of several cytokines may involve the downstream
CXC chemokines. These receptors are members of production of angiostatic chemokines such as IP-10 and
the rhodopsin-like, seven transmembrane domain Mig [32, 52, 222, 1788].
receptor family [1147]. As examples of chemokine– Ephrins and ephs, their receptors, are also involved
receptor interactions, CXCR1 and CXCR2 bind in cell migration. They act as chemodirectants during
IL-8 and GCP-2 (granulocyte chemotactic pro- morphogenesis, inflammation and immune response.
tein-2) [5]. CXCR3 binds IP-10 (interferon-induced Ephrins and ephs are found on a wide variety of cells.
protein-10) and Mig (monokine induced by γ inter- In contrast with the chemokines, which mainly stimu-
feron) [894]. SDF-1 (Stromal derived factor-1), a late migration and adhesion, the ephrins are chemore-
pre-B-cell growth factor as well as a chemokine, pulsive agents which tend to be transiently expressed
204 Cytokines

at times when the movement of cells needs to be system is involved in activation-induced cell death in
redirected. lymphocytes. The tumor cells that express FasL are gener-
Chemokines are involved in many inflammatory dis- ally negative for Fas and tend to be resistant to apoptosis
eases and in the development of cancer. Cell movement [700, 1468, 1469]. The expression of FasL may explain the
and angiogenesis are hallmarks of malignancy. Elevated general phenomenon of immune resistance (immune
levels of angiogenic chemokines are associated with privilege), by which certain normal tissues (eye chamber,
tumor progression and metastasis [63, 976, 1167]. IL-8 testis) and tumors are able to evade immune effector cells.
production has been correlated with tumor vascularity Cells that express surface FasL may induce apoptotic death
and progression [976, 2250]. Chemokines are of interest in lymphocytes [670, 700]. In tumor biology this idea has
in the design of therapeutic agents, including antitumor been referred to as “FasL counterattack.” in which infiltrating
vaccines. For example, lymphotactin is produced by lymphocytes may be killed upon contact with FasL+ tumor
NK cells and attracts T cells [949]. A vaccine using cells [135, 1468, 1469]. This theory is interesting but con-
tumor cells co-transfected to produce lymphotactin and troversial. Certain reports show that tumors expressing FasL
IL-2, induced potent antitumor immunity in mice [432]. may be rejected [61, 124, 519, 1778]. In transplantation
In another study, a vaccine with Lewis lung carcinoma research, some have tried to engineer graft tissues with
antigen-pulsed dendritic cells, transfected to produce FASL to make them less immunogenic but with they have
lymphotactin, induced a much more potent antitumor met with variable success [31, 916].
response than a control vaccine lacking lymphotactin. Tumor cells may contain Fas. When this is the case,
In mice with established tumors, vaccination with the tumor development and therapy may be influenced by
antigen-pulsed lymphotactin-DCs inhibited metastasis. Fas-induced apoptosis. Dendritic cells express FASL, as
Only one relatively small 104 DC dose was required to well as other proapoptotic cytokines TNF, LTα1β2,
immunize the mice [241]. TRAIL, and may kill tumor cells directly [1166]. CTLs
express FASL. Irradiation was found to increase Fas on
tumor cells and prior irradiation enhanced apoptotic
response to adoptive CTL therapy [281]. Vitamin E
Fas ligand stimulated Fas and Fas ligand levels in certain cultured
breast cancer cells and induced death by apoptosis
Inducer of Apoptosis [2037]. In some leukemias and solid tumors, drugs such
Fas ligand (FasL) is a transmembrane protein in the as adriamycin and methotrexate induce Fas and FasL
TNF family. It is expressed on activated lymphocytes causing death [566].
and on a variety of other cells including certain tumor Capillary leak syndrome involves endothelial cell
cells. In T cells, FasL is induced by TCR activation. Its damage. It is associated with chronic infection and it is
production is inhibited by retinoids and glucocorticoids a dose-limiting toxicity in the use of IL-2 and certain
[2107, 2228], by inhibitors of protein kinase C and other cytokines to treat cancer patients. Capillary dam-
calcium mobilization [2107], and by cyclosporin [217]. age appears to be due to cytotoxic lymphocytes [375].
Fas is the receptor for FasL [1410, 1839, 1976]. Fas Studies using perforin knockout mice and mice with
(also known as Apo 1, CD95) is expressed on lympho- defective FasL and Fas genes showed that both perforin
cytes and many different types of cells including tumor and Fas-FasL were responsible [1612].
cells. There are soluble forms of Fas; they are gener-
ated by alternative mRNA splicing [253]. The FasL–
Fas system is responsible for the induction and
regulation of programmed cell death in the peripheral
FLT-3 ligand
immune system. Apoptosis is a way to regulate and ter- Stimulator of Early Hematopoietic
minate an ongoing immune response, or to eliminate
useless or potentially damaging immune cells [1019,
Stem Cells and Dendritic Cells
1184].The importance of the FasL/Fas system in the Flt-3 ligand is a membrane-bound cytokine. In structure
regulation of lymphocytes can be seen in the autoim- and activity, it is related to macrophage colony stimulating
mune diseases and lymphoproliferative disorders in factor and stem cell factor [710, 1175]. The human
mice, associated with mutations in the FasL (gld) and gene is located on chromosome 19 [1176, 1265]. It is
Fas (lpr) genes [330, 1410, 1698, 1976]. expressed by a wide variety of hematopoietic and non-
Various types of cancer and tumor cell lines express FasL hematopoietic tissues [710, 1176]. Flt-3 ligand is produced
[135, 700, 1452, 1469, 1703, 1814, 1897]. The Fas–FasL in both membrane bound and soluble forms, both active.
Walter M. Lewko and Robert K. Oldham 205

Soluble forms are produced by proteolysis [1177] or NK cells in blood, bone marrow, spleen and liver [1555,
alternative mRNA splicing [1176, 1265]. Soluble Flt-3 1801]. It appears to do this by stimulating proliferation
ligand is elevated in serum of patients with certain ane- of pro-NK cells and the production of mature, non-activated
mias [1179]. In patients on myelosuppressive chemo- NK. Flt-3 ligand increases NK cell responsiveness. IL-2
therapy, elevated plasma Flt-3 ligand is prognostic for rapidly induces proliferation, cytotoxicity and LAK
poor recovery from thrombocytopenia [170]. activity in Flt3 ligand-treated cells [1801].
Flt-3 (fms-like tyrosine kinase-3, CD135) is the Flt3 ligand influences certain non-hematopoietic
receptor for Flt-3 ligand. It is a transmembrane protein. cells. Osteoclasts respond to M-CSF with increased
Flt-3 was named for its similarity to fms, the receptor differentiation; Flt-3 ligand can substitute for M-CSF
for M-CSF [1679, 1680]. Flt-3 is also referred to as flk-2 [1069]. Neural stem cells express Flt-3; Flt-3 ligand
(fetal liver kinase 2) [1251] and Stk-1 (stem cell tyrosine inhibited EGF and FGF-2 induced proliferation. Neurons
kinase-1) [1837]. It is a 158 kDa glycoprotein. The are also Flt-3+. In culture, Flt-3 ligand synergized with
human gene is on chromosome 13q12 [1680]. This nerve growth factor to promote neuron survival [195].
receptor is expressed mainly by hematopoietic stem Flt-3 is expressed by many myeloid and lymphocytic
cells and progenitor cells [1248]; it is low or missing on leukemias [157, 370, 1286, 453, 1271, 1678]. When cells
most mature cells [370, 1251, 1286, 1837]. Flt-3 ligand are positive for the receptor, flt-3 ligand may stimulate
binds and activates the receptor. Dimerization of Flt-3 growth in culture. In AML, 20–25% of adult patients
ligand appears to be required for receptor dimerization have a mutation in flt3. This mutation is an internal
and signaling [710, 1177, 1178]. tandem duplication. The presence of this mutation is an
Flt-3 ligand has a role in the proliferation, survival and adverse prognostic factor. This is a gain of function muta-
differentiation of hematopoietic progenitor cells [1269]. tion that confers growth advantage on the cells [2151];
Mice lacking Flt-3 ligand have reduced numbers of signaling is constitutively activated [725]. Selective
myeloid progenitors, B-lymphoid progenitors, NK cells, tyrosine kinase inhibitors are being tested in patients with
and dendritic cells [1272]. Mice lacking Flt-3 appear flt3+ tumors. In a trial on elderly patients with AML,
healthy. However, their stem cells are deficient in their responses were observed for lestaurtinib (CEP701) in
ability to reconstitute lymphoid and myeloid cells when both mutated and wild type Flt3 tumors [989].
transplanted into irradiated mice [1192]. Mice injected Tumor bearing mice treated with Flt3 ligand generate
sc with Flt-3 ligand show a remarkable increase in antitumor responses. Several preclinical studies have
hematopoietic precursors [193, 194]. Flt-3 ligand stimu- shown this in lymphoma, leukemia, and several types of
lates the growth and survival of early progenitor cells solid tumors [283, 320, 499, 1183, 2097]. Soluble and
[909, 1298, 1448]. GM-CSF, IL-3, SCF, IL-11 and IL-12 membrane bound forms of Flt-3L were effective.
are synergistic [909, 1298] while TNF-α and TGFβ are Dendritic cells and CD8 T cells appeared to be respon-
inhibitory [2068]. Flt3 ligand acts together with IL-7 or sible for antitumor activity though NK cells might also
SCF to stimulate B cell development from progenitors in be involved [1546, 1555, 1804, 1826]. In a mouse model
bone marrow and thymus [768, 823, 1272, 1424, 2067]. for metastasis, local irradiation plus subsequent Flt-3
Flt-3 ligand has a remarkable influence on the devel- ligand treatment enhanced survival. This appeared to be
opment of dendritic cells. Mice treated with Flt-3 ligand due to increased presentation of antigen derived from
produce greater numbers of dendritic cells [452, 1224, dying tumor cells. In vivo manipulation of dendritic
1819, 1890]. Spleen cells with markers for dendritic cells was used to induce immune response in three
cells went from less than 1% of total in controls to 20% mouse model tumors. Mice were treated with several
in Flt-3 treated mice [1224]. DCs from treated mice daily Flt3 ligand injections to induce DCs, then with CG
expressed more costimulatory CD80 and CD86 [1890]. oligonucleotide to mature the DCs in the presence of
Patients treated with Flt-3 ligand, also showed a remark- tumor peptide antigen. Existing tumors were inhibited
able increase in dendritic cells [1221]. Flt-3 ligand and and the mice were protected against a new challenge
stem cell factor cooperate to induce recruitment and [1490]. It should be noted again that Flt-3 ligand acts
expansion of CD34+ CD14+ dendritic cell precursors together with other cytokines. For example, Flt-3 ligand
(CFU-DC). Flt-3 ligand and SCF are not required for and CD40 ligand were shown to be synergistic in the
the differentiation step. Other cytokines, including induction of antitumor immunity. Blocking CD40
GM-CSF, IL-4, TGF-β and TNF-α stimulate maturation obviated the effect of Flt3 ligand [179].
of the expanded precursors [367]. Flt3 ligand does not directly influence the growth of
Flt-3 ligand also stimulates the production of NK most types of non-hematological tumor cells in culture,
cells [194]. Treatment of mice increased the number of although it has been reported that neural crest tumors
206 Cytokines

express Flt-3 and that cell lines treated with Flt-3 ligand [1168] thymic epithelium [1067] and synoviocytes [395,
showed increased growth and decreased apoptosis [1999]. 249]. The gene for LIF is on chromosome 22q12 where
Flt-3 is an apparent site of immune suppression. it colocalizes with the gene of OSM. In immune cells,
Vascular endothelial cell growth factor is produced by secretion of LIF is stimulated by endotoxin, IL-1 and
many tumors; it is an angiogenic factor and it is also TNF [1297]. In thymic epithelial cells, EGF and TGF-β
immunosuppressive. VEGF interferes with Flt-3- stimulate LIF production [1743].
mediated DC production. It appears to do so by inhibiting The receptor for LIF has two subunits, LIFRβ and the
the activation of the transcription factor NF-κB [1483]. common gp130 utilized by IL-6 family cytokines [602].
There is interest in Flt-3 ligand for the treatment of LIF has a role in the production of CD4 and CD8 T
cancer patients undergoing stem cell transplants. cells. LIF is secreted by thymic epithelium [1067]. Too
Patients may be pre-treated with Flt-3 ligand to mobi- much or too little LIF causes abnormal T cell develop-
lize stem cells to increase the number available for har- ment [1295]. In transgenic mice that overproduced LIF,
vest. Flt-3 may also be used to expand these cells in thymic epithelial architecture was disrupted and cortical
culture. Studies in normal volunteers and in cancer thymocytes were decreased [1804]. In LIF deficient
patients showed that Flt-3 ligand is relatively safe. Few mice, thymocytes were produced but they were insensi-
side effects were observed [1098, 1221]. As seen in tive to ConA activation [498].
mice and other animals, Flt-3 ligand caused a remark- LIF regulates inflammation. Depending on the set-
able increase in stem cells and dendritic cells in blood ting, LIF may be pro- or anti-inflammatory. LIF knock-
and other tissue sites [1221]. Flt-3 ligand is effective out mice showed abnormal inflammatory response in
alone but most likely it will be used in combination with nervous tissues [579, 1926, 2003]. Injection of rLIF into
other cytokines such as G-CSF and GM-CSF [193, 194, skin and joints induces swelling and infiltration [248,
1920]. In culture, stem cells and progenitor cells may 1277]. LIF is elevated in ulcerative colitis; LIF stimu-
proliferate for several weeks when Flt-3 ligand is added lates the growth of cancer cells and may thus have a role
alone or together with other cytokines such as IL-1, Il-3, in the development of colon cancer associated with this
Il-6, IL-11, IL-12, erythropoietin, SCF and thrombopoi- inflammatory disease [622]. LIF is proinflammatory,
etin [381, 578, 872, 1564, 1568, 1860]. Flt-3 ligand but there is also evidence LIF can inhibit inflammation.
appears to maintain the stem cells, to sustain long-term In knockout mice lacking LIF, inflammation is exacer-
hematopoiesis after reimplantation [381, 1564]. bated in footpads injected with Freund’s complete adju-
Trials have begun in cancer patients, using Flt-3 ligand vant. Delivery of LIF using an adenoviral vector
to treat patients or to generate dendritic cells for vaccina- suppressed inflammation [2296]. In psoriasis, LIF and
tion. In a preliminary study, colon cancer patients injected IL-11 have similar depressive effects on inflammation
with Flt3 ligand showed increased lymphocytes in blood, and cytokine production [2022].
increased percent dendritic cells in PBMCs and increased LIF is involved in septic shock. The blood of patients
dendritic cells infiltrating tumors [1366]. In another report, had elevated LIF as well as TNF, IL-1 and IL-6; the level
patients were vaccinated with antigen-pulsed autologous of these cytokines was correlated with the severity of the
dendritic cells. The patients were pre-treated with Flt-3 disease [2077, 2118, 2119]. LIF and IL-6 appear to be
ligand to mobilize DCs. The DCs were harvested, loaded induced by TNF-α [880]. Blocking antibodies for LIF
ex vivo with a peptide of carcinoembryonic antigen and depress the lethality of endotoxemia [166]. Interestingly,
then reinfused. The preliminary results were promising. treatment of mice with LIF before a lethal dose of endo-
Two of 12 patients showed dramatic regressions; one had toxin favored survival of the mice [26, 2117]. This may
a mixed response. Anticancer activity correlated with the be due to induction of acute phase proteins that have
generation of specific CD8 T cells [549]. antiinflammatory, protective effects [1258].
LIF has growth, differentiation and metabolic effects
in non-hematopoietic tissues [759, 1042, 1295, 1297].
LIF reversibly inhibits the differentiation of embryonic
Leukemia inhibitory factor stem cells [1449]. In fetal pituitary corticotropes, LIF
T Cell Development, Regulator of inhibits cell division, stimulates differention and
increases ACTH production [1884]. LIF regulates neu-
Inflammation, Embryonic Development ronal differentiation and increases survival [1345]. It
Leukemia inhibitory factor (LIF) is a member of the IL-6 stimulates bone development and extracellular matrix
cytokine family [2273]. It is a pleiotropic cytokine; many metabolism. LIF regulates mammary cell growth and
of its effects overlap with those of IL-6. LIF is produced development [1137]. Maternal expression of LIF is
by fibroblasts [2008], macrophages [51] endothelial cells required for blastocyst implantation [1892].
Walter M. Lewko and Robert K. Oldham 207

Mice injected with LIF suffer weight loss associ- As typical of TNF-family members, LIGHT induces
ated with cachexia. Weight loss is a characteristic of apoptosis. Normally, only cells that express both HVEM
several gp130 signaling cytokines, IL-6, IL-11, CNTF and LT-β receptors are susceptible to LIGHT-induced
and NNT [1784]. cell death. No apoptosis was observed in cells express-
LIF has some remarkable effects in cancer. As men- ing only one of them [719, 2278]. Because lymphocytes
tioned, it is required for normal immune and inflammatory do not express LT-β receptor, LIGHT does not cause
response. In tumor cells, LIF has both negative and posi- apoptosis in these cells.
tive direct effects on growth. LIF was discovered based on TR6 (also called DcR3) is another member of the
its ability to arrest growth and induce differentiation in TNF receptor family. TR6 is a soluble, non-signaling
murine myeloid leukemia cells [604]. In cultured human protein that binds both LIGHT and FasL and is, there-
glioma cells, LIF inhibits growth and induces astrocyte- fore, a decoy receptor. TR6 appears to have a regulatory
like differentiation [701]. But LIF can stimulate growth in role in LIGHT and FasL mediated cell death [2265].
many types of tumor cells including human multiple LIGHT is expressed on immature dendritic cells.
myeloma [2285] colon cancer [964], breast cancer [425, Engagement of LIGHT costimulates T cell proliferation.
500, 948] and prostate cancer [948]. Other cytokines in Blockade of LIGHT, using soluble recombinant receptors,
the IL-6 family similarly influence tumor cell growth inhibits DC-mediated allogeneic T cell response [1957].
based on gp130 signaling [2285]. There is interest in LIGHT stimulates inflammatory response. Triggering
agents that regulate growth by this signaling pathway. HVEM together with toll-like receptors on neutrophils
synergistically stimulates respiratory burst, degranulation,
secretion of IL-8 and opsonization of particles [722].
At an early stage in liver regeneration, lymphocytes
LIGHT infiltrate the hepatic tissue. Hepatocyte cell division is
Costimulatory Molecule; stimulated and this depends on the expression of T cell-
LIGHT and liver cell-LTβR [45].
T Cell Activator; Inducer of Apoptosis
LIGHT costimulates T cell anticancer immunity.
LIGHT is a 240 amino acid cytokine that was first dis- Typically immune cells express LIGHT but it is also
covered in the cDNA library of activated T cells. It is a found in melanoma cells and on microvesicles released
member of the TNF family [1044, 1254]. LIGHT is from melanoma; co-culture of microvesicles with lym-
homotrimeric and it is produced in both soluble and phocytes in the presence of IL-2 costimulated CD8 T cell
membrane bound forms, both active. LIGHT is expressed proliferation and apoptotic death in the melanoma cells
by T cells, granulocytes, monocytes, immature dendritic [1368]. Human breast cancer cells transfected with LIGHT
cells and certain tumors [719, 1368, 1957, 1959, 2278]. cDNA triggered immune response and regression in an in
LIGHT binds two receptors, HVEM (herpes virus vivo model system [2278]. NK cells express HVEM;
entry mediator) and lymphotoxin β receptor (LTβR) tumor cells that express LIGHT activate NK cells to kill
[2278]. HVEM is a member of the TNFR family [1044, tumors in a manner that requires IFNγ and CD8 cells. The
1352, 1958]. It is the cell receptor for herpes simplex NK cells provided help (IFN) in the activation of CTLs
infection [1344, 1254]. HVEM is also referred to as [509]. With respect to apoptosis, LIGHT triggers cell
TR2 (TNF-related receptor 2) [1044] and ATAR (another death in tumor cells. Normally, two receptors (HVEM and
TRAF-associated receptor) [808]. It is a 283 amino acid LTβR) are required for LIGHT-induced apoptosis but in
transmembrane protein. Signaling involves TRAF bind- tumor cells only one, the LTβR, was necessary [1662].
ing and the activation of NF-κB, Jun N-terminal kinase
and AP1 [808, 1231, 1254]. HVEM is localized on T
cells, B cells, NK cells, monocytes, endothelial cells
and immature dendritic cells [720, 1044, 1344, 1958]. Lymphotoxin-a (Tumor Necrosis
Its levels are up regulated by cell activation and down
regulated by LIGHT [1958]. HVEM is also a receptor
Factor b)
for lymphotoxin-α [1254]. Lymphoid Organogenesis; Inflammation;
In T cells, LIGHT costimulates proliferation, cytokine
secretion and surface protein levels [720, 1044]. Disruption
T Cell, B Cell, Bone Cell Development
of LIGHT, interferes with lymphnode formation, depresses Lymphotoxin α (LT α) was discovered in the condi-
T cell proliferation in a MLR, the secretion of IL-2, IFN-γ, tioned medium of activated T cells [505, 1078, 1695].
IL-4 and TNF-α [720, 1739]. Disruption of LIGHT also It was named for its lymphocyte origin and its toxicity to
prolongs allograft survival [2237]. certain cells. The gene for human LTα is on chromosome 6,
208 Cytokines

close to the TNF-α gene. LTα is a TNF family member; to stimulate bone resorption [1883]. In a model for
it is also referred to as TNF-β [9, 1553]. LTα is pro- periodontal disease, soluble recombinant receptors for
duced by Th1 cells, CD8 T cells, early B cells, NK cells IL-1 and LT-α, acted as antagonists and depressed
and also by astrocytes. LTα is synthesized as a 202 recruitment of inflammatory cells, osteoclast activity,
amino acid precursor with a signal peptide and it is bone loss and periodontal destruction [83].
cleaved to 171 and 194 amino acid forms [9]. LTα self-
associates to form homotrimers (LTα3) with a confor-
mation similar to TNF-α. It is a soluble cytokine. The
homotrimer binds both TNFRI and TNFRII [209, 212,
Lymphotoxin-b
777, 1112]. Because the TNFRII-LTα3 complex does Lymphoid Organogenesis; Inflammatory;
not signal, for LTα3 TNFRII is a decoy receptor and an
NK, Dendritic Cell Development
antagonist [1281]. LTα3 also binds herpesvirus entry
mediator (HVEM), a receptor for LIGHT [1254]. Lymphotoxin-β (LT-β) is a 33 kDa transmembrane pro-
Lymphoid organ development depends on LT-α and tein that was originally cloned from a T cell hybridoma
other cytokines such as LT-β, TNF-α and LIGHT. [212, 685]. It is related to LT-α in structure and func-
LT-α(−/−) mice, were born without lymph nodes or with tion. LT-β is expressed mainly on activated T cells, B
defective nodes. Peyer’s patches were also lacking. cells and NK cells [2116]. LT-β chains form homotrim-
Spleen histology was abnormal [110, 419, 1004, 1706]. ers and heterotrimers with LTα; LTα1-LT-β2 is the major
Both LT-α and LT-β appeared to be required for normal form [209, 212, 777, 2116].
lymphoid organogenesis [1249, 1445, 1636]. LT-β binds the LT-β receptor (LT-βR). This receptor is
LT-α and TNF-α are involved in the formation of related to the TNF receptors. It binds LT-β in its trimer
B cell follicles, T and B cell segregation, follicular and heterotrimeric forms; it does not bind TNF-α or the
dendritic cell clustering and the formation of germinal LT-α homotrimer [209, 211, 212, 355, 766]. LIGHT is
centers. Mice deficient in either LT-α or TNF-α also a ligand for the LT-βR [2278]. Follicular stromal
failed to form germinal centers [502, 2110]. cells, monocytes, fibroblasts, smooth muscle and skeletal
LT-α (−/−) mice have impaired effector T cells. These muscle cells express the LT-βR. It is also on human mel-
mice were much more susceptible than controls to HSV anoma and adenocarcinoma cell lines [211, 402, 2051].
infection. CD8 T cells were induced normally but their LT-β, LT-α, TNF-α and LIGHT are all critical in the
effector functions were depressed. The cells failed to development of lymphoid organs [1249, 1445, 1636,
become CTL and did not secrete INF-γ when stimulated 1739]. LT-β was also required for the homing of den-
with antigen [1035]. LT-α was also required for the dritic cells to lymph nodes [2206]. LT-β induces IFN-β
development of memory B cells and their capacity to and this promoted survival of lymphocytes during
respond to antigen [571]. murine cytomegalovirus infection [110].
LT-α has a major part in inflammation. It induces the LT-α and LT-β stimulate the development of NK
expression of leukocyte adhesion molecules VCAM-1 cells. LT-β(−/−) mice had fewer NK cells [841, 861,
and ICAM-1 on endothelial cells [265, 1581]. In a 1846] and LT-βR was required for normal NK develop-
mouse model, antibodies to LT-α and TNF-α prevented ment [841, 2205]. The effect of LT-β was not dependent
allergic encephalitis [1694]. Transgenic mice overex- on IL-15, another essential cytokine in NK maturation
pressing LT-α have remarkable inflammation in the sites [841]. It appeared that an effect of LT-β on marrow
of targeted expression. The infiltrate consists of T cells, stromal cells was critical for an early step in NK devel-
B cells, macrophages, and dendritic cells [1020, 1569]. opment [2205].
TNF receptors were studied in this system. Mice lacking LT-α and LT-β are required for the normal develop-
TNFRI failed to develop inflammation. TNFRI appears ment of NK T cells. These cells are important in that
to be the primary receptor involved in LTα-induced they may be stimulated to quickly produce large amounts
inflammation. Lack of TNFRII did not did not influence of IL-4, IFN-γ, and IL-10. They are involved in certain
inflammatory response. Lack of the related cytokine types of antitumor activity, the prevention of autoim-
lymphotoxin β did not prevent inflammation but altered munity and protection against bacterial and parasitic
the response [1705]. infections. Mice that are LT-α(−/−) and LT-β(−/−) have
LT-α is involved in bone and tooth metabolism. reduced NK T cells and they are low in IL-4 and IL-10.
TNF-α and LT-α stimulate osteoblast and osteoclast Both LT-α and LT-β are needed suggesting LT-α1β2
activity [1988]. IL-1 acts synergistically with TNF (not the homotrimer) is the active form [483].
Walter M. Lewko and Robert K. Oldham 209

LT-β has both cell mediated and direct effects on cancer.


It is required for normal production of dendritic, NK,
Oncostatin M
NK T and LAK cells. Mice lacking the LT-βR showed Lymph Node/T Cell Development;
enhanced tumor growth and metastasis [861]. Direct LT Inflammation; Growth and Wound Repair
effects have also been observed in certain human adeno-
carcinoma cells that have the LT-β receptor. Signaling Oncostatin M (OSM) is a 28 kDa cytokine. It was dis-
through the LT-βR induced cell death in culture and covered in the conditioned medium of lymphocytes as a
arrested tumor growth in vivo [211, 1194]. Human mel- factor capable of inhibiting certain cancer cells [207,
anoma cell lines also express the LT-βR. When the 1210, 2274]. Activated T cells, monocytes and neutro-
receptor was activated, growth was inhibited and proin- phils produce OSM [207, 213, 666, 826]. The gene for
flammatory cytokines were secreted [402]. OSM is on chromosome 22q12 where it colocalizes
with the gene for LIF. It is a member of the IL-6 cytokine
family that utilizes gp130 as a receptor component
[1664, 2273].
Novel neurotrophin In humans, OSM binds and signals through two
B Cell Development; Regulator receptors. The first is the receptor for LIF, gp130-LIFRβ.
It is responsible for effects induced in common by OSM
of Inflammation and LIF [601, 602]. The second is gp130-OSMRβ,
Novel neurotrophin (NNT) is a member of the IL-6 which is a specific OSM receptor and is responsible for
cytokine family. It is also referred to as B cell stimulat- the effects particular to OSM [1373, 1984]. OSM recep-
ing factor-3 and cardiotrophin-like cytokine. NNT is a tors are on a variety of cells including tumor cells. The
225 amino acid protein. It is homologous with IL-6 fam- OSM mechanism of signaling involves MAP kinase,
ily members, in particular cardiotrophin-1 (CT-1) and JAK-STAT and the PIP3-kinase pathways [38, 99,
ciliary neurotrophic factor (CNTF). The gene for NNT 837, 1009].
is on chromosome 11q13 where it is close to the gene Oncostatin M has remarkable effects on lymph node
for CNTF (11q12). mRNA for NNT is produced by development and the production of lymphocytes.
lymph nodes and spleen and to a lesser extent by other Thymus is a key site for T cell processing. But even in
tissues [1783, 1808]. athymic animals a certain amount of T cell produc-
The receptor for NNT contains ciliary neurotro- tion occurs in bone marrow, liver, intestine and in
phin Rα, gp130 and LIFRβ. Signaling, involves the lymphnodes. In transgenic mice overexpressing oncostatin
JAK/STAT pathway and the activation of STAT-3. In M, thymus atrophies and T cells (immature and mature)
this way NNT resembles other IL-6 family members accumulate in the lymphnodes [175, 325, 1209]. High
[384, 1088]. OSM endows lymph nodes with the ability to sustain
NNT stimulates B cells. Increased B cell production T cell development. OSM also mobilizes lymphocytes
and secretion of IgM, IgE, IgG was observed in mice causing them to move into lymphnodes and to recycle
treated with NNT [1784, 1783]. These mice suffered into circulation.
weight loss. NNT, like certain other IL-6 family mem- OSM has multiple effects related to wound repair and
bers, induces cachexia. As its name implies, this cytokine inflammation. It is a growth stimulator for fibroblasts
is stimulates nerve growth. In culture, NNT increased [793] and vascular smooth muscle cells [679]. OSM is a
the survival of chick embryo neurons [1784]. Gene defi- fibroblast activator. It stimulates growth of 3T3 cells,
ciency studies showed that NNT and the ciliary neu- dermal fibroblasts and synoviocytes [694]. OSM
rotrophic factor receptor were required for motor neuron increases the production of extracellular matrix colla-
development [396, 1099]. In mice, NNT induced the gen [836, 837] and glycosaminoglycan [469]. It stimu-
production of serum amyloid A protein. It also potenti- lates the production and activation of collagenases that
ated IL-1-induced secretion of glucocorticoids. Many of degrade the extracellular matrix [705, 1644]. Lytic
these effects are shared with certain other members of enzymes and collagen metabolism are part of the growth
the IL-6 family, but there are differences. NNT was process in connective tissue. OSM also increases IL-6
characteristic in that it did not induce hematopoiesis; production in fibroblasts [208] which itself stimulates
NNT induced growth of M1 macrophage cells whereas fibroblast mitosis and collagen production [468].
IL-6. LIF, OSM and CT-1 inhibited M1 growth and Transgenic mice that overproduce OSM have develop-
induced differentiation [1784]. mental abnormalities and visceral fibrosis [1209].
210 Cytokines

OSM has both stimulatory and regulatory effects on sition for bone calcification [82, 182, 413]. OPN is not
inflammation. It is increased in the synovial fluids of normally present in soft tissues, but it does accumulate
rheumatoid arthritis patients [817]. In a mouse model at sites of abnormal calcium deposition, such as athero-
for RA, OSM appeared to suppress inflammation and sclerotic lesions and calcified heart valves [542, 1875].
tissue damage [2092]. It stimulated the production of OSN appears to be controlling calcium deposition, for
acute phase proteins, which are known to have antiin- in OSN deficient mice, vascular calcification is exacer-
flammatory activity. [1642]. In fibroblasts, OSM inhib- bated [1867].
ited the production of IL-1-stimulated proinflammatory In bone marrow, OPN appears to control stem cell
cytokines IL-8 and GM-CSF [1643]. OSM also increases migration and the size of the stem cell pool to prevent
fibroblast production of tissue inhibitor of metallopro- excessive expansion [1456, 1893]. Osteopontin controls
tease (TIMP) [1644], which inhibits collagenase activ- the activation of T cells. It favors Th1 immune response
ity and tissue destruction. On the other hand, OSM has [79]. Mice deficient in OPN fail to develop Th1 immunity
been shown to have proinflammatory effects. When during viral and bacterial infection; production of IL-12
injected into joints, it caused cartilage resorption and and IFN-γ is decreased while IL-10 is increased [79].
inhibited proteoglycan production resulting in tissue Attenuation of experimental autoimmune encephalitis,
damage [127]. And OSM alone, and together with IL-1 a Th1-related disease, was observed in OPN deficient
and TNF-α, stimulates production of several matrix mice [881]. OPN appears to stimulate Th1 response at
metalloproteinases in cartilage [356, 1009]. the level of dendritic cells. OPN promotes migration of
As implied by its name, OSM has been shown to dendritic cells from tissue sites to lymphnodes; it
inhibit growth in several cancers including melanoma induces TNF and IL-12 (Th1) cytokines; and it increases
[207, 2274], myeloid leukemia [213], glioma cells [702] adhesion and costimulatory molecules [1635]. Though
and breast cancer [1137]. There is interest in OSM for it favors the Th1 pathway, OPN stimulates B cell prolif-
these cytostatic effects and for its role in immune eration and Ig production.
response. But OSM has not been tested clinically. It Osteopontin is involved in acute and chronic inflam-
should be noted, OSM is a growth factor for Kaposi’s mation. It has chemotactic, pro-inflammatory effects
sarcoma [1308]. Certain of its proinflammatory effects but it also has prominent regulatory functions. Elevated
might exacerbate infiltration and angiogenesis [1605] OPN levels have been observed in atherosclerosis [87],
glomerulonephritis [816] and in granulomatous diseases
[1430]. During heart failure, increased OPN levels are
observed in myocardial cells [1831]. In a mouse model
Osteopontin for rheumatoid arthritis, OPN appeared to cause joint
Pro Th1, Nitric Oxide Synthetase destruction by promoting angiogenesis and cartilage
cell apoptosis [2268]. However, OPN depresses nitric
Inhibiting Cytokine
oxide synthetase and NO production in macrophages
Osteopontin (OPN) is a secreted phosphoglycoprotein. [694, 1657], kidney epithelium [814] and endothelial
It is also referred to as early T lymphocyte activation cells [1771]. Inhibition of nitric oxide appears to be a
protein-1 (Eta-1). OPN is produced by activated T cells, major regulatory function of OPN limiting damage dur-
macrophages, osteoblasts, endothelial cells, brain and ing inflammation.
certain epithelial cells. Its expression is increased by In endothelial cells, OPN acts as a survival, cell adhe-
IL-1, IFN-γ TNF-α, bFGF, phorbol esters, glucocorti- sive and chemotactic factor. It organizes angiogenesis.
coids and 1,25 dihydroxyvitamin D3 [79]. OPN is also Fibroblast growth factor 2 (3, 1068) stimulates OPN
produced by transformed cells [1785]. production by endothelial cells. In turn OPN induces
There are two receptors for osteopontin, integrin monocyte chemotaxis. Tissue becomes infiltrated with
αVβ3 on endothelial cells, fibroblasts and other non- monocytes. OPN stimulates monocytes to produce
hematopoietic cells, and CD44 that is on leukocytes [79, TNF-α and IL-8, which are both angiogenic [1068].
694]. The synthetic peptide GRGDSP blocks integrin This mechanism could have a role in normal wound
binding and activation [694]. Integrin αVβ3, CD44 and repair or in pathology such as cancer.
osteopontin are cell surface extracellular matrix compo- Several properties of OPN may contribute to cancer
nents which function structurally and in signaling. cell growth and behavior. Variable osteopontin levels
OPN has an important role in bone growth and remod- are observed in cancer. Increased OPN secretion was
eling. OPN binds osteoclasts and functions in osteoclast associated with decreased nitric oxide production and
recruitment. OPN is a regulator of hydroxyapatite depo- decreased killing of tumor cells by macrophages and
Walter M. Lewko and Robert K. Oldham 211

endothelial cells. In this way, OPN may have a role in OX40L is capable of reverse signaling back into the
immune escape by cancer cells [413]. OPN also induces cells that express it [1511, 1908]. In intermediate stage
cyclooxygenase, and the progression of prostatic cancer dendritic cells, OX40L ligation, by interaction with T
[877]. OPN induces monocyte secretion of IL-1β [1421] cells or by agonist Mabs, caused reverse signaling to the
and other growth and proangiogenic factors. OPN is dendritic cell, enhancing maturation and the secretion of
expressed by normal brain and astrocytoma cells; it cytokines TNF-α, IL-12, IL-1β and IL-6 [1511].
increases astrocytoma cell migration [440]. Hepatocyte B cell-deficient mice produce a poor T cell response
growth factor activates a genetic program in epithelial [1141]. B cells function during antigen presentation;
cells for cell dissociation, growth and invasion. This they appear to stimulate T cells after their initial contact
program is upregulated during progression in many with DCs. OX40L has a role in this process. OX40L is
tumors. HGF induces osteopontin; antibody studies sug- expressed on splenic B cells. When B and T cells inter-
gest that OPN mediates HGF-induced invasive behavior act, OX40 engagement stimulates T cell growth and
[1280]. Melanoma produces OPN; in melanocytes OPN IL-2 release [1908, 1909]; reverse signaling through
functions as an antiapoptotic survival factor [608]. OX40L on the B cells induces B cell growth and
Osteopontin has been observed in many breast cancers. differentiation.
Its presence is often associated with calcification. OX40 ligand is found mainly on antigen presenting
Several studies have related osteopontin accumulation cells. However, OX40L is also present on CD4 T cells
in breast cancer with decreased survival [770, 1696, and it appears to have a role in T cell response [1862].
1834, 2036]. When activated in culture, T cells that were genetically
deficient in OX40L proliferated less than normal T cells
and when they were transferred back into mice, sur-
vival was reduced [1862]. It thus appears that T cell
OX40 ligand OX40L provides additional signals to sustain CD4 T
Costimulation in T Cell, B Cell cell longevity.
OX40 ligand has been shown to shut down IL-10 pro-
Development/Memory; Inflammation
ducing regulatory T cells. OX40 L inhibited the genera-
OX40 ligand (OX40L) is a member of the TNF family. tion of the regulatory cells and additionally it inhibited
It is a 32–34 kDa, 183 amino acid, membrane bound IL-10 production in differentiated IL-10 producing reg-
glycoprotein. OX40L is expressed mainly on dendritic ulatory T cells [865].
cells [632, 1511], B cells [1908], endothelium and acti- OX40L is expressed on vascular endothelial cells. It
vated T cells [117, 851, 1571]. may function during inflammation in the binding and
OX40 (CD134) is the receptor for OX40L. It is a extravasation of activated OX40-expressing T cells
48 kDa, 250 amino acid, membrane glycoprotein. OX40 [851]. The OX40–OX40L system appears to be involved
was originally described in CD4 T cells as a surface in several inflammatory diseases including allergic
antigen, which was similar to the nerve growth factor encephalomyelitis [1436, 2134], graft versus host dis-
receptor [1213, 1542]. It is found only on activated T ease [1909, 1910, 2000], inflammatory bowel disease
cells [232, 2000]. Normally, its levels are very low and [751] and rheumatoid arthritis [215]. In certain cases,
rise following activation [232, 655]. OX40 signaling amelioration of the disease has been demonstrated when
involves TRAF-2, TRAF-3 and NF-κB [62]. the animals were treated with anti-OX40 antibodies
The OX40L–OX40 system functions as a costimulator [751, 1910, 2134]. Exacerbation of inflammatory dis-
during antigen presentation for the activation and ease has been shown in transgenic animals overexpress-
increased longevity of T cells, in particular CD4+ Th ing OX40 [1436]. Tumor infiltrating lymphocytes were
cells. Mice deficient in OX40 signaling show reduced T also positive suggesting a role for OX40 in host response
cell response to infection. B cell response appears to be to cancer [472, 2073].
normal [294, 1006, 1396, 1571, 2000]. OX40L–OX40 There is interest in costimulatory cytokines as adju-
interactions occur in addition to B7-CD28 signaling vants during vaccination. OX40 ligation by agonistic
[111, 660, 386, 887, 2243] between AP and T cells. OX40L antibodies promoted memory with increased survival of
acts synergistically with B7 to costimulate CD4 Th cell specific T cells [655]. OX40 costimulation could even
expansion and the secretion of IL-2, IL-4 and IL-5 reverse established anergy [112]. It has been shown that
[655]. OX40L was required for priming [1436] and OX40 signaling by several means enhanced immune
sustained later stage CD4 T cell proliferation and response during cancer vaccination [27, 44, 379, 669,
memory [654, 655]. 836, 914, 1362, 1529] OX40 costimulation together
212 Cytokines

with GM-CSF was able to produce a strong response to OPG have altered B cells; transitional B cells are
the naturally occurring Her2/neu tumor antigen, capable increased and proB cells are more sensitive to IL-7
of inducing regression in established tumors [1397]. growth stimulation [2270]. RANKL is a morphogenesis
OX40L and OX40 are on HTLV-infected T cell leu- factor; it is involved in the early development of
kemias and the virus regulates expression. OX40L lymphnodes. Mice deficient in RANKL and RANK had
and OX40 may be involved in the development and abnormal lymphnode development and B cell production
growth of this cancer [117, 1321]. [416, 1003].
Estrogens tend to inhibit B cell lymphopoiesis during
pregnancy [970]. Estrogens stimulate stromal cell secre-
tion of factors that are inhibitory [1845]. Estrogens also
RANKL/TRANCE stimulate the production of OPG [778].
B Cell, DC, Osteoclast Development, RANKL stimulates osteoclast maturation and bone
development. Osteoprotegerin regulates osteoclast
Lymph Node Organogenesis
activity [1050, 1828, 2221]. Macrophages and osteo-
RANKL (receptor activator of NF-κB ligand) is a mem- clasts share a common lineage [517]. Peripheral blood
brane bound, TNF-related protein, It functions in the monocytes cultured with RANKL, G-CSF and TGF-β
development of osteoclasts, lymphocytes and in lymph formed osteoclasts [807, 913, 1793]. B cells may also be
node organogenesis [450, 1003]. RANKL is also involved in osteoclast development. B cells are a source
referred to as TRANCE (TNF-related activation-induced of stimulatory RANKL. B-lymphoid progenitor cells
cytokine), osteoclast differentiation factor [2235] and are also a source of osteoclast precursor cells [1215].
osteoprotegerin ligand. RANKL is expressed on T cells Mice lacking the RANKL and RANK have problems
[48, 903, 1003, 2193, 2194], B cells [2269], dendritic with bone and vascular development. The mice lack
cells [48, 2192] and bone marrow stromal/osteoblast osteoclasts and develop osteoporosis [218]. Mice defi-
cells [48, 1950, 2269]. In T cells RANKL levels increase cient in OPG are viable but osteoclast activity goes
with TCR activation [2106]. unchecked and as they age, the mice develop severe
RANK and osteoprotegerin (OPG) are receptors for osteoporosis [1328, 2270].
RANKL. They are membrane-bound proteins and While little has been done with the RANK/RANKL
members of the TNFR family. RANK is found on a system in cancer, there is interest in costimulatory mol-
variety of cell types, in particular, osteoclast precursors ecules that may enhance anticancer immune response. It
[1003] dendritic cells [48, 1003, 2193, 2221] and B has been shown that DNA vaccines that include the
cells [2269, 2270]. OPG is also referred to as follicular RANKL gene and dendritic cell vaccines that express
dendritic cell-derived receptor-1 and osteoclastogene- RANK/RANKL produce more potent T cell responses
sis inhibitory factor [218, 825, 2221, 2235]. OPG has [1322, 2155]. It may be that controlling OPG will be
membrane bound and soluble forms [2221, 2269]. OPG useful during immune therapy and vaccination [2270].
and RANK are both upregulated by CD40L, an impor-
tant cytokine in germinal center and B cell develop-
ment [2269]. RANK is the active receptor, which
induces response in target cells; OPG is a non-signaling
Stem cell factor
decoy receptor that competes with RANK [1813, 2270]. Early Progenitor and Mast
Inhibitory activity is mainly associated with soluble
form of OPG. The function of the bound form is not
Cell Growth Factor
certain. There is a report that it may be able to induce Stem cell factor (SCF) was discovered in rat liver as a
apoptosis [2221]. Other investigators were unable to protein responsible for the outgrowth of very early pro-
detect any signaling [2269]. genitor cells in bone marrow [2305]. SCF is also referred
RANKL has several functions in the immune system. to as kit ligand, steel factor and mast cell growth factor
In dendritic cells, it stimulates antigen presentation [48, [47, 345, 2006, 2169]. There are two forms of SCF,
903, 1003, 2192], Bcl-XL antiapoptotic activity and sur- soluble and membrane bound, produced by alternative
vival [2193]. OPG suppresses this [2270]. The dendritic splicing of the same pre-mRNA [47, 345, 2006, 2007].
cells from OPG-deficient mice are altered in that they Several types of cells express SCF, including bone mar-
are activated and show increased capacity to stimulate T row stroma, fibroblasts, liver and spleen [1244, 1248].
cells [2270]. RANKL stimulates normal B cell develop- The receptor for SCF turned out to be the product
ment. OPG also regulates this process. Mice lacking of the protooncogene c-kit [47, 345, 543, 2169, 2304].
Walter M. Lewko and Robert K. Oldham 213

This receptor is a protein tyrosine kinase [2219]. It is with progression in breast cancer [1426] and melanoma
found on stem cells, progenitor cells and mast cells [1428] reminiscent of the loss of estrogen receptor,
[1947, 2033, 2143]. A ligand-induced dimerization is which occurs with progression in breast cancer.
part of the receptor activation process. SCF induces SCF is being tested for its ability to synergize with
the downregulation of its own receptor by internal- G-CSF (filgrastim) to mobilize CD34+ progenitor cells
ization [2238]. into peripheral blood. It has been shown that SCF in
SCF stimulates growth of early progenitor cells combination with G-CSF may be useful to stimulate
(hematopoietic, lymphoid, and myeloid). Other cytok- hematopoietic recovery after chemotherapy [506, 2124].
ines induce differentiation and SCF may act synergisti- SCF with G-CSF also decreased the number of aphere-
cally with them [373, 2007, 2033]. SCF has a particularly ses needed to obtain progenitor cells for autologous
remarkable effect on mast cells. Mice genetically defi- transplantation [562].
cient for SCF suffer anemia and are very deficient in
tissue mast cells [609, 977, 978]. SCF acts on early
mast cell progenitors to stimulate outgrowth. IL-3 acts
at a later stage to stimulate further growth and antipara-
Tumor Necrosis Factor a
site activity [2266]. SCF depresses apoptosis and thus Inflammation, Immune Regulation,
serves as a mast cell survival factor [834]. rhuSCF
Apoptosis, Endothelial Damage
injected sc into patients activates mast cell degranula-
tion and proliferation with a wheal and flare response at TNF-α was first described in 1975 as a factor found in
the injection site [346]. animals treated with BCG or LPS. It was named for its
Mast cells and SCF may be involved in fibrosis. ability to induce hemorrhagic necrosis in tumors [250, 661,
Fibroblasts are a major source of SCF. Mast cells stim- 2174]. TNF-α was later found to be identical to cachectin,
ulated by fibroblast SCF release histamine and eotaxin, a factor responsible for metabolic wasting in patients with
the eosinophil chemokine [780]. SCF also stimulates advanced cancer or infection [150–153]. Human TNF-α
mast cell adhesion to fibronectin of fibroblasts and exists in both soluble and membrane bound forms [1549].
other cells [382]. Mast cells induce fibroblasts to The soluble 17 kDa protein is generated from the 26 kDa
produce collagen. Protracted inflammation produces membrane-bound form by a protease, TNF-α converting
fibrosis [322, 1105]. In this way SCF may be involved enzyme (TACE) [161, 1377]. TNF-α forms a trimer in
in the development of fibrotic reactions characteristic solution [75]. The soluble form is more potent and appears
of chronic inflammatory disease and tumors. In humans, to be responsible for most TNF-α bioactivity. The mem-
IL-4 depresses the growth and survival of mast cells by brane-bound form is also active; it binds and signals
down regulating the expression of c-kit [1824]. through its receptor and by reverse signaling back into its
γδ T cells appear to have a role in innate and adaptive parental cell [91, 714, 753].
response to viruses and other parasites [1782]. The pro- TNF-α is produced by a number of different types of
liferation of γδ T cells appears to depend on SCF. Mice activated cells including macrophages, T cells (CD4+
lacking SCF receptor lacked intestinal γδ T cells while Th1 and CD8+) [634] and B cells [2029, 2116], den-
αβ T cells were not affected [1054]. dritic cells [2293], neutrophils [2069], adipocytes
SCF and its receptor are produced by several different [954], keratinocytes [1135], mast cells [158, 644],
types of tumors [1244, 1427] and may stimulate growth. mammary epithelium [2049], colon epithelium [907],
Tumors that have tested positive for c-kit and for SCF pancreatic β cells [2220], osteoblasts [1330], astrocytes
include small cell lung carcinoma [748, 1701, 1781], [1077], neurons [1972] and steroid-producing adrenal
breast [1426], testicular [1907], uterine, cervical and cells [637]. Pathogen molecules such as LPS and bacte-
ovarian cancer [853] and melanoma [1428]. While it is rial DNA stimulate TNF production in macrophages
not unusual to have the same cell express both the [241, 1304, 1866, 1876]. TNF-α expression increases
cytokine and its receptor, SCF will usually influence with activation of T cell receptors [634] and B cell
tumor growth by a paracrine mechanism. In this way the receptors [2029].
normal fibroblast component of a tumor could serve as TNF-α production is well regulated. In macrophages,
the source of SCF for c-kit+ carcinoma cells. For therapy, for example, LPS induces TNF-α; then TNF induces
it is possible that SCF blockade may inhibit tumor growth IL-10 [1579, 2114] and IL-10 feeds back on TNF-α
or that presence of this cytokine or c-kit on tumor cells and other inflammatory cytokines [613, 802] in a regu-
may be used to target antibodies, drugs or toxins. latory loop. Release of TNF-α from macrophages is
Interestingly, loss of c-kit expression has been observed inhibited by glucocorticoids, progesterone [1309], and
214 Cytokines

by estrogens [288, 1614, 1796]. Glucocorticoids are MAPK and NFκB are also activated. Apoptosis is
well known antiinflammatory agents. Progesterone has induced by a caspase protease cascade, starting with
some glucocorticoid activity; antiinflammatory effects caspase-8. The caspases clip and activate a series of
may be beneficial during pregnancy. enzymes and other proteins, producing the morphol-
There are at least three receptors for TNF-α. TNFRI ogy and DNA fragmentation observed in apoptotic
(55 kDa) and TNFRII (75 kDa) bind both TNF-α and cells [75, 2094]. TNF-α also induces Fas expression in
LT-α. A third receptor, described in liver, binds TNF-α cells such as CD4+ T cells, another way in which it
but not LT-α [1760]. Nearly all mammalian cells express causes cell death by apoptosis [2292].
TNFRI [659]. It appears to be responsible for most TNF-α not only induces apoptosis but it also regu-
TNF-α effects [2094]. The cytoplasmic part of TNFRI lates it. Activation of NF-κB by TNF-α induces anti-
contains a death domain that is involved in apoptosis. apoptotic survival factors that interfere with caspase
TNF-α signaling also involves activation of NF-κB and activation (691). TNF signaling induces death and
p38 MAP kinase pathways, which induce inflammation NF-κB, independently. Sensitivity to TNF-induced
and other non-apoptosis responses [2066]. apoptosis was enhanced in cells that were not able to
TNFRII preferentially binds membrane bound TNF-α activate NF-κB. Thus TNF-α stimulates apoptosis and
[665]. TNFRII lacks a death domain though clearly it later suppresses it [2050, 2099].
stimulates apoptosis [1976, 2288]. It seems that TNFRII TNF-α influences many processes in immune
does not induce apoptosis directly but rather it cooper- response. In dendritic cells, TNF stimulates maturation,
ates with TNFRI; a complex is formed which induces activation and antigen presentation for the induction of
apoptosis (397). Mice lacking TNFRII appeared to specific T cells [1648]. TNF stimulates IL-12 secretion
develop normally but were less sensitive to TNF-α and in macrophages and other cells; IL-12 enhances T cell
its toxicity [495]. development. In T cells, TNF-α and IL-1 together induce
TNF receptors I and II are shed from cells and found Th1 development and secretion IFN-γ [1809]. In many
in tissue fluids [493, 1060]. The soluble receptors are target cells, TNF-α and IFN-γ stimulate MHCI expres-
generated by the protease TNF receptor releasing enzyme sion to enhance antigen presentation and susceptibility to
[659, 738, 925, 1389, 1464, 1844]. Production of shed CTL-induced killing [1515]. TNF-α is a major stimula-
TNFR is specific and regulated. Activation by LPS or by tor of outgrowth in γδ T cells. This response was corre-
TNF-α itself induces TNFR shedding from many types lated with TNFRII levels [1053]. TNF-α has a major part
of cells [131, 344, 1083, 1587, 1889]. In patients with in the control of certain infections. For example, TNF-α
inflamed livers, soluble TNFR levels were correlated works together with γ-IFN and IL-4 on macrophages to
with disease severity [2184]. Shed receptors bind TNF-α control tuberculosis [545]. In part, this is due to the
and act as competitive antagonists. The soluble receptor induction of nitric oxide synthetase [126, 1729]. TNF-α
thus has an antiinflammatory effect [892, 1889]. IL-10, also induces apoptosis in the mycobacteria-laden mac-
an antiinflammatory and immunosuppressive cytokine, rophages, sequestering the pathogens in apoptotic bodies
stimulates the release of soluble TNFR from monocytes [140, 457]. In neutrophils, TNF-α increased complement
as part of its mechanism of action [905]. receptor CD11b, adhesion to endothelium, release of
TNF stimulates the secretion of a number of cytokines reactive oxygen, degranulation, phagocytosis and ADCC
including TNF-α itself, IL-1, IL-6, IL-8, IFN-γ, GM-CSF, [586, 983, 1429, 1792]. TNF-α also increased cellular
M-CSF, PDGF, IL-10 and NGF. Directly or indirectly, it leukocyte adhesion molecules such as ICAM-1, VCAM-1
increases the expression of several growth factor recep- and E-Selectin in endothelium [1193], renal tubule epi-
tors, adhesion molecules, collagenases and plasminogen thelium [2209] and in liver [2184]. These adhesion mol-
activator. TNF-α and IL-1 are key proinflammatory ecules function during tissue infiltration to direct effector
cytokines. They share several biological functions [150, cell migration and activity.
152, 1498] even though they are not related molecularly In capillary endothelium, TNFα is angiogenic and
nor do they share receptors. It appears that both cytok- functions in wound repair [1084]. TNF stimulates fibro-
ines, on binding their receptors, activate several down- blast growth and enzyme secretion associated with ves-
stream protein kinases in common [691, 698]. sel formation [698]. However TNF-α also causes
TNF-α has a remarkable ability to induce death by damage and it is believed to be responsible for vascular
apoptosis in many different types of cells for its role in leak syndrome [458, 1607].
the regulation of morphogenesis, inflammation and TNF-α is involved in bone metabolism. TNF-α is
immune response [1726]. When TNF binds RI and produced by osteoblasts; it stimulates osteoblast mitosis
RII, death domains are activated [1965, 2094]. p38 [1330] and bone resorption by osteoclasts [1988].
Walter M. Lewko and Robert K. Oldham 215

TNF-α is produced by lipocytes and it may have a GM-CSF and TNF-α and then pulsed with specific
role in obesity. TNF-α mRNA levels are increased in the tumor antigens prior to inoculating mice [262, 1259].
lipocytes of obese patients; levels decrease with weight TNF-α generally inhibits tumor growth, however,
loss [954]. there are cases in which it stimulates growth. TNF
TNF-α is involved in inflammatory and autoimmune appears to be an autocrine and paracrine growth factor
diseases. Most of the deleterious TNF effects appear to for certain ovarian cancers. TNF-α induced its own
be mediated by TNFRI [37, 1685]. In septic shock, LPS production. IL-1 also stimulated TNF-α levels and
and bacterial DNA act synergistically to stimulate ovarian cancer growth [2207]. In the B16 mouse mela-
TNF-α production [590]. Mice treated with recombi- noma model, TNF-α stimulated tumor metastasis to
nant soluble TNF receptor are protected from toxicity lung. The induction of VCAM-1 by TNF-α may have
and death [1100]. Collagen-induced arthritis was atten- been responsible [1486].
uated in mice treated with antibodies to TNFRI and in TNF-α causes thrombocytopenia. Studies in mice
mice genetically deficient for TNFRI [1355, 1991]. A showed this depended on TNFRI. Since platelets did not
protease inhibitor which blocks both TACE and matrix have the TNFR, TNF-α appeared to be stimulating
metalloproteases (induced by TNF) produced good platelet activation and consumption indirectly, possibly
responses in rat arthritis models [343]. Antibodies to by increasing thrombin, plasmin or 5-hydroxytryptam-
TNF-α (Infliximab, Etanercept) have been approved ine which are platelet agonists [1941].
for use in patients with rheumatoid arthritis and are Clinical studies with tumor necrosis factor in cancer
being tested in patients with Crohn’s disease and pso- patients have been extensive. Unfortunately, the results
riasis [292, 1203, 1454, 1721]. Interestingly, in experi- are disappointing. TNF-α has many functions, perhaps
mental diabetes and in certain other inflammatory too many to be useful clinically. At effective doses,
diseases, TNF-α appears to have a two-part influence. TNF-α causes substantial multiorgan toxicity, hypoten-
TNF-α expressed early was required for disease pro- sion and flu-like symptoms [1318]. As a single agent, it
gression. With prolonged exposure, TNF-α suppressed has not shown much antitumor effect [220, 522, 1836,
inflammatory and autoimmune responses [315, 642, 2149]. By administering TNF-α in isolated limb perfu-
1609]. Certain autoimmune diseases were exacerbated sion [559, 1128, 1496], in organ perfusion [25] and
in mice lacking TNF and TNFR. The antiinflammatory intravesicularly (bladder cancer) [1786], higher doses
effects of long term TNF appear to be due to the sup- may be used while containing the toxicity. In these set-
pression of T cell growth and cytokine secretion, tings, TNF-α has been given in combination with INFγ
associated with downregulation of the T cell receptor and chemotherapy (e.g. melphalan). This largely avoids
[668, 730, 870, 1864]. the systemic toxicities. More substantial effects have
TNF inhibits tumor growth. It does this several ways. been observed. Unfortunately, TNF-α administered
In culture, TNF has a direct effect on many tumor cell intrapleurally and intraperitoneally have not been very
lines [250, 1921]. The addition of IFN-γ often has a syn- useful in the control of malignant effusions [771, 1877].
ergistic effect [563, 2174]. In animals, TNF acts sys- Current strategies with TNF-α emphasize in vitro use to
temically to bring about tumor regression [250]. TNF produce dendritic cells and TNF gene transfected tumor
may act directly on the tumor cells, but its most remark- cells for vaccines. The TNF gene is also being inserted
able effect is on tumor’s vascular endothelium [1435]. into activated lymphocytes to exploit the capacity for
Vessels break down and clotting occurs, shutting off the tumor-specific T cells to home into the target site where
tumor’s blood supply. TNF also stimulates the immune TNF is released locally.
system and upregulates surface antigens involved in Recombinant soluble TNF receptor and antibodies to
tumor cell recognition. TNF and IL-2 synergistically TNF are being tested for control of TNF-related toxici-
increased cytotoxicity in LAK cells [1517], NK cells ties during immunotherapy. In one study, soluble recep-
[1513] and TIL [1118, 2047, 2111]. tor was administered in combination with IL-2.
TNF may be useful as an adjuvant in tumor vaccines. IL-2-related toxicity (due to TNF release) was modu-
In animal models, tumor cells engineered to produce lated while the IL-2 antitumor response was preserved
TNF induced immune response [30, 162, 775]. The [2020]. Thalidomide, the infamous sedative and terato-
engineered cells were not as tumorigenic; animals with gen [1291], is once again being prescribed as an immu-
regressed tumors were immune to subsequent tumor nomodulatory agent and it is being tested in cancer
challenge. Further, TNF-α has been used to produce patients. Thalidomide suppresses TNF-α production,
dendritic cells for immunization. Dendritic cells pre- inflammation and certain cell surface adhesion molecules
pared from bone marrow were grown in culture with [1356, 1462, 1719]. It is also antiangiogenic [953].
216 Cytokines

TRAIL that stimulate dendritic cell maturation [1689] and serve


as antigen for immune response [20, 779].
Apoptosis in Neoplastic Cells; TRAIL regulates inflammation and autoimmune
Regulation of Inflammation disease. This has been studied in mouse models for
autoimmune arthritis [1856] and encephalomyelitis
Tumor necrosis factor-related apoptosis-inducing ligand [757]. Blockade using soluble receptor to TRAIL exac-
(TRAIL, also called apo-2 ligand) is a 281 amino acid erbated these diseases while TRAIL expression dimin-
transmembrane protein and a member of the tumor ished them. TRAIL apparently decreases the activation
necrosis factor family [1576, 2166]. Most leukocytes of autoimmune T cells.
and certain tumor cells express TRAIL [2166]. In T The anticancer effects of TRAIL are remarkable but
cells, TRAIL is induced by activation and by IFN-α/β complex. Mice with genetic TRAIL deficiencies had
[939, 1776]. Cyclosporin and glucocorticoids inhibit its increased risk for hematological malignancies [2276] and
expression [1406, 2107]. methylcholanthrene-induced sarcomas [352]. TRAIL-
There are at least five receptors for TRAIL; TRAIL-R1 induces apoptosis in most melanoma cell lines [1985,
(DR4) [1528], TRAIL-R2 (DR5) [1555, 1751, 1772, 2283]. In human colon and breast cancers growing in
2089], TRAIL-R3 (DcR1) [404, 1341, 1805], TRAIL-R4 nude mice, activation of TRAIL receptors induced apop-
(DcR2) [403, 1232, 1527] and osteoprotegerin [490]. tosis and inhibited growth [81, 319, 2090, 2106]. These
TRAIL receptors are found on many cells, often together studies show that TRAIL and TRAIL R1 and R2 may
with TRAIL. TRAIL-R1 and TRAIL-R2 contain cyto- have therapeutic potential in the treatment of cancer. But
plasmic death domains and induce apoptosis in cells, not all cancers respond. And in certain tumors TRAIL
mainly neoplastic cells. TRAIL may induce apoptosis or induces NF-κB, which promotes survival and growth
regulate cell growth in normal T cells, mast cells and [480]. Drugs that inhibited NF-κB signaling sensitized
hepatocytes [138, 401, 888, 896, 1856]. cells to apoptosis and increased anticancer response to
By comparison with TNF-α and FasL, TRAIL has TRAIL [104, 1140, 1172, 2098].
little normal tissue toxicity [81, 19]. Two possible Clinical trials have been initiated in cancer patients
explanations for this lack of toxicity are competitive using apoptosis-inducing TRAIL receptor antibodies
antagonism and induction of antiapoptotic proteins. [1580, 2001].
TRAIL-R3 and TRAIL-R4 bind TRAIL but do not
induce apoptosis. They lack cytoplasmic death domains
and thus serve as decoy receptors [80, 1227, 1555]. In a
similar way, osteoprotegerin is a soluble receptor that TWEAK
binds TRAIL and competitively antagonizes its apop- Cell Growth, Survival;
totic activity [490]. Cancer cells may produce OPG and
for these tumors OPG can function as a paracrine
Angiogenesis; Inflammation
survival factor [785, 1809]. TWEAK (tumor necrosis factor-like weak inducer of
Normal cells and TRAIL-resistant tumor cells gener- apoptosis) is a member of the TNF cytokine family. It is
ally express antiapoptotic proteins such as FLIP [671, expressed in many types of cells. Tweak is synthesized
856, 2283] and IAP [403, 404, 420, 560, 2100]. These as a membrane-bound protein that is cleaved to release
interfere with caspase activation and function as sur- a soluble, active, trimeric cytokine [304]. The TWEAK
vival factors. Drugs that control survival factors potenti- receptor is Fn14. It is a TNF receptor family member.
ate anticancer effects of TRAIL [1140]. Fn14 was discovered as a cell surface protein that was
TRAIL is responsible for cell contact-induced tumori- induced by fibroblast growth factor [523, 2164]. Fn14 is
cidal activities of CD4 T cells [1985], NK cells [939], upregulated in injured blood vessels [2164] and in
monocytes [672] and dendritic cells [289, 510]. In CD4 regenerating liver [523]. The cytoplasmic tail binds and
cells, for example, studies using blocking and activating signals through TNF associated factors-1, 2, 3 and 5
antibodies showed that TRAIL-induced apoptosis is one [206, 2164]. TWEAK activates NFκB, which translo-
of the ways cytotoxic CD4 T cells kill melanoma [1985]. cates to the nucleus to induce genes; TWEAK activates
The upregulation of TRAIL on T cells may be respon- caspases 3 and 8 in those cells that undergo Tweak-
sible for certain antitumor effects of interferons α and β induced apoptotic death [206, 2019].
[939]. Dendritic cells can kill tumor cells directly by In a cultured monocyte line, TWEAK induced the
TRAIL-induced apoptosis. In addition to decreased cells to differentiate into osteoclasts. Fn14 was not the
tumor burden, DC-TRAIL provides apoptotic bodies receptor for this differentiation since the cell line lacks
Walter M. Lewko and Robert K. Oldham 217

Fn14 and inactivating mabs to Fn14 did not block the 6. Adorini L, Guery JC, Trembleau S. Manipulation of the Th1/Th2
cell balance: an approach to treat human autoimmune diseases?
Tweak effect. This study suggested the existence of a Autoimmunity 1996; 23: 53.
second TWEAK receptor [1583]. It was recently reported 7. Agematsu K, Kobata T, Sugita K, et al. Direct cellular communica-
that CD163 is an additional binding protein for TWEAK. tions between CD45RO and CD45RA T cell subsets via CD27/
CD70. J Immunol 1995; 154: 3627–3635.
It is present on monocytes and macrophages where 8. Agematsu K, Kobata T, Sugita K, et al. Role of CD27 in T cell
it is known to function as a scavenger for hemoglobin. immune response: analysis by recombinant soluble CD27. J
CD163 has been shown to compete with Fn14 for Immunol 1994; 153: 1421.
9. Aggarwal BB, Hanzel WJ, Moffat B, Kohr WJ, Harkins RN.
TWEAK; may serve as an alternative receptor [185]. Primary structure of human lymphotoxin derived from 1788
TWEAK is a remarkably proinflammatory cytokine. It lymphoblastoid cell line. J Biol Chem 1985; 260: 2334–2344.
induces the expression of factors such as IL-6, IL-8, 10. Aggarwal S, Ghilardi N, Xie MH, et al. Interleukin-23 promotes a
distinct CD4 T cell activation state characterized by production of
RANTES, MCP-1, IP-10 and ICAM-1 in a number of Interleukin-17. J Biol Chem 2003; 278: 1910–1914.
cell types including fibroblasts [305], keratinocytes [891], 11. Aggarwal S, Xie MH, Maruoka M, et al. Acinar cell of pancreas
synoviocytes [305], bronchial epithelial cells [2216], are a target of interleukin-22. J Interferon Cyt Res 2001; 21: 1047.
12. Agostini C, Trentin L, Facco M, et al. Role of IL-15, IL-2 and their
macrophages [967], mesangial cells [237], astrocytes receptors in the development of T cell alveolitis in pulmonary
[1702] and endothelial cells [713]. Typically, the addition sarcoidosis. J Immunol 1996; 157: 910.
13. Airoldi I, Giri G, Marshall JD, et al. Expression and function of
of IFN-γ has a synergistic effect. TWEAK levels are ele- IL-12 and IL-18 receptors on human tonsillar B cells. J Immunol
vated in tissues of mice with experimental autoimmune 2000; 165: 6880–6888.
encephalitis [417] and collagen-induced arthritis [1556]. 14. Akahoski T, Oppenheim JJ, and Matsushima K. Induction of high
affinity interleukin 1 receptor on human peripheral blood lympho-
TWEAK is angiogenic. It induces mitosis and migra- cytes by glucocorticosteroid hormones. J Exp Med 1988; 167:
tion in human endothelial and aortic smooth muscle 924–936.
cells. FGF-2 and VEGF, which are also angiogenic, 15. Akashi K, Kondo M, Von Freeden-Jeffry R, Murray R, Weissman
IL. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor defi-
stimulate TWEAK receptor levels. TWEAK induced cient mice. Cell 1997; 89: 1033.
strong vessel growth in the rat cornea angiogenesis 16. Akashi K. Role of interleukin 4 in the negative regulation of leuke-
assay [449, 713, 1180, 2164]. mia cell growth. Leukemia and Lymphoma 1993; 9: 205–209.
17. Al Jabaari B, Ladyman HM, Larche M, et al. Elevated expression
TWEAK appears to be involved in neoplastic pro- of interleukin 4 receptor in carcinoma. A target for immunotherapy?
gression. The Fn14 gene is rapidly induced during liver British J Cancer 1989; 59: 910–914.
regeneration; Fn14 is overexpressed in hepatocellular 18. Albanesi C, Scarponi C, Cavani A, et al. Interleukin 17 is produced
by both Th1 and Th2 lymphocytes, modulates interferon-γ and
carcinomas [523]. When human embryonic kidney cells interleukin-4-induced activation of human keratinocytes. J Invest
were transfected to express TWEAK, these cells formed Dermatol 2000; 115: 81.
larger and more vascular tumors in nude mice [774]. 19. Albanesi C, Scarponi C, Sebastiani S, et al. IL-4 enhances kerati-
nocyte expression of CXCR3 agonistic chemokines. J Immunol
Glioblastoma specimens and glioma cells in culture 2000; 165: 1395–1402.
overexpress Fn14. And when glioma cells were treated 20. Albert ML, Sauter B, BharDwar N. Dendritic cells acquire antigen
from apoptotic cells and induce class I-restricted CTLs. Nature
with TWEAK, the cells became resistant to apoptosis- 1998; 392: 86.
based therapy. Tweak activates the NFκB path for 21. Alderson MR, Armitage RJ, Tough TW, et al. CD40 expression by
growth and survival. All told, blocking Fn14 may be of human monocytes: regulation by cytokines and activation of
monocytes by the ligation for CD40. J Exp Med 1993; 178: 669.
benefit in the management of glioblastoma and other 22. Alderson MR, Sassenfeld HM, Widmer MB. Interleukin 7 enhances
TWEAK/Fn14 expressing cancers [2018]. cytotoxic T lymphocyte generation and induces lymphokine
activated killer cells from peripheral blood. J Exp Med 1990; 172:
577–587.
23. Alderson MR, Smith CA, Tough TW, et al. Molecular and biologi-
References cal characterization of human 4-1BB and its ligand. Eur J Immunol
1994; 24: 2219–2227.
1. Aarvak T, Chaubaud M, Miossec P, Natvig JB. IL-17 is produced by 24. Alderson MR, Tough TW, Ziegler SF, Grabstein KW. Interleukin 7
some proinflammatory Th1/Th0 cells but not by Th2 cells. J induces cytokine secretion and tumoricidal activity by human
Immunol 2000; 162: 1246–1251. peripheral blood monocytes. J Exp Med 1991; 173: 923–930.
2. Abehsira-Amar O, Gilbert M, Joliy M, et al. IL-4 plays a dominant 25. Alexander HR, Bartlett DL, Libutti SK, et al. Isolated hepatic per-
role in the differential development of Th0 into Th1 and Th2 cells. J fusion with tumor necrosis factor and melphalan for unresectable
Immunol 1992; 148: 3820–3829. cancers confined to the liver. J Clin Oncol 1998; 16: 1479–1486.
3. Abraham JA, Mergia A, Whang JL, et al. Nucleotide sequence of a 26. Alexander HR, Wong GGH, Doherty GM, et al. Differentiation
bovine clone encoding the angiogenic protein, basic fibroblast factor/leukemia inhibitory factor protection against lethal endotox-
growth factor. Science 1986; 233: 545. emia in mice: synergistic effect with interleukin-1 and tumor
4. Acres B, Gantzer M, Remy C, et al. Fusokine interleukin-2/interleu- necrosis factor. J Exp Med 1992; 175: 1139.
kin-18, a novel potent innate and adaptive immune stimulator with 27. Ali S, Ahmad M, Lynam J, et al. Anti-tumor efficacy of OX40L in
decreased toxicity. Cancer Res 2005; 65: 9536–9546. murine tumor model. Vaccine 2004; 22: 3585–3594.
5. Addison CL, Daniel TO, Burdick MD, et al. The CXC chemokine 28. Allakhverdi Z, Smith DE, Comeau MR, et al. The ST2 ligand
receptor 2, CXCR2, is the putative receptor for ELR+ CXC chemokine- IL-33 potently activates and drives maturation of human mast
induced angiogenic activity. J Immunol 2000; 165: 5269–5277. cells. J Immunol 2007; 2051–2054.
218 Cytokines

29. Allevena P, Giardina G, Bianchi G, Mantovani A. IL-15 is chemot- 49. Andoh A, Zhang Z, Inatomi O, et al. Interleukin-22, a member of
actic for natural killer cells and stimulates their adhesion to vascu- the IL-10 subfamily induces inflammatory responses in colonic sub-
lar endothelial cells. J Leukocyte Biol 1997; 61: 729. epithelial myofibroblasts. Gastroenterology 2005; 129: 969–984.
30. Allione A, Consalvo M, Nanni P, et al. Immunizing and curative 50. Andrews JVR, Schoof DD, Bertagnolli MM, et al. Immunomodulatory
potential of replicating and nonreplicating murine mammary effects of Interleukin 12 on human tumor infiltrating lymphocytes. J
adenocarcinoma cells engineered with interleukin (IL)-2, IL-4, Immunotherapy 1993; 14: 1–10.
IL-6, IL-7, IL-10, Tumor Necrosis Factor and γ-interferon gene 51. Anegon I, Moreau J-F, Godard A, et al. Production of human inter-
or admixed with conventional adjuvants. Cancer Res 1994; 54: leukin for DA cells (HILDA)/leukemia inhibitory factor (LIF) by
6022–6026. activated monocytes. Cell Immunol 1990; 130: 50.
31. Allison J, Georgiou HM, Strasser A, Vaux DL. Transgenic expres- 52. Angiolillo AL, Sqadari C, Tosato G. A role for the interferon-in-
sion of CD95 ligand on islet β cells induces a granulocytic infiltra- ducible protein 10 in inhibition of angiogenesis by interleukin-12.
tion but does not confer immune privilege upon islet allograft. Proc Ann N Y Acad Sci 1996; 795: 158–167.
Natl Acad Sci USA 1997; 94: 3943. 53. Anguita J, Barthold SW, Samanta S, Ryan J, Fikrig E. Selective
32. Altenburg A, Baldus SE, Smola H, Pfister H, Hess S. CD40 ligand– anti-inflammatory action of interleukin-11 in murine Lyme dis-
CD40 interaction induces chemokines in cervical carcinoma cells ease: arthritis decreases while carditis persists. J Infect Dis
in synergism with IFN-γ. J Immunol 1999; 162: 4140–4147. 1999; 179: 734.
33. Alters SE, Gadea JR, Holm B, Lebkowski J, Philip R. IL-13 can 54. Ank N, West H, Bartholdy C, et al. Lambda interferon (IFN-λ), a
substitute for IL-4 in the generation of dendritic cells for the induc- type III interferon, is induced by viruses and IFNs and displays
tion of cytotoxic T lymphocytes and gene therapy. J Immunother potent antiviral activity against select virus infections in vivo. J
1999; 22: 229–236. Virol 2006; 80: 4501–4509.
34. Alves NL, Arosa FA, and van Lier RA. IL-21 sustains CD28 55. Annunziato F, Cosmi L, Santarlasci V, et al. Phenotypic and functional
expression on IL-15 activated human naive CD8+ T cells. J features of human Th17 cells. J Expl Med 2007; 204: 1849–1861.
Immunol 2005; 175: 755–762. 56. Antony PA, Restifo NP. CD4+ CD25+ T regulatory cells, immuno-
35. Alzona M, H-M Jack, Fisher RI, Ellis TM. CD30 defines a subset of therapy of cancer and interleukin-2. J Immunother 2005; 28:
activated human T cells that produce IFN-γ and IL-5 and exhibit 120–128.
enhanced B cell helper activity. J Immunol 1994; 153: 2861–2867. 57. Antonysamy MA, Fanslow WC, et al. Evidence for a role of IL-17
36. Amakawa R, Hakem A, Kundig KM, et al. Impaired negative in organ allograft rejection: IL-17 promotes functional differentia-
selection of T cells in Hodgkin’s disease antigen CD30 deficient tion of dendritic cell progenitors. J Immunol 1999; 162: 577–584.
mice. Cell 1996; 84: 551. 58. Aoki T, Kikuchi H, Miyatake S-I, et al. Interleukin 5 enhances
37. Amar S, Van Dyke TE, Eugster HP, Schultze N, Koebel P, interleukin 2 mediated lymphokine activated killer activity. J Exp
Bluethmann H. TNF-induced cutaneous necrosis is mediated by Med 1989; 23: 163–177.
tumor necrosis factor receptor R1. J Inflamm 1997; 47: 180. 59. Aoki T, Tashiro K, Miyatake S, et al. Expression of murine IL-7 in
38. Amaral MC, Miles CS, Kumar G, Nel AE. Oncostatin M stimu- a murine glioma cell line results in reduced tumorigenicity in vivo.
lates tyrosine protein phosphorylation in parallel with activation of Proc Natl Acad Sci USA 1992; 89: 3850–3894.
p42MAPK/ERK-2 in Kaposi’s cells: evidence that this pathway is 60. Aoki Y, Jaffee ES, Chang Y, et al. Angiogenesis and hematopoiesis
important in Kaposi’s cell growth. J Clin Invest 1993; 93: 848. induced by Kaposi’s sarcoma associated herpes virus-encoded
39. Ambrus JL and Fauci AS. Human B cell lymphoma cell line pro- interleukin-6. Blood 1999; 93: 4034.
ducing B cell growth factor. J Clin Invest 1985; 75: 732–730. 61. Arai H, Chan SY, Bishop DK, Nabel GJ. Induction of the alloanti-
40. Ambrus JL Chesky L, Stephany D, et al. Functional studies exam- body response by CD95 ligand. Nat Med 1997; 3: 843.
ining the subpopulation of human B lymphocytes responding to 62. Arch RH, Thompson CB. 4-1BB and Ox40 are members of a tumor
high molecular weight B cell growth factor. J Immunol 1990; 145: necrosis factor (TNF)-nerve growth factor subfamily that bind
3949–3955. TNF receptor-associated factors and activate nuclear factor κB.
41. Ambrus JL, Jurgenson CH, Broom EJ, Fauci AS. Purification to Mol Cell Biol 1998; 19: 558.
homology of a high molecular weight human B cell growth factor. 63. Arenberg DA, Keane MP, DiGiovine B, et al. Epithelial-neutrophil
J Exp Med 1985; 162: 1319–1335. activating peptide (ENA-78) is an important angiogenic factor in
42. Ambrus JL, Pippin J, Joseph A, et al. Identification of a cDNA for non-small cell lung carcinoma. J Clin Invest 1998; 102: 465.
a human high molecular weight B cell growth factor. Proc Nat 64. Arenberg DA, Kunkel SL, Polverini PJ, et al. Interferon-γ-inducible
Acad Sci USA 1993; 90: 6330–6334. protein 10 (IP-10) is an angiostatic factor that inhibits human non-
43. Amiel C, Darcissac E, Truong MJ, et al. Interleukin 16 (IL-16) small cell lung cancer (NSCLC) tumorigenesis and spontaneous
inhibits human immunodeficiency virus replication in cells from metastasis. J Exp Med 1996; 184: 981–992.
infected subjects and serum IL-16 levels drop with disease pro- 65. Arend WP, Malyak M, Smith Jr MF, et al. Binding of IL-1α, IL-1β
gression. J Infect Dis 1999; 179: 83–91. and IL-1 receptor antagonist by soluble IL-1 receptors and levels of
44. Andarini S, Kikuchi T, Nukiwa M, et al. Adenovirus vector medi- soluble IL-1 receptors in synovial fluids. J Immunol 1994; 153:
ated in vivo gene transfer of OX40 ligand to tumor cells enhances 4766–4774.
antitumor immunity to tumor-bearing hosts. Cancer Res 2004; 64: 66. Arens R, Schepers K, Nolte MA, et al. Tumor rejection induced by
3281–3287. CD70-mediated quantitative and qualitative effects on effector
45. Anders RA, Shubudhi SK, Wang J, et al. Contribution of the lym- CD8+ T cell formation. J Exp Med 2004; 199: 1595–1605.
photoxin β receptor to liver regeneration. J Immunol 2005; 175: 67. Arienti F, Belli F, Napolitano F, Sule-Suso J, et al. Vaccination of
1295–1300. melanoma patients with interleukin 4 gene-transduced allogeneic
46. Anderson DM, Kumaki S, Abdieh M, et al. Functional character- cells. Hum Gene Ther 1999; 10: 2907–2916.
ization of the human interleukin 15 receptor α chain and close 68. Ariizumi K, Kitajima T, Bergstresser PR, Takashima A. Interleukin-1
linkage of the IL-15RA and IL-2RA genes. J Biol Chem 1995; β converting enzyme in murine Langerhans cells and epidermal
270: 29862. derived dendritic cell lines. Eur J Immunol 1995; 25: 2137.
47. Anderson DM, Lyman S, Baird A, et al. Molecular cloning of mast 69. Ariizumi K, Meng Y, Bergstresser PR, Takashima A. INF-γ-
cell growth factor, a hematopoietin that is active in both membrane Dependent IL-7 gene regulation in keratinocytes. J Immunol 1995;
bound and soluble forms. Cell 1990; 63: 235–243. 154: 6031–6039.
48. Anderson DM, Marakovsky E, Billingsley WL, et al. A homolog of 70. Arima M, Plitt J, Stelatto C, et al. Expression of interleukin-16 by
the tumor necrosis factor and its ligand enhance T cells growth and human epithelial cells. Inhibition by dexamethasone. Am J Respir
dendritic cell function. Nature 1997; 390: 175. Cell Mol Biol 1999; 21: 684–692.
Walter M. Lewko and Robert K. Oldham 219

71. Armitage RJ, MacDuff BM, Eisenman J, et al. IL-15 has stimula- chemokine promoter activation in intestinal epithelial cells. J
tory activity in the induction of B cell proliferation and differen- Immunol 1999; 162: 5337.
tiation. J Immunol 1995; 154: 483–490. 94. Azimi N, Brown K, Bamford RN, et al. Human T cell lympho-
72. Armitage RJ, Macduff BM, Spriggs MK, Fanslow WC. Human tropic virus type I Tax protein trans-activates interleukin-15
B cell proliferation and Ig secretion induced by recombinant gene transcription through an NF-κB site. Proc Natl Acad Sci
CD40 ligand and modulated by soluble cytokines. J Immunol USA 1998; 95: 2452.
1993; 150: 3671. 95. Azimi N, Jacobsen S, Leist T, Waldmann TA. Involvement of
73. Armitage RJ, Namen AE, Sassenfeld HM, Grabstein KH. Regulation IL-15 in the pathogenesis of human T lymphotropic virus type
of T cell proliferation by IL-7. J Immunol 1990; 144: 938. I-associated myelopathy/tropical spastic paraparesis: implica-
74. Armitage RJ, Zeigler SF, Friend DJ, et al. Identification of a tions for therapy with a monoclonal antibody directed to the IL-2/
novel low affinity receptor for human interleukin-7. Blood 1992; IL-15 receptor. J Immunol 1999; 163: 4064.
7: 1738. 96. Azuma C, Tanabe T, Konishi M, et al. Cloning of cDNA for
75. Armitage RJ. Tumor necrosis factor receptor superfamily mem- human T-cell replacing factor (interleukin-5) and comparison with
bers and their ligands. Curr Opinion Immunol 1994; 6: 407–413. the murine homologue. Nucleic Acid Res 1986; 14: 9149–9158.
76. Aronson FR, Libby P, Brandon EP, et al. IL-2 rapidly induces 97. Bacon CM, McVicar DW, Ortaldo JR, et al. Interleukin-12
natural killer cell adhesion to human endothelial cells. A potential induces tyrosine phosphorylation of JAK2 and TYK2: differen-
mechanism for endothelial injury. J Immunol 1988; 141: 158. tial use of tyrosine kinases by IL-2 and IL-12. J Exp Med 1995;
77. Asadullah K, Docke WD, Ebeling M, et al. Interleukin 10 treat- 181: 399–404.
ment of psoriasis: clinical results of a phase 2 trial. Arch Dermatol 98. Bacon KB, Westwick J, Camp RDR. Potent and specific inhibi-
1999; 135: 187–192. tion of IL-8, IL-1α and β induced in vitro human lymphocyte
78. Asao H, Okuyama C, Kumaki S, et al. The common γ-chain is an migration by calcium channel antagonists. Biochem Biophys Res
indispensable subunit of the IL-21 receptor. J Immunol 2001; Commun 1989; 165: 349.
167: 1–5. 99. Badache A, Hynes NE. Interleukin 6 inhibits proliferation and, in
79. Ashkar S, Weber GF, Panoutsakopoulou V, et al. ETA-1: an early cooperation with epidermal growth factor receptor autocrine
component of type-1 immunity. Science 2000; 287: 860. loop, increases migration of T47D breast cancer cells. Cancer
80. Ashkenazi A, Dixit VM. Death receptors: Signaling and modula- Res 2001; 61: 383–391.
tion. Science 1998; 281: 1305. 100. Bagby GC Jr, Dinarello CA, Wallace P, et al. Interleukin 1 stimu-
81. Ashkenazi A, Pai RC, Fong S, et al. Safety and antitumor activity lates granulocyte macrophage colony-stimulating activity release
of recombinant Apo2 ligand. J Clin Invest 1999; 104: 155. by vascular endothelial cells. J Clin Invest 1985; 78: 1316.
82. Asou Y, Rittling H, Yoshitake K, et al. Osteopontin facilitates 101. Baggiolini M, Kernan P. Neutrophil activation: control of shape
angiogenesis, accumulation of osteoclasts, and resorption of change exocytosis and respiratory burst. News Physiol Sci
ectopic bone. Endocrinology 2001; 142: 1325–1332. 1992; 7: 215.
83. Assuma R, Oates T, Cochran D, Amar S, Graves DT. IL-1 and 102. Baier M, Bannert N, Werner A, Lang K, Kurth R. Molecular
TNF antagonists inhibit the inflammatory response and bone loss cloning, sequence, expression and processing of the interleukin
in experimental periodontitis. J Immunol 1998; 160: 403–409. 16 precursor. Proc Natl Acad Sci USA 1997; 94: 5273–5277.
84. Atkins MB, Robertson MJ, Gordon M, et al. Phase I evaluation 103. Baier M, Werner A, Bannert N, Metzner K, Kurth R. HIV sup-
of recombinant human interleukin 12 in patients with advanced pression by interleukin-16. Nature 1995; 378: 563.
malignancies. Clin Cancer Res 1997; 3: 409–417. 104. Bakker TR, Reed T, Renno T, Jongeneel CV. Efficient adenovirus
85. Atkins MB, Vachino G, Tilg HJ, et al. Phase I evaluation of transfer of NF-κB inhibitor sensitizes melanoma cells to tumor
thrice-daily intravenous bolus interleukin-4 in patients with necrosis factor-mediated apoptosis. Int J Cancer 1999; 80: 320.
refractory malignancy. J Clin Oncol 1992; 10: 1802–1809. 105. Balasa B, Krahl T, Patstone G, et al. CD40 ligand–CD40 ligation
86. Attia P, Maker AV, Haworth LR, et al. Inability of a fusion protein interactions are necessary for the initiation of insulinitis and dia-
of IL-2 and diphtheria toxin (Denileukin Diftitox, DAB389IL-2, betes in nonobese diabetic mice. J Immunol 1997; 159: 4260.
ONTAK) to eliminate regulatory T lymphocytes in patients with 106. Baluna R, Vitetta ES. Vascular leak syndrome: a side effect of
melanoma. J Immunother 2005; 28: 582–592. immunotherapy. Immunopharmacology 1997; 37: 117–132.
87. Attur MD, Dave MN, Clancey RM, et al. Functional genomic 107. Bamford RN, DeFilippis AP, Azimi N, Kurys G, Waldmann TA.
analysis in arthritis affected cartilage: yin-yang regulation of The 5′ untranslated region, signal peptide, and the coding
inflammatory mediators by integrins. J Immunol 2000; 164: 2684. sequence of the carboxyl terminus of IL-15 participate in its
88. Attur MG, Patel RN, Abramson SB, Amin AR. Interleukin-17 multifaceted translational control. J Immunol 1998; 160:
upregulation of nitric oxide production in human osteoarthritis 4418–4426.
cartilage. Arthritis Rheum 1997; 40: 1050. 108. Bamford RN, Grant AJ, Burton JD, et al. The interleukin (IL) 2β
89. Auron PE, Webb AC, Rosenwasser LJ, et al. Nucleotide sequence chain is shared by IL-2 and a cytokine, provisionally designated
of human monocyte interleukin 1 precursor cDNA. Proc Natl IL-T that stimulates T cell proliferation and the induction of lym-
Acad Sci USA 1984; 81: 7907. phokine activated killer cells. Proc Nat Acad Sci USA 1994; 91:
90. Avanzi GC, Brizzi MF, Gianotti J, et al. M-07e human leukemic 4940–4944.
factor-dependent cell line provides a rapid and sensitive bioassay 109. Banchereau J, Bazan D, Blanchard D, et al. The CD40 antigen
for the human cytokines GM-CSF and IL-3. J Cell Physiol 1990; and its ligand. Annual Rev Immunol 1994; 12: 881.
145: 458–464. 110. Banks TA, Richert S, Benedict CA, et al. A lymphotoxin-IFN-β
91. Aversa G, Punnonen J, de Vries JE. The 26 kDa transmembrane axis essential for lymphocyte survival revealed during cytomega-
form of tumor necrosis factor α on activated CD4+ T cell clones lovirus infection. J Immunol 2005; 174: 7217–7225.
provides a costimulatory signal for human B cell activation. J 111. Banks TA, Rouse BT, Kerley MK, et al. Lymphotoxin-α-deficient
Exp Med 1993; 177: 1575. mice: effects on secondary lymphoid organ development and
92. Avice MN, Demeure CE, Delespesse G, Rubio M, Armant M, humoral immune response. J Immunol 1995; 44: 1685.
Sarfati M. IL-15 promotes IL-15 production by human mono- 112. Bansal-Pakala P, Jember AG, Croft M, et al. Signaling through
cytes via T cell-dependent contact and may contribute to IL-12 OX40 (CD134) breaks peripheral T cell tolerance. Nat Med
mediated IFN-γ secretion by CD4+ T cells in the absence of TCR 2001; 7: 607–912.
ligation. J Immunol 1998; 161: 3408. 113. Barker BR, Parvani JG, Meyer D, et al. IL-21 induces apoptosis
93. Awane M, Andres PG, Li DJ, Reinecker HC. NF-κB-inducing of antigen-specific T lymphocytes. J Immunol 2007; 179:
kinase is a common mediator of IL-17-, TNF-α- and IL-1β-induced 3596–3603.
220 Cytokines

114. Battaile R, Barlogie B, Lu ZY, et al. Biologic effects of anti-inter- 136. Bennett SRM, Carbone FR, Karamalis F, et al. Help for cytotoxic
leukin-6 murine monoclonal antibody in advanced multiple T-cell responses is mediated by CD40 signaling. Nature 1998;
myeloma. Blood 1995; 86: 685–691. 393: 478.
115. Batten M, Li J, Yi S, et al. Interleukin 27 limits autoimmune 137. Benveniste EN, Tang LP, Law RM. Differential regulation of
encephalitis by suppressing the development of interleukin astrocyte TNF-α expression by cytokines TGF-β, IL-6, and
17-producing T cells. Nat Immunol 2006; 7: 929–936. IL-10. Int J Dev Neurosci 1995; 13: 341.
116. Bauer JH, Liu KD, You Y, Lai SY, Goldsmith MA. 138. Berent-Maoz B, Philiponsky AM, Daigle I, et al. Human mast
Heteromerization of the γ c chain with the interleukin-9 receptor cells undergo TRAIL-induced apoptosis. J Immunol 2006; 176:
α subunit leads to STAT activation and prevention of apoptosis. J 2272–2278.
Biol Chem 1998; 273: 9255–9260. 139. Berkman N, Robichaud A, Robbins RA, et al. Inhibition of
117. Baum PR, Gayle RB III, Ramsdell RF, et al. Molecular character- inducible nitric oxide synthase expression by interleukin-4 and
ization of murine and human OX40/OX40 ligand systems: iden- interleukin-13 in human lung epithelial cells. Immunol 1996;
tification of a human OX40 ligand as the HTLV-1-regulated 89: 363.
protein gp34. EMBO J 1994; 13: 3992. 140. Bermudez LE, Parker A, Petrofsky M. Apoptosis of
118. Baxevanis CN, Gritzapis AD, Papamichail M. In vivo antitumor Mycobacterium avium-infected macrophages is mediated by
activity of NKT cells activated by the combination of IL-12 and both tumor necrosis factor (TNF) and Fas, and involves the
IL-18. J Immunol 2003; 171: 2953–2959. activation of caspases. Clin Exp Immunol 1999; 116: 94.
119. Bazan JF, Bacon CB, Hardiman G, et al. A new class of membrane- 141. Bernstein HB, Plasterer MC, Schiff SE, et al. CD4 expression on
bound chemokine with CX3C motif. Nature 1997; 385: 640. NK cells: ligation of CD4 induces cytokine expression and cell
120. Bazan JF, Timans JC, Kastelein RA. A newly defined interleu- migration. J Immunol 2006; 177: 3669–3676.
kin-1? Nature 1996; 379: 591. 142. Berro AI, Perry GA, Agarwal DK. Increased expression and acti-
121. Beatty PR, Krams SM, Martinez OM. Involvement of IL-10 in vation of CD30 induce apoptosis in human eosinophils. J
the autonomous growth of EBV-transformed B cell lines. J Immunol 2004; 173: 2173–2183.
Immunol 1997; 158: 4045–4051. 143. Bertagnolli M and Herrmann S. IL-7 supports the generation of
122. Beckman MP, Cosman D, Fanslow W, et al. The interleukin 4 cytotoxic T lymphocytes from thymocytes. J Immunol 1990;
receptor: structure function and signal transduction. Chem 145: 1706–1712.
Immunol 1992; 51: 107. 144. Bertini R, Bianchi M, Ghezzi P. Adrenalectomy sensitizes mice
123. Bedard M, Mclure CD, Schiller NL, et al. Release of interleukin to the lethal effects of interleukin 1 and tumor necrosis factor. J
8, interleukin 6, and colony stimulating factors by upper airway Exp Med 1988; 167: 1708.
epithelial cells: implications for cystic fibrosis. Am J Respir Cell 145. Bertoglio S, Melioli G, Baldini E, et al. Intraperitoneal infusion
Mol Biol 1993; 9: 455. of recombinant interleukin-2 in malignant ascites in patients with
124. Behrens CK, Igney FH, Arnold B, Moller P, Krammer PH. CD95 gastrointestinal and ovarian cancer. Acta Med Austriaca 1989;
ligand-expressing tumors are rejected in anti-tumor TCR trans- 16: 81–83.
genic perforin knockout mice. J Immunol 2001; 166: 3240–3247. 146. Bertram EM, Dawicki W, Sedgmen B, et al. A switch in costimu-
125. Bejarano MT, de Waal Malefyt R, Abrams JS, et al. Interleukin lation from CD28 to 4-1BB during primary versus secondary
10 inhibits allogeneic proliferative and cytotoxic T cell responses CD8 cell response to influenza in vivo. J Immunol 2004; 172:
generated in primary mixed lymphocyte culture. Int Immunol 981–988.
1992; 4: 1389. 147. Bertram EM, Lau P, Watts TH. Temporal segregation of 4-1BB
126. Bekker L-G, Freeman S, Murray PJ, Ryffel B, Kaplan G. TNF-α versus CD28-mediated costimulation: 4-1BB ligand influences T
controls intracellular mycobacterial growth by both inducible nitric cell numbers late in primary response and regulates the size of the
oxide synthetase dependent and inducible nitric oxide synthetase- T cell memory response following influenza infection. J Immunol
independent pathways. J Immunol 2001; 166: 6728–6734. 2002; 168: 3777–3785.
127. Bell MC, Carrol GJ, Chapman HM, Mills JN, Hui W. Oncostatin 148. Besedovsky H, del Rey A, Sorkin E, Dinarello CA.
M induces leukocyte infiltration and cartilage proteoglycan deg- Immunoregulatory feedback between interleukin-1 and gluco-
radation in vivo in goat joints. Arth Rheu 1999; 42: 2543. corticoid hormones. Science 1986; 233: 652–654.
128. Belladonna ML, Renauld JC, Bianchi R, et al. IL-23 and IL-12 149. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental
have overlapping but distinct effects on murine dendritic cells. J pathways for the generation of pathogenic effector TH17 and
Immunol 2002; 168: 5448–5454. regulatory T cells. Nature 2006; 441: 235–238.
129. Bellone G, Carbone A, Smire C, et al. Cooperative induction of a 150. Beutler B, Cerami A. The biology of cachectin/TNF: a primary
tolerogenic dendritic cells phenotype by cytokines secreted by mediator of the host immune response. Annual Rev Immunol
pancreatic carcinoma cells. J Immunol 2006; 177: 3448–3460. 1989; 7: 625.
130. Bellone G, Trinchieri G. Dual stimulatory and inhibitory effect of 151. Beutler B, Greenwald D, Hulmes JD, et al. Identity of tumour
NK cell stimulatory factor/IL-12 on human hematopoiesis. J necrosis factor and the macrophage secreted factor cachectin.
Immunol 1994; 153: 930–937. Nature 1985; 316: 552–554.
131. Bemelmans MH, Abramowicz D, Gouma DJ, Goldman M, 152. Beutler B, Mahoney J, Le Trang N, et al. Purification of cachec-
Buurman WA. In vivo T cell activation by anti-CD3 monoclonal tin, a lipoprotein lipase-suppressing hormone secreted by endo-
antibody induces soluble TNF receptor release in mice: effects of toxin-induced RAW 2264.7 cells. J Exp Med 1985; 161: 984.
pentoxifylline, methylprednisone and anti-TNF and anti-γ anti- 153. Beutler B, Milsark I, Cerami A. Passive immunization against
bodies. J Immunol 1994 153; 499. cachectin/tumor necrosis factor protects mice from lethal effect
132. Ben-Sasson SZ, Le Gros G, Conrad DH, et al. IL-4 production by of endotoxin. Science 1985; 229: 869.
T cells from naive donors. J Immunol 1990; 145: 1127–1136. 154. Beutler BA, Cerami A. Recombinant interleukin 1 suppresses
133. Benchetrit F, Ciree A, Vives V, et al. Interleukin-17 inhibits tumor lipoprotein lipase activity in 3T3-L1 cells. J Immunol 1985;
cell growth by means of a T-cell-dependent mechanism. Blood 135: 3969–3971.
2002; 99: 2114–2121. 155. Bigler RD, Bushkin Y, Chiorazzi N. S152 (CD27). A modulating
134. Benjamin D, Sharma V, Knobloch TJ, et al. B cell IL-7. Human dissulfide-linked activation antigen. J Immunol 1988; 141: 21.
B cell lines constitutively secrete IL-7 and express IL-7 recep- 156. Bird TA, Davies T, Balswin SA, Saklatvala J. Interleukin 1 stim-
tors. J Immunol 1994; 152: 4749–4757. ulates hexose transport in fibroblasts by increasing the expression
135. Bennett MW, O’Connell J, O’Sullivan GC, et al. The Fas coun- of glucose transporters. J Biol Chem 1990; 265: 13578.
terattack in vivo: Apoptotic depletion of tumor infiltrating 157. Birg F, Courcoul M, Rosnet O, et al. Expression of FMS/KIT-like
lymphocytes associated with Fas ligand expression by human gene FLT3 in human leukemias of the myeloid and lymphoid
esophageal carcinoma. J Immunol 1998; 160: 5669–5675. lineages. Blood 1992; 80: 2584–2593.
Walter M. Lewko and Robert K. Oldham 221

158. Bissonnette EY, Chin B, Befus AD. Interferons differentially dendritic cells and generation of antitumor immunity in vivo. J
regulate histamine and TNF-α in rat intestinal mucosal mast Immunol 1999; 163: 1289–1297.
cells. Immunol 1995; 86: 12–17. 180. Born TL, Tomassen T, Bird TA, Sims Je. Cloning of a novel
159. Bjorek P, Bamchereua J, Flores-Romo L. CD40 ligation counter- receptor subunit, AcPL, required for interleukin-18 signaling. J
acts Fas-induced apoptosis of human dendritic cells. Int Immunol Biol Chem 1998; 273: 29445.
1997; 9: 365. 181. Borrow PA, Tishon A, Lee S, et al. CD40 ligand-deficient mice
160. Black RA, Kronheim SR, Cantrell M, et al. Generation of bio- show deficits in antiviral immunity and have impaired memory
logically active interleukin-1β by proteolytic cleavage of an inac- CD8+ CTL response. J Exp Med 1996; 183: 2129.
tive precursor. J Biol Chem 1988; 263: 9437. 182. Boskey AL, Maresca M, Ullrich W, et al. Osteopontin–
161. Black RA, Rauch CT, Kozlosky CJ, et al. A metalloproteinase hydroxyapatite interactions in vitro: inhibition of hydroxyapatite
disintegrin that releases tumor necrosis factor α from cells. formation and growth in a gelatin-gel. Bone Miner 1993; 22:
Nature 1997; 385: 729–733. 147–159.
162. Blankenstein T, Qin Z, Uberla K, et al. Tumor suppression after 183. Boulton TG, Stahl N, Yancopoulos GD. Ciliary neurotrophic fac-
cell targeted tumor necrosis factor α gene transfer. J Exp Med tor/leukemia inhibitory factor/interleukin 6/oncostatin M family
1991; 173: 1047–1052. of cytokines induces tyrosine phosphorylation of a common set
163. Blazar BR, Taylor PA, Smith S, Vallera DA. Interleukin-10 of proteins overlapping those induced by other cytokines and
decreases survival in murine recipients of major histocompati- growth factors. J Biol Chem 1994; 269: 11648.
bility complex disparate donor bone marrow grafts. Blood 184. Boussaud V, Soler P, Morreau J, Goodwin RG, Hance AJ.
1995; 85: 842. Expression of three members of the TNF-R family of receptors
164. Blevin MP, Anderson KV. A conserved signaling pathway: the (4-1BB, lymphotoxin-β and Fas) in human lung. Eur Respir J
Drosophila Toll-dorsal pathway. Ann Rev Cell Dev Biol 1996; 1998; 12: 926–931.
12: 393. 185. Bover LC, Cardo-vila M, Kiniyasu A, et al. A previously unrec-
165. Blin N, Stafford DW. Isolation of high molecular weight DNA. ognized protein–protein interaction between Tweak and CD163:
Nucleic Acid Res 1976; 3: 2303. potential biological implications. J Immunol 2007; 178:
166. Block MI, Berg M, McNamara MJ, et al. Passive immunization 8183–8194.
of mice against D factor blocks lethality and cytokine release 186. Bowen MA, Lee RK, Miragliotta G, Nam SY, Podack ER.
during endotoxemia. J Exp Med 1993; 178: 1085. Structure and expression of murine CD30 and its role in cytokine
167. Blotta MH, Marshall JD, De Kruyff RH, Umetsu DT. Cross- production. J Immunol 1996; 156: 442–449.
linking of the CD40 ligand on human CD4+ lymphocytes gener- 187. Bowman MR, Crimmins AV, Yetz-Aldape J, et al. The cloning of
ates a costimulatory signal that upregulates IL-4 synthesis. J CD70 and its identification as the ligand for CD27. J Immunol
Immunol 1996; 156: 3133. 1994; 152: 1756.
168. Blum-Degen D, Muller T, et al. Interleukin-1β and interleukin-6 188. Bracho F, Krailo MD, Shen V, et al. A phase I clinical, pharmaco-
are elevated in cerebrospinal fluid of Alzheimer’s and de novo logical, and biological trial of interleukin 6 plus granulocyte-
Parkinson’s disease patients. Neurosci Lett 1995; 202: 17. colony stimulating factor after ifosamide, carboplatin, and
169. Blumberg H, Conklin D, Xu WF, et al. Interleukin 20: discovery, etoposide in children with recurrent/refractory solid tumors:
receptor identification, and role in epidermal function. Cell 2001; enhanced hematologic responses but a high incidence of grade
104: 9–19. III/IV constitutional toxicities. Clin Cancer Res 2001; 7: 58–67.
170. Blumenthal RD, Lew W, Juweid M, et al. Plasma FLT-3 ligand 189. Bradding P, Feather IH, Howarth PH, et al. Interleukin 4 is
levels predict bone marrow recovery from myelosuppressive localized and released by human mast cells. J Exp Med 1992;
therapy. Cancer 2000; 88: 333–343. 176: 1381.
171. Boehme SA, Lio FM, Maciejewski-Lenoir D, Bacon KB, Conlon 190. Bradley EC, Louis AC, Paradise CM, et al. Antitumor response in
PJ. The chemokine fractalkine inhibits Fas-mediated cell death in patients with metastatic renal cell carcinoma is dependent upon
brain microglia. J Immunol 2000; 165: 397–403. regimen intensity (meeting abstract). Proc Am Soc Clin Oncol
172. Bogdan C, Thuring H, Dlaska M, Rollinghoff M, Weiss G. 1989; 8: A519.
Mechanism of suppression of nitric oxide release by IL-13: influ- 191. Bradley TR, Williams N, Kriegler AB, Fawcett J, Hodgson GS.
ence of the macrophage population. J Immunol 1998; 159: 4506. In vivo effects of interleukin-1α on regenerating marrow myeloid
173. Bogdan C, Vodovotz Y, Nathan CF. Macrophage inactivation by colony forming cells after treatment with S-fluorouracil.
interleukin 10. J Exp Med 1991; 174: 1549. Leukemia 1989; 3: 893.
174. Bohlen H, Kessler M, Sextro M, Diehl V, Tesch H. Poor clinical 192. Brandt K, Bufone-Paus S, Foster DC, et al. Interleukin-21 inhib-
outcome of patients with Hodgkin’s disease and elevated inter- its dendritic cell activation and maturation. Blood 2003; 102:
leukin-10 serum levels. Clinical significance of interleukin 10 4090–4098.
serum levels for Hodgkin’s disease. Ann Hematol 2000; 79: 193. Brasel K, McKenna HJ, Charrier K, et al. Flt3 ligand synergizes
110–113. with granulocyte-macrophage colony stimulating factor or gran-
175. Boileau C, Houde M, Dulude G, et al. Regulation of extrathymic ulocyte colony stimulating factor to mobilize hematopoietic pro-
T cell development and turnover by oncostatin M. J Immunol genitor cells into the peripheral blood of mice. Blood 1997; 90:
2000; 164: 5713–5720. 3781–3788.
176. Boldt DH, Mills BJ, Gemlo BT, et al. Laboratory correlates of 194. Brasel K, McKenna HJ, Morrisey PJ, et al. Hematologic effects
adoptive immunotherapy with recombinant interleukin-2 and of flt3 ligand in vivo in mice. Blood 1996; 88: 2004–2012.
lymphokine-activated killer cells in human. Cancer Res 1988; 195. Brazel CY, Ducceschi MH, Pytowski B, Levison SW. The flt3
48: 4409. tyrosine kinase receptor inhibits neural stem/progenitor cell pro-
177. Bolesta E, Kowalczyk A, Wierzbicki A, et al. Increased level and liferation and collaborates with NGF to promote neuronal sur-
longevity of protective immune responses induced by DNA vival. Mol Cell Neurosci 2001; 18: 381–393.
vaccine expressing the HIV-1 env glycoprotein when combined 196. Breder CD, Dinarello CA, Saper CB. Interleukin-1 immunoreac-
with IL-21 and IL-15. J Immunol 2006; 177: 177–191. tive innervation of the hypothalamus. Science 1988; 240: 321.
178. Boniface K, Bernard F-X, Garcia M, et al. IL-22 inhibits epider- 197. Brennan FM, Chantry D, Jackson A, et al. Inhibitory effects of
mal differentiation and induces proinflammatory gene expression TNF α antibodies on synovial cell interleukin 1 production in
and migration in human keratinocytes. J Immunol 2005; 174: rheumatoid arthritis. Lancet 1989; 11: 244.
3695–3702. 198. Brenne AT, Ro TB, Waage A, et al. Interleukin-21 is a growth and
179. Borges L, Miller RE, Jones J, Ariail K, et al. Synergistic action of survival factor for human myeloma cells. Blood 2002; 99:
fms-like tyrosine kinase and CD40 ligand in the induction of 3756–3762.
222 Cytokines

199. Bresnihan B, Alvaro-Garcia JM, Cobby M, et al. Treatment of 221. Buhlman JE, Foy TM, Aruffo A, et al. In the absence of a CD40
rheumatoid arthritis with recombinant human interleukin-1 signal, B cells are tolerogenic. Immunity 1995; 2: 645.
receptor antagonist. Arthritis Rheum 1998; 41: 2196–2204. 222. Bukowski RM, Raymond P, Molto L, et al. Interferon-γ and CXC
200. Brodie SJ, Lewisohn DA, Patterson BK, et al. In vivo migration chemokine induction by interleukin 12 in renal cell carcinoma.
and function of transferred HIV-1-specific cytotoxic T cells. Nat Clin Cancer Res 1999; 5: 2780–2789.
Med 1999; 5: 34–41. 223. Bulfone-Paus S, Ungureanu D, Pohl T, et al. Interleukin-15 pro-
201. Brody DT, Durum SK. Membrane IL-1: IL-1α precursor binds to tects from lethal apoptosis in vivo. Nat Med 1997; 3: 1124.
the plasma membrane via a lectin-like interaction. J Immunol 224. Bullock TN, Yagita H. Induction of CD70 on dendritic cells
1989; 143: 1183. through CD40 or TCR stimulation contributes to the develop-
202. Broide DH, Paine MM, Firestein GS. Eosinophils express inter- ment of CD8+ T cell responses in the absence of CD4+ T cells.
leukin 5 and granulocyte macrophage colony stimulating factor J Immunol 2005; 174: 710–717.
mRNA at sites of allergic inflammation in asthmatics. J Clin 225. Burd PR, Thompson WC, Max EE, Mills FC. Activated mast
Invest 1992; 90: 1414. cells produce interleukin-13. J Exp Med 1995; 181: 1373.
203. Brooks B, Rees RC. Human recombinant IL-4 suppresses the 226. Burger R, Neipel F, Fleckenstein B, Savino R, et al. Human her-
induction of human IL-2 induced lymphokine activated killer pesvirus type 8 interleukin-6 homologue is functionally active on
(LAK) activity. Clin Exp Immunol 1988; 74: 162. human myeloma cells. Blood 1998; 91: 1858.
204. Brown MA, Pierce JH, Watson CJ, et al. B cell stimulatory fac- 227. Burgess SJ, Marusina AI Pathmanathan I, et al. IL-21 down-
tor-1/Interleukin-4 is expressed by normal and transformed mast regulates NKG2D/DAP10 expression on human NK and CD8+ T
cells. Cell 1987; 50: 809–818. cells. J Immunol 2006; 176: 1490–1497.
205. Brown PM, Tagari P, Rowan KR, et al. Epitope labeled soluble 228. Burton JD, Bamford RM, Peters C, et al. A lymphokine provi-
human interleukin-5 (IL-5) receptors: affinity cross-link labeling, sionally designated interleukin T and produced by a human adult
IL-5 binding and biological activity. J Biol Chem 1995; 270: 29236. T cell leukemia line stimulates T cell proliferation and the induc-
206. Brown SA, Richards CM, Hanscom HN, et al. The Fn14 cyto- tion of lymphokine activated killer cells. Proc Nat Acad Sci USA
plasmic tail binds tumor necrosis factor receptor associated fac- 1994; 91: 4935–4939.
tors 1, 2, 3, and 5 and mediates nuclear factor-κB activation. 229. Cagnard N, Letourneur F, Essabani A, et al. Interleukin-32,
Biochemical J 2003; 371: 395–403. CCL2, PF4F1 and GFD10 are the only cytokine/chemokine
207. Brown TJ, Lioubin MN, Marquardt H. Purification and charac- genes differentially expressed by in vitro cultured rheumatoid
terization of cytostatic lymphokines produced by activated and osteoarthritis fibroblast-like synoviocytes. Eur Cytokine
human T lymphocytes: synergistic antiproliferative activity of Netw 2005; 16: 289–292.
transforming growth factor-β1, interferon-γ, and oncostatin M 230. Cai L, Starovasnik MA, Hoque DA, et al. Pathways by which
for human melanoma cells. J Immunol 1987; 139: 2977–2983. interleukin 17 induces cartilage breakdown in vitro and in vivo.
208. Brown TJ, Rowe JM, Shoyab H. Regulation of IL-6 expression Cytokine 2001; 16: 10–21.
by oncostatin M. J Immunol 1991; 147: 2175. 231. Cai X, Gommoll-CP J, Justice L, Narula SK, Fine JS. Regulation
209. Browning JL, Douglas I, Ngam-ek A, et al. Characterization of of granulocyte colony stimulating factor gene expression by
surface lymphotoxin forms. J Immunol 1996; 154: 33. interleukin-17. Immunol Lett 1998; 62: 51.
210. Browning JL, Mattaliano RJ, Pinchang CE, et al. Disulfide 232. Calderhead DM, Buhman JE, van den Eertwegh AJ, et al. Cloning
scrambling of interleukin-2: HPLC resolution of the three possi- of mouse Ox40: a T cell activation marker that may mediate T–B
ble isomers. Anal Biochem 1986; 155: 123. cell interactions. J Immunol 1993; 151: 5261.
211. Browning JL, Miatkowski K, Sizing I, et al. Signaling through 233. Camerini D, Walz G, Loenen WAM, Borst J, Seed B. The acti-
the lymphotoxin β receptor induces the death of some adenocar- vation antigen CD27 is a member of the nerve growth factor/
cinoma tumor cell lines. J Exp Med 1996; 183: 867. tumor necrosis factor receptor gene family. J Immunol 1991;
212. Browning JL, Ngam-ek A, Lawton P, et al. Lymphotoxin β, a 147: 3165.
novel member of the TNF family that forms a heteromeric com- 234. Cameron LA, Taha RA, Tsicopoulos A, et al. Airway epithelium
plex with lymphotoxin on the cell surface. Cell 1993; 72: 847. express interleukin-18. Eur Respir J 1999; 14: 553–559.
213. Bruce AG, Hoggart IH, Rose TM. Oncostatin M is a differentia- 235. Campbell IL, Abraham CR, Masliah E, et al. Neurologic disease
tion factor for myeloid leukemia cells. J Immunol 1992; 142: induced in transgenic mice by cerebral overexpression of inter-
1271–1275. leukin-6. Proc Natl Acad Sci USA 1993; 90: 10061.
214. Brugger W, Heimfeld S, Berenson RJ, Mertlesman R, Kanz L. 236. Campbell KA, Ovendale PJ, Kennedy MK, et al. CD40L is
Reconstitution of hematopoiesis after high-dose chemotherapy required for protective cell-mediated immunity to Leishmania
by autologous progenitor cells generated ex vivo. N Eng J Med major. Immunity 1996; 4: 283.
1995; 333: 283–287. 237. Campbell S, Burkly LC, Gao H-X, et al. Proinflammatory effects
215. Brugnoni D, Bettinardi A, Malacarne F, Airo P, Cattaneo R. of TWEAK/Fn14 interactions in glomerular mesangial cells.
CD134/OX40 expression by synovial fluid CD4+ T lymphocytes J Immunol 2006; 176: 1889–1898.
in chronic synovitis. Br J Rheumatol 1998; 37: 584. 238. Cannons JL, Lau P, Ghumman B, et al. 4-1BB ligand induces cell
216. Brunda MJ, Luistro L, Warrier RR, et al. Antitumor and antimeta- division, sustains survival, and enhances effector function of
static activity of interleukin-12 against murine tumors. J Exp CD4 and CD8 T cells with similar efficacy. J Immunol 2001; 167:
Med 1993; 178: 1223–1230. 1313–1324.
217. Brunner T, Yoo NJ, LaFace D, Ware CF, Green DR. Activation- 239. Cao H, Wolff RG, Meltzer MS, Crawford RM. Differential regu-
induced cell death in murine T cell hybridomas: differential regu- lation of class II determinants on macrophages by IFN-γ and
lation of Fas (CD95) versus Fas ligand expression by cyclosporin IL-4. J Immunol 143; 1989: 3524.
A and FK506. Int Immunol 1996; 8: 1017. 240. Cao R, Farnebo J, Kurimoto M, Cao Y. Interleukin-18 acts as an
218. Bucay N, Sarosi I, Dunstan CR, Morony S, et al. Osteoprotegerin angiogenesis and tumor suppressor. FASEB J 1999; 13: 2195.
deficient mice develop early onset osteoporosis and arterial calci- 241. Cao X, Zhang W, He L, Xie Z, et al. Lymphotactin-gene-modified
fication. Genes Dev 1998; 12: 1260. bone marrow dendritic cells act as more potent adjuvants for pep-
219. Budagian V, Bulanova E, Orinska Z, et al. Reverse signaling tide delivery to induce specific immunity. J Immunol 1998; 161:
through membrane bound interleukin-15. J Biol Chem 2004; 6238–6244.
279: 42192–42201. 242. Cao XX, Mohuiddin I, Chada S. Adenovirus transfer of mda-7
220. Budd GT, Green S, Baker LH, et al. A Southwest Oncology leads to BAX up-regulation and apoptosis in mesothelioma cells
Group phase II trial of recombinant tumor necrosis factor in met- and is abrogated by over expression of BCL-XL. Mol Med 2002;
astatic breast cancer. Cancer 1992; 68: 1694–1695. 8: 869–876.
Walter M. Lewko and Robert K. Oldham 223

243. Cao Z, Henzel WJ, Gao X. IRAK: a kinase associated with the 263. Caux C, Vanbervliet B, Massactier C, et al. Activation of
interleukin-1 receptor. Science 1996; 271: 1128. human dendritic cells by CD40 crosslinking. J Exp Med 1994;
244. Caput D, Laurent P, Kaghad M, et al. Cloning and characterization 180: 1263.
of a specific Interleukin (IL)-13 binding protein structurally 264. Cavallo F, Signorelli P, Giovarelli M, et al. Antitumor efficacy of
related to the IL-5 receptor α chain. J Biol Chem 1996; 271: adenocarcinoma cells engineered to produce interleukin 12
16921–16926. (IL-12) or other cytokines compared to exogenous IL-12. J Natl
245. Carbone E, Ruggiero G, Terrazzano G, et al. A new mechanism Canc Inst 1997; 89: 1049.
of NK cytotoxicity activation: the CD40:CD40 ligand interac- 265. Cavender DE, Edelbaum D, Ziff M. Endothelial cell activation
tion. J Exp Med 1997; 183: 2053. induced by tumor necrosis factor and lymphotoxin. Am J Pathol
246. Carrascal MT, Mendoza L, Valcaracel M. Interleukin-18 binding 1986; 134: 551.
protein reduces B16 melanoma hepatic metastasis by neutraliz- 266. Cayabyab M, Phillips JH, Lanier LL. CD40 preferentially co-
ing adhesiveness and growth factors of sinusoidal endothelium. stimulates activation of CD4+ T lymphocytes. J Immunol 1994;
Cancer Res 2003; 63: 491–497. 152: 1523.
247. Carriere V, Roussel L, Ortega N, et al. IL-33, the IL-1-like cytokine 267. Cella M, Scheidegger D, Palmer-Lehman K, et al. Ligation of
ligand for ST2 receptor, is a chromatin-associated nuclear factor in CD40 on dendritic cells triggers production of high levels of
vivo. Proc Natl Acad Sci USA 2007; 104: 282–287. interleukin-12 and enhances T cell stimulatory capacity: T-T help
248. Carroll GJ, Bell MC, Chapman HM, Mills JN, Robinson WF. via APC activation. J Exp Med 1996; 184: 747.
Leukemia inhibitory factor induces leukocyte infiltration and car- 268. Center DM, Berman JS, Kornfeld H, Theodore AC, Cruikshank
tilage proteoglycan degradation in goat joints. J Interferon WW. The lymphocyte chemoattractant factor. J Lab Clin Med
Cytokine Res 1995; 15: 567. 1995; 125: 167–172.
249. Carson WE, Giri JG, Lindenmann MJ, et al. Interleukin (IL) 15 is 269. Center DM, Cruikshank WW, Zhang Y. Pro-IL-16 regulation of T
a novel cytokine that activates human natural killer cells via com- cell proliferation: p27Kip1-dependent G0/G1 arrest mediated by
ponents of the IL-2 receptor. J Exp Med 1994; 180: 1395. inhibition of Skp2 transcription. J Immunol 2004; 172:
250. Carswell EA, Old LJ, Kassel RL, et al. An endotoxin induced 1654–1660.
serum factor that causes necrosis of tumors. Proc Natl Acad Sci 270. Center DM, Cruikshank WW. Modulation of lymphocyte migra-
USA 1975: 72: 3666–3670. tion by human lymphokines. I. Identification and characterization
251. Carter LL, Dutton RW. Relative perforin and Fas-mediated of a chemoattractant activity for lymphocytes from mitogen stim-
lysis in T1 and T2 CD8 effector populations. J Immunol ulated mononuclear cells. J Immunol 1982; 128: 2563.
1995; 155: 1028. 271. Center DM, Kornfeld H, Cruikshank WW. Interleukin-16 and its
252. Carveth HJ, Bohnsack JF, McIntyre TM, et al. Neutrophil acti- function as a CD4 ligand. Immunol Today 1996; 17: 476.
vating factor (NAF) induces polymorphonuclear leukocyte 272. Cerretti DP, Kozlowsky CJ, Mosely B, et al. Molecular cloning of
adherence to endothelial cells and to subendothelial matrix pro- the IL-1 β converting enzyme. Science 1992; 256: 97.
teins. Biochem Biophys Res Commun 1989; 162: 387. 273. Cerutti A, Schaffer A, Goodwin RG, et al. Engagement of CD153
253. Cascino I, Fiucci G, Papoff G, and Ruberti G. Three functional (CD30 Ligand) by CD30+ T cells inhibits class switch DNA
soluble forms of the human apoptosis-inducing Fas molecule are recombination and antibody production in human IgD+ IgM+ B
produced by alternative splicing. J Immunol 1995; 154: 2706. cells. J Immunol 2000; 165: 786–794.
254. Casey LS, Lichtman AH, Boothby M. IL-4 induced IL-2 receptor 274. Cerutti A, Schaffer A, Zan H, et al. CD30 is a CD40-inducible
p75 β chain gene expression and IL-2 dependent proliferation in molecule that negatively regulates CD40-mediated immunoglob-
mouse T lymphocytes. J Immunol 1992; 148: 3418–3426. ulin class switching. Immunity 1998; 9: 247.
255. Cassatella M, Bazzoni F, Ceska M, et al. IL-8 production by 275. Chabaud M, Durand JM, Buchs N, et al. Human Interleukin-17:
human polymorphonuclear leukocytes: the chemoattractant a T cell derived proinflammatory cytokine produced by the rheu-
formylmethionyl leucylphenylalanine induces the pathway. J matoid synovium. Arthritis Rheum 1999; 42: 963.
Immunol 1992; 148: 3216. 276. Chabaud M, Fossiez F, Taupin JL, Miossec P. Enhancing effect of
256. Cassatella MA, Gasperini S, Bovolenta C, et al. Interleukin-10 IL-17 on IL-1 induced IL-6 and leukemia inhibitory factor pro-
(IL-10) selectively enhances CIS3/SOCS3 mRNA expression duction by rheumatoid arthritis synoviocytes and its regulation
in human neutrophils: evidence for and IL-10 induced pathway by Th2 cytokines. J Immunol 1998; 161: 409.
that is independent of STAT protein activation. Blood 1999; 277. Chada S, Bocangel D, Ramesh R, et al. mda-7/IL-24 kills pancre-
94: 2880. atic cancer cells by inhibition of the Wnt/PI3K signaling path-
257. Cassatella MA, Meda L, Gasperini S, Calzetti F, Bonora S. ways: identification of IL-20 receptor-mediated bystander activity
Interleukin-10 (IL-10) upregulates IL-1 receptor antagonist pro- against pancreatic cancer. Mol Ther 2005; 11: 724–733.
duction from lipopolysaccharide stimulated human polymorpho- 278. Chada S, Sutton RB, Ekmekcioglu S, et al. MDA-7/IL-24 is a
nuclear leukocytes by delaying mRNA degradation. J Exp Med unique cytokine-tumor suppressor in the IL-10 family. Int
1994; 179: 1695. Immunopharmacol 2004; 4: 649–667.
258. Castell JV, Gomez-Lechnon MJ, David M, et al. Interleukin-6 is 279. Chakir J, Shannon J, Molet S, et al. Airway remodeling-associ-
the major regulator of acute phase protein synthesis in adult ated mediators in moderate to severe asthma: effect of steroids on
human hepatocytes. FEBS Lett 1989; 242: 237. TGF-β, IL-11, IL-17 and type I and type III collagen expression.
259. Castigli E, Pahwa R, Good RA, Geha RS, Chatila TA. Molecular J Allergy Clin Immunol 2003; 111: 1293–1298.
basis of a common lymphokine deficiency in a patient with 280. Chakrabarty S, Nagata M, Yasuda H, et al. Critical roles of CD30/
severe combined immunodeficiency. Proc Natl Acad Sci USA CD30L interactions in murine autoimmune diabetes. Clin Exp
1993; 90: 4728. Immunol 2003; 133: 318–325.
260. Castle BE, Kishimoto K, Stearns C, Brown ML, Kehry MR. 281. Chakraborty M, Abrams SI, Camphausen K, et al. Irradiation of
Regulation of expression of ligand for CD40 on T helper lympho- tumor cells upregulates Fas and enhances CTL lytic activity and
cytes. J Immunol 1993; 151: 1777. CTL adoptive immunotherapy. J Immunol 2003; 170: 6338–6347.
261. Caudell EG, Mumm JB, Poindexter N, et al. The protein product 282. Chakraborty NG, Li L, Sporn JR, et al. Emergence of regula-
of tumor suppressor gene, melanoma differentiation associated tory CD4+ T cell response to repetitive stimulation with antigen-
gene 7, exhibits immunostimulatory activity and is designated presenting cells in vitro: implications in designing antigen
IL-24. J Immunol 2002; 168: 6041–6046. presenting cell-based tumor vaccines. J Immunol 1999; 162:
262. Caux C, Dezutter-Dambuyant D, Schmitt D, Banchereau J. 5576–5583.
GM-CSF and TNF cooperate in the generation of dendritic 283. Chakravarty PK, Alfieri A, Thomas EK, et al. FLT-3 ligand
Langerhans cells. Nature 1992; 360, 258. administration after radiation therapy prolongs survival in a
224 Cytokines

murine model of metastatic lung cancer. Cancer Res 1999; 59: 304. Chicheportiche Y, Bourbon PR, Xu H, et al. TWEAK, a new
6028–6032. secreted ligand in the tumor necrosis family that weakly induces
284. Chan SH, Perussia B, Gupta JW, et al. Induction of interferon γ apoptosis. J Biol Chem 1997; 272: 32401–32410.
production by natural killer cell stimulatory factor: characteriza- 305. Chicheprotiche Y, Chicheportiche R, Sizing I, et al. Proinflammatory
tion of responding cells and synergy with other inducers. J Exp activity of TWEAK on human dermal fibroblasts and synovio-
Med 1991; 173: 869–879. cytes: blocking and enhancing effects of anti-TWEAK monoclonal
285. Chan WL, Pejnovic N, Lee CA, Al-Ali NA. Human IL-18 recep- antibodies. Arthritis research 2001; 4: 126–133.
tor and ST2L are stable and selective markers for the respective 306. Chien Y, Gascoigne N, Kavaler J, et al. Somatic recombination in
type 1 and type 2 circulating lymphocytes. J Immunol 2001; 167: a murine T-cell-receptor gene. Nature 1984; 309: 322.
1238–1244. 307. Chilton PM, Fernandez-Botran R. Production of soluble IL-4
286. Chang C-I, Zoghi B, Liao JC, Kuo L. The involvement of tyrosine receptors by murine spleen cells is regulated by activation and
kinases, cyclic AMP/protein kinase A, and p38 mitogen-activated IL-4. J Immunol 1993; 151: 5907–5917.
protein kinase in IL-13 mediated arginase I induction in mac- 308. Chiu C-P, Moulds C, Coffman RL, et al. Multiple biological
rophages: its implications in IL-13 inhibited nitric oxide produc- activities are expressed by a mouse interleukin 6 cDNA clone
tion. J Immunol 2000; 165: 2134–2141. isolated from bone marrow stromal cells. Proc Nat Acad Sci USA
287. Chang H, Hanawa H, Liu H, et al. Hydrodynamic-based delivery 1988; 85: 7099–7103.
of an interleukin-22-Ig fusion gene ameliorates experimental 309. Chiyo M, Shimozato O, Yu Y, et al. Expression of IL-27 in murine
autoimmune myocarditis in rats. J Immunol 2006; 3635–3643. carcinoma cells produces antitumor effects and induces protec-
288. Chao T-C, Van Alter PJ, Greager JA, Walter RJ. Sex steroid hor- tive immunity in inoculated host animals. Int J Cancer 2005; 115:
mones regulate the release of tumor necrosis factor by mac- 437–442.
rophages. Cell Immunol 1995; 160: 43. 310. Chizzonite R, Truitt T, Kilian PL, et al. Two high affinity inter-
289. Chapoval AI, Tamda K, Chen L. In vitro inhibition of a broad leukin 1 receptors represent separate gene products. Proc Natl
spectrum of tumor cell lines by activated human dendritic cells. Acad Sci USA 1989; 86: 8029.
Blood 2000; 95: 2346. 311. Cho D, Song H, Kim YM, et al. Endogenous interleukin-18 mod-
290. Charlton B, Lafferty KJ. The Th1/Th2 balance in autoimmunity. ulates immune escape of murine melanoma cells by regulating
Curr Opinion Immunol 1995; 7: 793–798. the expression of Fas ligand and reactive oxygen intermediates.
291. Chattopadhyay S, Tracy E, Liang P, et al. Interleukin-31 and Cancer Res 2000; 60: 2703–2709.
oncostatin-M mediate distinct signaling reactions and response 312. Choi BK, Asai T, Vinay DS, et al. 4-1BB-mediated amelioration
patterns in lung epithelial cells. J Biol Chem 2007; 282: of experimental autoimmune uveoretinitis is caused by indoleam-
3014–3026. ine 2,3-diogygenase-dependent mechanisms. Cytokine 2006; 34:
292. Chaudhari U, Romano P, Mulcahy LD, et al. Efficacy and safety 233–242.
of infliximab monotherapy for plaque-type psoriasis: a random- 313. Chouat G, Milani AA, Martal J, et al. IL-10 prevents naturally
ized trial. Lancet 2001; 357: 1842–1847. occurring fetal loss in the CBA × DBA/2 mating combination and
293. Chazen GD, Pereira GMB, Le Gross G, et al. Interleukin 7 is a T local defect in IL-10 production in the abortion prone combina-
cell growth factor. Proc Nat Acad Sci USA 1989; 86: tion is corrected by in vivo injection of IFN-γ. J Immunol 1995;
5923–5927. 154: 4261.
294. Chen IA, McAdam AJ Buhlman JE, et al. Ox40-ligand has a criti- 314. Chowers Y, Cahalon L, Lahav M, et al. Somatostatin through its
cal costimulatory role in dendritic cell:T Cell interactions. specific receptor inhibits spontaneous and TNF-α and bacterial-
Immunity 1999; 11: 689. induced IL-8 and IL-1β secretion from intestinal epithelial cells.
295. Chen J, Chada S, Mhashilkar A, Miano JM. Tumor suppressor J Immunol 2000; 165: 2955–2961.
MDA-7/IL-24 selectively inhibits vascular smooth muscle cell 315. Christen U, Wolfe T, Mohrle U, et al. A dual role of TNF-α in
growth and migration. Mol Ther 2003; 8: 220–229. type 1 diabetes: Islet-specific expression abrogates the ongoing
296. Chen L, Mory Y, Zilberstein A, Revel M. Growth inhibition of autoimmune process when induced late but not early during
human breast carcinoma and leukemia/lymphoma cell lines by pathogenesis. J Immunol 2001; 166: 7023–7032.
recombinant interferon-β2. Proc Natl Acad Sci USA 1988; 85: 316. Chtanova T, Tangye SG, Newton R, et al. T follicular helper cells
8037. express a distinctive transcriptional profile, reflecting their role as
297. Chen WF, Zlotnik A. IL-10: a novel cytotoxic T cell differentia- non-Th1/Th2 effector cells that provide help for B cells. J
tion factor. J Immunol 1991; 147: 528. Immunol 2004; 173: 68–78.
298. Chen Y, Langrish CL, McKenzie B, et al. Anti-IL-23 therapy 317. Chu NR, DeBenedette MA, Stiernholm BJN, Barber BH, Watts
inhibits multiple inflammatory pathways and ameliorates auto- TH. Role of IL-12 and 4-1BB ligand in cytokine production by
immune encephalitis. J Clin Invest 2006; 116: 1317–1326. CD28+ and CD28− T cells. J Immunol 1997; 158: 3081–3089.
299. Cheneval D, Ramage P, Kastelic T, et al. Increased mature 318. Chua AO, Chizzonite R, Desai BB, et al. Expression cloning of a
interleukin-1β (IL-1β) secretion form THP-1 cells induced by human IL-12 receptor component: a new member of the cytokine
nigericin is a result of activation of p45 IL-1β-converting enzyme superfamily with strong homology with gp130. J Immunol 1994;
processing. J Biol Chem 1998; 273: 18846. 153: 128.
300. Chernoff AE, Granowitz EV, Shapiro L, et al. A randomized con- 319. Chuntarapai A, Dodge K, et al. Isotype-dependent inhibition of
trolled trial of IL-10 in humans: inhibition of inflammatory tumor growth in vivo by monoclonal antibodies to death receptor
cytokine production and immune response. J Immunol 1995; 4. J Immunol 2001; 166: 4891–4898.
154: 5492–5499. 320. Ciavarra RP, Somers KD, et al. Flt3 ligand induces transient
301. Cheung PF, Wong CK, Ip WK and Lam CW. IL-25 regulates the tumor regression in an ectopic treatment model of major histo-
expression of adhesion molecules on eosinophils: mechanism of compatibility complex-negative prostate cancer. Cancer Res
eosinophilia in allergic inflammation. Allergy 2006; 61: 878–885. 2000; 60: 2081–2084.
302. Chiaramonte MG, Mentink-Kane M, Jacobson BA, et al. 321. Claasen JL, Levine AD, Buckley RH. Recombinant human IL-4
Regulation and function of the interleukin-13 receptor α2 during induces IgE and IgG synthesis by normal and atopic donor mono-
a T helper cell type 2-dominant response. J Exp Med 2003; 197: nuclear cells. J Immunol 1991; 144: 123–130.
687–701. 322. Clamin H. Mast cells, T-cells, and abnormal fibrosis. Immunol
303. Chiarle R, Podda A, Prolla G, et al. CD30 overexpression Today 1985; 6: 192.
enhances negative selection in the thymus and mediates pro- 323. Clark-Lewis I, Kent SBH, Schrader JW. Purification to apparent
grammed cell death via a Bcl-2-sensitive pathway. J Immunol homogeneity of a factor stimulating the growth of multiple lin-
1999; 163: 194. eages of hemopoietic cells. J Biol Chem 1984; 259: 7488.
Walter M. Lewko and Robert K. Oldham 225

324. Clark-Lewis I, Schrader JW. P-cell stimulating factor: biochemi- 346. Costa JJ, Demetri GD, Harrist TJ, et al. Recombinant human
cal characterization of a new T-cell-derived factor. J Immunol stem cell factor (kit ligand) promotes human mast cell and mel-
1981; 127: 1941. anocyte hyperplasia and functional activation in vivo. J Exp Med
325. Clegg CH, Haugen HS, Rulffes JT, Friend SL, Farr AG. 1996; 2681–2686.
Oncostatin M transforms lymphoid tissue function in transgenic 347. Costello R, Brailly H, Mallet F, et al. Interleukin-7 is a potent
mice by stimulating lymphnode T cell development and thymus costimulus of the adhesion pathway involving CD2 and CD28
autoantibody production. Exp Haematology 1999; 27: 712. molecules. Immunology 1993; 80: 451–457.
326. Coccia EM, Passini N, Battistini A, et al. Interleukin-12 induces 348. Couderc B, Zitvogel L, Douin-Echinard V, et al. Enhancement of
expression of interferon regulatory factor-1 via signal transducer antitumor immunity by expression of CD70 (CD27 ligand) or
and activator of transcription-4 in human T helper type 1 cells. J CD154 (CD40 ligand) costimulatory molecules in tumor cells.
Biol Chem 1999; 274: 6698–6703. Cancer Gene Ther 1998; 5: 163–175.
327. Coceani F, Lees J, Reford J, Bishai I. Interleukin-1 receptor 349. Coughlin CM, Salany KE, Wysocka M, et al. Interleukin-12 and
antagonist: effectiveness against interleukin-1 fever. Can J interleukin-18 synergistically induce murine tumor regression,
Physiol Pharmacol 1992; 7: 1590. which involves inhibition of angiogenesis. J Clin Invest 1998;
328. Cocks BG, de Waal Malefyt R, Galizzi JP, de Vries JE. IL-13 101: 1441.
induces proliferation and differentiation of human B cells acti- 350. Coughlin CM, Salhany KE, Gee MS, et al. Tumor cell responses
vated by the CD40 ligand. Int Immunol 1993; 5: 657. to IFNγ affect tumorigenicity and response to IL-12 therapy and
329. Coffman RL, Bond MW, Carty J, et al. B cell stimulatory factor antiangiogenesis. Immunity 1998; 9: 25.
1 enhances the IgE response of lipopolysaccharide-activated B 351. Crawford R, Finboom D, Ohara J, et al. B cell stimulatory factor
cells. J Immunol 1986; 136: 4538–4541. 1 (Interleukin 4) activates macrophages for increased tumori-
330. Cohen PL and Eisenberg RA. Lpr and gld: single gene models of cidal activity and expression of Ia antigens. J Immunol 1987;
systemic autoimmunity and lymphoproliferative diseases. Ann 139: 135.
Rev Immunol 1991; 9: 243. 352. Cretney E, Takeda K, Yagita H, et al. Increased susceptibility to
331. Cohen SBA, Katsikis PD, Feldman M, Londei E. IL-10 enhances tumor initiation and metastasis in TNF-related apoptosis-induc-
expression of the IL-2 receptor α chain on T cells. Immunol ing ligand-deficient mice. J Immunol 2002; 168: 1356–1361.
1994, 83: 329. 353. Crooks GM, Hao QL, Peterson D, Barsky LW, Brockstoce D.
332. Cohen SBA, Parry SL, Feldman M, Foxwell B. Autocrine and IL-3 increases production of B lymphoid progenitors from human
paracrine regulation of human T cell IL-10 production. J Immunol CD34+ CD38−cells. J Immunol 2000; 165: 2382–2389.
1997; 158: 5596–5602. 354. Croston GE, Cao Z, Goeddel DV. NF-κB activation by interleu-
333. Cohney SJ, Sanden D, Cacalano NA, et al. SOCS-3 is tyrosine kin-1 (IL-1) requires an IL-1 receptor-associated protein kinase
phosphorylated in response to interleukin-2 and suppresses activity. J Biol Chem 1995; 270: 16514.
STAT5 phosphorylation and lymphocyte proliferation. Mol Cell 355. Crowe PD, van Arsdale TL, Walter BN, et al. A lymphotoxin-β
Biol 1999; 19: 4980. specific receptor. Science 1994; 264: 707.
334. Colotta F, Re F, Muzio M, Bertini R, et al. Interleukin-1 type II 356. Crowell S, Knauper V, Stewart ML, et al. Induction of martix
receptor: a decoy target for IL-1 that is regulated by IL-4. Science metalloproteinase activation cascades based on membrane-type
1993; 261: 472. matrix metalloproteinase associated activation of gelatinase A,
335. Collision LW, Workman CI, Kuo TT, et al. The inhibitory gelatinase B, and collagenase 3. Biochem J 1998, 331: 453.
cytokine IL-35 contributes to regulatory T-cell function. Nature 357. Crown J, Jakubowski A, Kemeny N, et al. A phase I trial of
2007; 450: 566–569. recombinant human interleukin-1β alone and in combination
336. Colombel JF, Rutgeerts P, Malchow H, et al. Interleukin-10 with myelosuppressive doses of 4-fluorouracil in patients with
(Tenovil) in the prevention of post operative Crohn’s disease. Gut gastrointestinal cancer. Blood 1991; 78: 1420–1427.
2001; 49: 42–46. 358. Cruikshank WW, Berman JS, Theodore AC, Bernardo J, Center
337. Colotta F, Re F, Muzio M, Polentarutti N, et al. Interleukin-13 DM. Lymphokine activation of T4+ lymphocytes and monocytes.
induces expression and release of interleukin-1 decoy receptor in J Immunol 1987; 138: 3817.
human polymorphonuclear cells. J Biol Chem 1994; 269: 12403. 359. Cruikshank WW, Center DM, Nisar N, et al. Molecular and func-
338. Comes A, Rosso O, Orengo AM, et al. CD25+ regulatory T cell tional analysis of a lymphocyte chemoattractant factor: associa-
depletion augments immunotherapy of micrometastases by an tion of biologic function and CD4 expression. Proc Natl Acad Sci
IL-21-secreting cellular vaccine. J Immunol 2006; 176: 1750–1758. USA 1994; 91: 5109.
339. Consentino G, Soprana E, Thienes CP, et al. IL-13 down-regu- 360. Cruikshank WW, Center DM. Modulation of lymphocyte migra-
lates CD14 expression and TNF-α secretion in normal human tion by human lymphokines. II. Purification of a lymphotactic
monocytes. J Immunol 1995; 155: 3145–3151. factor (LCF). J Immunol 1982; 128: 2569.
340. Conti B, Jahng JW, Tinti C, et al. Induction of IFN-γ inducing 361. Cruikshank WW, Greenstein JL, Theodore AC, Center DM.
factor in the adrenal cortex. J Biol Chem 1997; 272: 2025–2037. Lymphocyte attractant factor induces CD-4 dependent intracyto-
341. Contractor NV, Bassiri H, et al. Lymphoid hyperplasia, autoim- plasmic signaling in lymphocytes. J Immunol 1991; 146: 2928.
munity, and compromised intestinal intraepithelial lymphocyte 362. Cruikshank WW, Long A, Tarpy RE, et al. Early identification of
development in colitis-free gnotobiotic IL-2 – deficient mice. J interleukin-16 (Lymphocyte chemoattractant factor) and mac-
Immunol 1998; 160: 385–394. rophage inflammatory protein 1α (MIP1α) in bronchoalveolar
342. Contractor V, Pippin J, Russell J, et al. Identification of cDNA lavage fluid of antigen-challenged asthmatics. Am J Respir Cell
and genomic localization for IL-14. FASEB J 1994; 8: A506. Mol Biol 1995; 13: 738–747.
343. Conway JG, Andrews RC, Beaudet B, Bickett DM, et al. 363. Cua DJ, Sherlock J, Chen Y, et al. Interleukin-23 rather than
Inhibition of tumor necrosis factor-α (TNF-α) production and interleukin-12 is the critical cytokine for autoimmune inflamma-
arthritis in the rat by GW3333, a dual inhibitor of TNF-α con- tion of the brain. Nature 2003; 421: 744–748.
verting enzyme and matrix metalloproteinases. J Pharmacol Exp 364. Cui X, Yang SC, Sharma S, et al. IL-4 regulates COX-2 and
Ther 2001; 298: 900–908. PGE2 production in human non-small-cell lung cancer. Biochem
344. Cope AP, Aderka D, Wallach D, et al. Soluble TNF receptor pro- Biophys Res Commun 2006; 343: 995–1001.
duction by activated T lymphocytes: differential effects of acute 365. Cumberbatch M, Dearman RJ, Kimber I. Constitutive and induc-
and chronic exposure to TNF. Immunology1995: 84: 21. ible expression of interleukin-6 by Langerhans cells and lymph
345. Copeland NG, Gilgert DJ, Cho BC, et al. Mast cell growth factor node dendritic cells. Immunology 1996; 87: 513–518.
maps near the steel locus on mouse chromosome 10 and is deleted 366. Cunningham CC, Chada S, Merritt JA, et al. Clinical and biologi-
in a number of steel alleles. Cell 1990; 63: 175–183. cal effects of an intratumoral injection of mda-7 (IL-24; INGN 241)
226 Cytokines

in patients with advancer carcinoma: a phase I study. Mol Ther 388. De Waal Malefyt R, Abrams J, Bennett B, et al. Interleukin 10
2005; 11: 149–159. (IL-10) inhibits cytokine synthesis by human monocytes: an
367. Curti A, Fogli M, Ratta M, Tura S, Lemoli RM. Stem cell factor autoregulatory role of IL-10 by monocytes. J Exp Med 1991b;
and FLT3 ligand are strictly required to sustain the long-term 174: 1209.
expansion of primitive CD34+ DR− dendritic cell precursors. J 389. DeBenedette MA, Chu NR, Pollok KE, et al. Role of 4-1BB
Immunol 2001; 166: 848–854. ligand in costimulation of T lymphocyte growth and its upregu-
368. D’Andrea A, Rangaraju M, Valiante NM, et al. Production of lation on M12 V lymphocytomas by cAMP. J Exp Med 1995;
natural killer cell stimulatory factor (interleukin 12) by peripheral 181: 985.
blood mononuclear cells. J Exp Med 1992; 176: 1387–1398. 390. DeBenedette MA, Wen T, Bachmann MF, et al. Analysis of
369. D’Andrea AM, Aste-Amezaga M, Valiante NM, et al. 4-1BB ligand (4-1BBL)-deficient mice and of mice lacking both
Interleukin-10 (IL-10) inhibits human interferon-γ production by 4-1BBL and CD28 reveals a role for 4-1BBL in skin allograft
suppressing natural killer cell stimulatory factor/IL-12 synthesis rejection and in the cytotoxic T cell response to influenza virus.
in accessory cells. J Exp Med 1993; 178: 1041. J Immunol 1999; 163: 4833–4841.
370. Da Silva N, Hu ZB, Ma W, et al. Expression of the Flt3 gene in 391. DeBenedette MA, Shahinian A, Mak TW, Watts TH. Costimulation
human leukemia-lymphoma cell lines. Leukemia 1994; 8: of CD28− T lymphocytes by 4-1BB ligand. J Immunol 1997; 158:
885–888. 551–559.
371. Dadi KH and Roifman CM. Activation of phosphatidylinositol-3 392. Debets R, Timens JC, Churakowa T, et al. IL-18 receptors, their
kinase by ligation of the interleukin 7 receptor on human thymo- role in ligand binding and function: Anti-IL-1RAcPL antibody, a
cytes. J Clin Invest 1993; 92: 1559–1563. potent antagaonist of IL-18. J Immunol 2000; 165: 4950–4956.
372. Dadi KH and Roifman CM. Interleukin 7 receptor mediates the 393. Debinski W, Obiri NI, Powers SK, Pastan I, Puri RK. Human
activation of phosphatidyl-3 inositol kinase in human B-cell pre- glioma cells overexpress receptors for IL-13 and are extremely
cursors. Biochem Biophys Res Commun 1993; 192: 450–464. sensitive to a novel chimeric protein composed of IL-13 and
373. Dai CH, Krantz SB, Zsebo KM. Human burst forming units Pseudomonas exotoxin. Clin Cancer Res 1995; 1: 1253.
erythroid need direct interaction with stem cell factor for further 394. DeBlaker-Hohe DF, Yamauchi A, Yu CR, et al. IL-12 synergizes
development. Blood 1991; 78: 2493–2497. with IL-2 to induce lymphokine activated cytotoxicity and gran-
374. Dalloul A, Laroche L, Bagot M, et al. Interleukin-7 is a growth zyme gene expression in fresh human NK cells. Cell Immunol
factor for Sezary lymphoma cells. J Clin Invest 1992; 90: 1995; 165: 33.
1054–1060. 395. Dechanet J, Taupin JL, Chomarat P, et al. Interleukin-4 but not
375. Damle NK, Doyle LV, Bender JR, Bradley EC. Interleukin 2-ac- interleukin-10 inhibits the production of leukemia inhibitory fac-
tivated human lymphocytes exhibit enhanced adhesion to normal tor by rheumatoid synovium and synoviocytes. Eur J Immunol
vascular endothelial cells and cause their lysis. J Immunol 1987; 1994; 24: 3222.
138: 1779. 396. DeChiara TM, Vejsada R, Poueymirou WT, et al. Mice lacking
376. Dandona P, Aljada A, Garg R, Mohanty P. Increase in plasma CNTF receptor, unlike mice lacking CNTF, exhibit profound
interleukin-10 following hydrocortisone injection. J Clin motor neuron deficits at birth. Cell 1995; 83: 313.
Endocrinol metal 1999; 84: 1141–1144. 397. Declercq W, Denecker G, Fiers W, Vandenabeele P. Cooperation
377. Danese S, de la Motte C, Reyes BM, et al. T cells trigger CD40- of both TNF receptors in inducing apoptosis: involvement of the
dependent platelet activation and granular RANTES release: TNF receptor-associated factor binding domain of the TNF
novel pathway for immune response amplification. J Immunol receptor 75. J Immunol 1998; 161: 390–399.
2004; 172: 2011–2015. 398. Defrance T, Carayon P, Billian G, et al. Interleukin 13 is a B cell
378. Danforth DN Jr, Sgagias MK. Interleukin-1α and interleukin-6 stimulating factor. J Exp Med 1994; 179: 135.
act additively to inhibit growth of MCF-7 breast cancer cells in 399. Defrance T, Fluckinger AC, Rossi JF, et al. Antiproliferative
vitro. Cancer Res 1993; 53: 1538–1545. effects of Interleukin-4 on freshly isolated non-Hodgkin malig-
379. Dannull J, Nair S, Su Z, et al. Enhancing the immunostimulatory nant B cell lymphoma cells. Blood 1992; 79: 990–996.
function of dendritic cells by transfection with mRNA encoding 400. Defrance T, Vanbervliet B, Briere F, et al. Interleukin-10 and
OX40 ligand. Blood 2005; 105: 3206–3213. transforming growth factor β cooperate to induce anti-CD40-ac-
380. Dannull J, Su Z, Rizzieri D. Enhancement of vaccine-mediated tivated naive human B cells to secrete immunoglobulin A. J Exp
antitumor immunity in patients after depletion of regulatory T Med 1992; 175: 671.
cells. J Clin Invest 2005; 115: 3623–3633. 401. Degli-Esposti M. To die or not to die: the quest of the trail recep-
381. Dao T, Ohashi K, Kayako T, Kurimoto M, Okamura H. tors. J Leukocyte Biol 1999; 65: 535.
Interferon-γ inducing factor, a novel cytokine, enhances Fas 402. Degli-Esposti MA, Davis-Smith T, Din WS, et al. Activation of
ligand mediated cytotoxicity of murine T helper cells. Cell lymphotoxin-β receptor by cross-linking induces chemokine pro-
Immunol 1996; 173: 230. duction and growth arrest in A375 melanoma cells. J Immunol
382. Dastych J, Metcalf DD. Stem cell factor induces mast cell adhe- 1997; 158: 1756.
sion to fibronectin. J Immunol 1994; 152: 213. 403. Degli-Esposti MA, Dougall WC, Smolak PJ, et al. The novel
383. Davis ID, Skrumsager BK, Cebon J, et al. An open-label, two receptor TRAIL-R4 induces NF-κB and protects against TRAIL-
arm, phase I trial of recombinant interleukin-21 in patients with mediated apoptosis, yet contains an incomplete death domain.
metastatic melanoma. Clin Cancer Res 2007; 13: 3630–3636. Immunity 1997; 7: 813.
384. Davis S, Aldrich TH, Stahl N, et al. LIFRβ and gp130 as het- 404. Degli-Esposti MA, Smolak PJ, Walczak H, et al. Cloning and
erodimerizing signal transducers of the tripartite CNTF receptor. characterization of TRAIL-R3, a novel member of the emerging
Science 1993; 260: 1805. TRAIL receptor family. J Exp Med 1997; 186: 1165.
385. De Bie JJ, Hendricks PA, Cruikshank WW, et al. Effects of inter- 405. DeGoer de Herve MG, Durali D, Tran TA, et al. Differential
leukin-16-blocking peptide on parameters of allergic asthma in a effect of agonistic anti-CD40 on human mature and immature
murine model. Eur J Pharmacol 1999; 383: 189–196. dendritic cells: the Janus face of anti-CD40. J Immunol Blood
386. De Smedt T, Smith J, Baum P, et al. OX40 costimulation enhances 2005; 106: 2806–2814.
the development of T cell responses induced by dendritic cells in 406. Dejana E, Bertocci F, Bortolami MC, et al. Interleukin 1 pro-
vivo. J Immunol 2002; 168: 661–670. motes adhesion to cultured endothelial cells. J Clin Invest 1988;
387. De Totero D, Meazza R, Zupo S, et al. Interleukin-21 (IL-21) 82: 1466.
receptor is upregulated by CD40 triggering and mediates 407. Del Peso L, Gonzalez-Garcia M, Page C, Herrara R, Nunez G.
proapoptotic signals in chronic lymphocytic leukemia B cells. Interleukin-3-induced phosphorylation of BAD through the
Blood 2006; 107: 3708–3715. protein kinase Akt. Science 1997; 278: 687–689.
Walter M. Lewko and Robert K. Oldham 227

408. Del Pozo V, De Andreas B, Martin E, et al. Eosinophil as antigen 430. Diehl L, Boer AT, Schoenberger SP, et al. CD40 activation in vivo
presenting cells: activation of T cell clones and T cell hybridoma overcomes peptide-induced peripheral cytotoxic T-lymphocyte
by eosinophils after antigen processing. Eur J Immunol 1992; and augments anti-tumor vaccine efficiency. Nat Med 1999;
22: 1919. 5: 774.
409. Del Prete G, Maggi E, Parronchi P, et al. IL-4 is an essential 431. Dillon SR, Sprecher C, Hammond A, et al. Interleukin 31, a
factor for the IgE synthesis induced in vitro by human T cell cytokine produced by activated T cells, induces dermatitis in
clones and their supernatants. J Immunol 1988; 140: 4193–4198. mice. Nat Immunol 2004; 5: 752–760.
410. Del Rey A, Besdovsky H, Sorkin E, Dinarello CA. Interleukin-1 432. Dilloo D, Bacon K, Holeden W, et al. Combined chemokine and
and glucocorticoid hormones integrate an immunoregulatory cytokine gene transfer enhances antitumor immunity. Nat Med
feedback circuit. Ann NY Acad Sci 1987; 496: 85. 1996; 2: 1090.
411. Della Bella s, Nicola S, Timofeeva I, et al. Are interleukin-16 and 433. Dilloo D, Brown M, Roscrow M, et al. CD40 ligand induces and
thrombopoietin new tool for the generation of dendritic cells? antileukemia response in vivo. Blood 1997; 90: 1927.
J Immunol 2004; 104: 4020–4028. 434. Dinarello CA. An IL-1 family member required caspase-1 pro-
412. Demoulin JB, Uyttenhove C, Van Roost E, et al. A single tyrosine cessing and signals through the ST2 receptor. Immunity 2005;
of the interleukin-9 (IL-9) receptor is required for STAT activa- 23: 461–462.
tion, anti-apoptotic activity, growth regulation by IL-9. Mol Cell 435. Dinarello CA, Cannon JG, Wolff SM, et al. Tumor necrosis factor
Biol 1996; 16: 4710–4716. (Cachectin) is an endogenous pyrogen and induces production of
413. Denhardt DT, Chambers AF. Overcoming obstacles to metastasis interleukin 1. J Exp Med 1986; 163: 1433.
defenses to host defenses: osteopontin (OPN) as a shield against 436. Dinarello CA, Goldin NP, Wolf SM. Demonstration and charac-
attack by cytotoxic host cells. J Cell Biochem 1994; 56: 48. terization of two distinct human leukocyte pyrogens. J Exp Med
414. Dennig D, O’ Reilly RJ. L7 induces surface expression of B7/BB1 on 1974; 139: 1369.
Pre-B cells and an associated increase in their costimulatory effects 437. Dinarello CA, Ikejima T, Warner SJ, et al. Interleukin-1 induces
on T cell proliferation. Cellular Immunol 1994; 153: 227–238. interleukin-1. I. Induction of circulating interleukin-1 in rabbits
415. Derocq J-M, Sequi M, Poinot-Chazel C, et al. Interleukin-13 in vivo and in human mononuclear cells in vitro. J Immunol
stimulates interleukin-6 production by human keratinocytes. 1987; 139: 1902.
FEBS Lett 1994; 343: 32. 438. Dinarello CA, Kim SH. IL-32, a novel cytokine with a possible
416. Desai BB, Quinn PM, Wolitzky AS, et al. IL-12 receptor II. role in disease. Ann Rheum Dis 2006; 65: Suppl 3: iii61–iii64.
Distribution and receptor expression. J Immunol 1992; 148: 439. Dinarello CA. IL18: A Th1-inducing proinflammatory cytokine
3125–3132. and a member of the IL-1 family. J Allergy Clin Immunol 1999;
417. Desplat-Jego S, Varriale S, Creidy R, et al. TWEAK is expressed 103: 11.
by glial cells, induces astrocyte proliferation and increases EAE 440. Ding O, Stewart J Jr, Prince CW, et al. Promotion of malignant
severity. J Neuroimmunol 2002; 133: 116–123. astrocytoma cell migration by osteopontin expressed in the normal
418. Desreumauz P, Janin A, Colombel JF, et al. Interleukin 5 mes- brain: differences in integrin signaling during cell adhesion to
senger RNA expression by eosinophils in the intestinal mucosa of osteopontin versus vitronectin. Cancer Res 2002; 62: 5336–5343.
patients with coeliac disease. J Exp Med 1992; 175: 293. 441. DiSanto JP, Bonnefoy JY, Gauchat JF, Fischer A, de Saint Badile
419. DeTogni PD, Goellner J, Ruddle NH, et al. Abnormal develop- G. CD40 ligand mutations in X-linked immunodeficiency with
ment of peripheral lymphoid organs in mice deficient in lympho- hyper-IgM. Nature 1993; 361: 541.
toxin. Science 1994; 264: 703. 442. Dokter WH, Sierdsema SJ, Esselink MT, et al. IL-7 enhances
420. Deveraux QL, Reed JC. IAP family proteins: suppressors of expression of IL-3 and granulocyte-macrophage-CSF mRNA in
apoptosis. Genes Dev 1999; 113: 239. activated human T cells by post-transcriptional mechanisms. J
421. Devos R, Guisez Y, Tavernier J, Plaetinck G. Recombinant solu- Immunol 1993; 150: 2584–2590.
ble interleukin-5 (hIL-5) receptor molecules: cross-linking and 443. Dokter WH, Sierdsema SJ, Esselink MT, et al. Interleukin 4
stoichiometry of binding to IL-5. J Biol Chem 1993; 268: 6581. mRNA and protein in activated human T cells are enhanced by
422. DeWaal Malefyt R, Figdor C, Huijbens R, et al. Effects of IL-13 Interleukin 7. Exp Hematology 1994; 22: 74–79.
on phenotype, cytokine production and cytotoxic function of 444. Domzig W, Stadler BM, Herberman RB. Interleukin 2 depen-
human monocytes. J Immunol 1993; 151: 6370–6381. dence of human natural killer (LAK) cell activity. J Immunol
423. DeWaal Malefyt R, Haansen J, et al. IL-10 and viral IL-10 1983; 130: 1970.
strongly reduce antigen specific T cell proliferation by diminish- 445. Donaldson DD, Whitters MJ, Fitz LJ, et al. The murine IL-13
ing the antigen presenting capacity of monocytes vial downregu- receptor α2: molecular cloning, characterization and comparison
lation of class II MHC expression. J Exp Med 1991; 174: 915. with murine IL-13 receptor α. J Immunol 1998; 161:
424. Dewhirst FE, Stashenko PP, Mole JE, Tsurumachi T. Purification 2317–2324.
and partial sequence of human osteoclast activating factor: iden- 446. Donepudi M, Quach DD, Mokyr MB. Signaling through CD40
tity with interleukin-1β. J Immunol 1985; 135: 2562–2568. enhances cytotoxic T lymphocyte generation by CD8+ T cells
425. Dhingra K, Sahin A, Emani K, Hortobagyi GN, Estrov Z. from mice bearing large tumors. Cancer Immunol Immunother
Expression of leukemia inhibitory factor and its receptor in breast 1999; 48: 153.
cancer: a potential autocrine and paracrine growth regulatory 447. Donohoe RE, Yang YC, Clark SC. Human P40 T cell growth
mechanism. Breast Cancer Res Treatment 1998; 48: 165–174. factor (interleukin 9) supports erythroid colony formation. Blood
426. Di Carlo E, Comes A, Basso S, et al. The combined action of IL-15 1990; 75: 2271–2275.
and IL-12 gene transfer can induce tumor cell rejection without T 448. Donohue JH, Rosenberg SA. The fate of interleukin-2 after in
and NK cell involvement. J Immunol 2000; 165: 3111–3118. vivo administration. J Immunol 1983; 130: 2203.
427. Di Carlo E, Comes A, Orengo AM, et al. IL-21 induces tumor 449. Donohue PJ, Richards CM, Brown SA, et al. TWEAK is an
rejection by specific CTL and IFN-γ-dependent CXC chemok- endothelial cell growth and chemotactic factor that also potenti-
ines in syngeneic mice. J Immunol 2004; 172: 1540–1547. ates FGF-2 and VEGF-A mitogenic activity. Arterioscler Throm
428. Dickensheets HL, Venkataraman C, Schindler U, Donnelly RP. Vasc Biol 2003; 23: 594–600.
Interferons inhibit activation of STAT6 by interleukin 4 in human 450. Dougall WC, Glaccum M, Charrier K, et al. RANK is essential
monocytes by inducing SOCS-1 gene expression. Proc Natl Acad for osteoclast and lymph node development. Genes Dev 1999;
Sci USA 1998; 96: 10800. 13: 2412.
429. Diegmann J, Junker K, Loncarevic IF, et al. Immune escape from 451. Doull IJ, Lawrence S, Watson M, et al. Allelic association of gene
renal cell carcinoma: CD70 mediates apoptosis in lymphocytes. markers on chromosomes 5q and 11q with atopy and bronchial
Neoplasia 2006; 8: 933–938. hyperresponsiveness. Am J Respir Crit Care Med 1996; 153: 1280.
228 Cytokines

452. Drakes ML, Lu L, Subbotin VM, Thomson AW. In vivo adminis- 473. Durkop H, Latza U, Hummel M, et al. Molecular cloning and
tration of Flt3 ligand markedly stimulates generation of dendritic expression of a new member of the nerve growth factor recep-
cell progenitors from mouse liver. J Immunol 1997; 159: 4268. tor family that is characteristic for Hodgkin’s disease. Cell
453. Drexler HG. Expression of FLT3 receptor and response to FLT3 1992; 68: 421.
ligand by leukemic cells. Leukemia 1996; 10: 588–599. 474. Dutcher JP, Creekmore S, Weiss GR, et al. A Phase II study of
454. Dripps DJ, Brandhuber BJ, Thompson RC, Eisenberg SP. interleukin-2 and lymphokine-activated killer cells in patients
Interleukin-1 (IL-1) receptor antagonist binds to the 80-kDa IL-1 with metastatic malignant melanoma. J Clin Oncol 1989; 7: 477.
receptor but does not initiate IL-1 signal transduction. J Biol 475. Dzierzak EA, Papayannopoulou T, Mulligan RG. Lineage spe-
Chem 1991; 266: 10331–10336. cific expression of a peptide β-globin gene in murine bone mar-
455. Du XX, Doershuk CM, Orazi A, Williams DA. A bone marrow row transplant recipients reconstituted with retrovirus-transduced
stromal derived growth factor, interleukin-11, stimulates recov- stem cells. Nature 1988; 331: 35–41.
ery of small intestinal mucosal cells after cytoablative therapy. 476. Eckenberg R, Moreau J-L, Melnyk O, Theze J. IL-2Rβ Agonist
Blood1994; 83: 33. P1–30 acts in synergy with IL-2, IL-4, IL-9 and IL-15: Biological
456. Du XX, Williams DA. Interleukin-11: review of molecular, cell and Molecular Effects. J Immunol 2000; 165: 4312–4318.
biology and clinical use. Blood 1997; 89: 3897. 477. Eckenberg R, Rose T, Moreau JL, et al. The first helix of IL-2
457. Duan L, Gan H, Arm J, Remold HG. Cytosolic phospholipase A2 folds as a homotetramer, acts as an agonist of the IL-2 receptor β
participates with TNF-α in the induction of apoptosis of human chain and induces lymphokine activated killer cells. J Exp Med
macrophages infected with Mycobacterium tuberculosis H37Ra. 2000; 131: 529.
J Immunol 2001; 166: 7469–7476. 478. Economou JS, McBride WH, Essner R, et al. Tumour necrosis
458. Dubinett SM, Huang M, Lichtenstein A, et al. Tumor necrosis factor production by IL-2-activated macrophages in vitro and in
factor-α plays a central role in interleukin-2 induced pulmonary vivo. Immunology 1989; 67: 514.
vascular leak and lymphocyte accumulation. Cell Immunol 1994; 479. Eder M, Geissler G, Ganser A. IL-3 in the clinic. Stem Cells
157: 170. 1997; 15: 327–333.
459. Dubucquoi S, Desreumaux P, Janin A, et al. Interleukin 5 synthe- 480. Ehrhardt H, Fuda S, Schmid I. TRAIL induced survival and pro-
sis by eosinophils: association with granules and immunoglobu- liferation in cancer cells resistant towards TRAIL-induced apop-
lin-dependent secretion. J Exp Med 1994; 179: 703. tosis mediated by NF-κB. Oncogene 2003; 22: 3842–3852.
460. Duda D, Sunamura M, Lozonschi L, et al. Direct in vitro evi- 481. Einarrson O, Geba GP, Zhu Z, Landry M, Elias JA. Interleukin-11:
dence and in vivo analysis of the antiangiogenesis effects of stimulation in vivo and in vitro by respiratory viruses and induc-
interleukin 12. Cancer Res 2000; 60: 1111. tion of airways hyper responsiveness. J Clin Invest 1996; 97: 915.
461. Dugas B, Renauld J-C, Pene J, et al. Interleukine-9 potentiates the 482. Eisenberg S, Brewer MT, Verderber E, et al. Interleukin-1
interleukine-4-induced immunoglobulin (IgG, IgM and IgE) pro- receptor antagonist is a member of the interleukin-1 gene fam-
duction by normal B lymphocytes. Eur J Immunol 1993; 23: 1687. ily: evolution of a cytokine control mechanism. Proc Natl Acad
462. Dumoutier L, Leemans C, Lejeune D, Kotenko SV, Renauld J-C. Sci USA 1991; 88: 5232.
STAT activation by IL-19, IL-20 and mda-7 through IL-20 recep- 483. Elewaut D, Brossay L, Santee SM, et al. Membrane lymphotoxin
tor complexes of two types. J Immunol 2001; 167: 3545–3549. is required for the development of different populations of NK T
463. Dumoutier L, Lejeune D, Colau D, Renauld J-C. Cloning and cells. J Immunol 2000; 165: 671–679.
characterization of IL-22 binding protein, a natural antagonist of 484. Elias JA, Wu Y, Zheng T, Panettieri R. Cytokine and virus stimu-
IL-10 related T cell-derived inducible factor/IL-22. J Immunol lated airway smooth muscle cells produce interleukin-11 and
2001; 166: 7090–7095. other IL-6-type cytokines. Am J Physiol (Lung Cell Mol Physiol)
464. Dumoutier L, Louahed J, Renauld J-C. Cloning and characteriza- 1997; 273: L648.
tion of IL-10 related T cell derived inducible factor (IL-TIF), a 485. Elias JA, Zheng T, Einerrson O, et al. Epithelial interleukin-11:
novel cytokine structurally related to IL-10 and inducible by regulation by cytokines, respiratory syncytial virus and retinoic
IL-9. J Immunol 2000; 164: 1814. acid. J Biol Chem 1994; 169: 22261.
465. Dumoutier L, Van Roost E, Ameye G, Michaux L, Renauld J-C. 486. Elias JA, Zheng T, Whiting NL, et al. Interleukin-1 and trans-
IL-TIL/IL-22: genomic organization and mapping of the human forming growth factor β regulation of fibroblast derived inter-
and mouse genes. Genes Immun 2000; 1: 488. leukin-11. J Immunol 1994; 152: 2421.
466. Dumoutier L, Van Roost E, Colau D, Renauld J-C. Human inter- 487. Eliopoulos A, Stack GM, Dawson CW, et al. Epstein-Barr virus
leukin-10-related T cell-derived inducible factor: molecular clon- encoded LMP1 and CD40 mediate IL-6 production in epithelial
ing and functional characterization as a hepatocyte-stimulating cells via an NF-κB pathway involving tumor necrosis factor
factor. Proc Natl Acad Sci USA 2000c; 97: 10144–10149. receptor-associated factors. Oncogene 1997; 14: 2899.
467. Dunbar CE, Kohn DB, Schiffman R, et al. Retroviral transfer of 488. Ellerhorst JA, Prieto VG, Ekmekcioglu S, et al. Loss of MDA-7
the cerebrosidase gene into CD34+ cells from patients with expression with progression of melanoma. J Clin Oncol 2002;
Gaucher disease: in vivo detection of transduced cells without 20: 1069–1074.
myeloablation. Hum Gene Ther 1998; 9: 2629. 489. Ellis TM, Simms PE, Slivnick DJ, Jack H-M, Fisher RI. CD30 is
468. Duncan MR, Berman B. Stimulation of collagen and gly- a signal-transducing molecule which defines a subset of human
cosaminoglycan production in cultured human adult dermal activated CD45RO+ T cells. J Immunol 1993; 151: 2380.
fibroblasts by recombinant human interleukin-6. Invest Dermatol 490. Emery JG, McDonnell P, Burke MB, et al. Osteoprotegerin is a
1991; 97: 686. receptor for the cytotoxic ligand TRAIL. J Biol Chem 1998;
469. Duncan MR, Hasan A, Berman B. Oncostatin M stimulates col- 273: 14363.
lagen and glycosaminoglycan production by cultured normal 491. Emmel EA, Verweij CL, Durand DB, et al. Cyclosporin A spe-
dermal fibroblasts: insensitivity of scleroderma and keloid fibro- cifically inhibits function of nuclear proteins involved in T cell
blasts. J Invest Dermatol 1995; 104: 128. activation. Science 1989; 246: 1617.
470. Durie FH, Aruffo A, Ledbetter J, et al. Antibody to the ligand of 492. Endo TA, Mashuhara M, Yokuchi M, et al. A new protein con-
CD40, gp39, blocks the occurrence of the acute and chronic taining as SF2 domain that inhibits JAK kinases. Nature 1997;
forms of graft-vs-host disease. J Clin Invest 1994; 94: 1333. 387: 921.
471. Durie FH, Fava RA, Foy TM, Aruffo A, Ledbetter JA, Noelle RJ. 493. Engelman H, Novick D, Wallach D. Two tumor necrosis factor
Prevention of collagen-induced arthritis with one antibody to binding proteins purified from human urine. J Biol Chem 1990;
gp39, the ligand of CD40. Science 1993; 261: 1328. 265: 1531.
472. Durkop H, Latza U, Himmelreich P, Stein H. Expression of the 494. Enk CD, Sredni D, Blauvelt A, Katz SI. Induction of IL-10 gene
human OX40 (hOX40) antigen in normal and neoplastic tissues. expression in human keratinocytes by UVB exposure in vivo and
Br J Haematol 1995; 91: 927. in vitro. J Immunol 1995; 154: 4851–4856.
Walter M. Lewko and Robert K. Oldham 229

495. Erickson SL, Sauvage FJD, Kikly K, et al. Decreased sensitivity 517. Faust J, Lacey DL, Hunt P, et al. Osteoclast markers accumulate
to tumor necrosis factor but normal T cell development in TNF- on cells developing from the peripheral blood mononuclear pre-
receptor-2-deficient mice. Nature 1994; 372: 560. cursors. J Cell Biochem 1999; 72: 67.
496. Ericson SG, Zhao Y, Gao H, et al. Interleukin-6 production by 518. Favre C, Saeland S, Caux C, et al. Interleukin-4 has growth
human neutrophils after Fc receptor cross-linking or exposure to promoting activity on cord blood cells. Blood 1990; 75: 67–73.
granulocyte colony stimulating factor. Blood 1998; 91: 2099–2107. 519. Favre-Felix N, Fromentin A, Hammann A, et al. The tumor
497. Ernst E. Plasma fibrinogen-an independent cardiovascular risk counterattack revisited: colon cancer cells do not induce T cell
factor. J Int Med 1990; 227: 365. apoptosis via the Fas (CD95, Apo-1) pathway. J Immunol 2000;
498. Escary J-L, Perreau J, Dumenil D, Ezine S, Brulet P. Leukemia 164: 5023–5027.
inhibitory factor is necessary for maintenance of hematopoietic 520. Fearon DT, Locksley RM. The instructive role of innate immu-
stem cells and thymic stimulation. Nature 1993; 363: 361. nity in the acquired immune response. Science 1996; 272: 50.
499. Esche C, Subbotin VM, Maliszewski C, Lotze MT, Shurin MR. 521. Fehninger TA, Shah MH, et al. Differential cytokine and
FLT3 ligand administration inhibits tumor growth in murine chemokine expression by human NK cells following activa-
melanoma and lymphoma. Cancer Res 1998; 58: 380–383. tion with IL-18 or IL-15 in combination with IL-12: implica-
500. Estrov Z, Samal B, Lapushin R, et al. Leukemia inhibitory factor tions for the innate immune response. J Immunology 1999;
binds to breast cancer cells and stimulates their proliferation. 162: 4511.
J Interferon Cytokine Res 1995; 15: 905–913. 522. Feldman ER, Creagan ET, Schaid DJ, Ahmann DL. Phase II trial
501. Ettinger R, Sims GP, Fairhurst AM, et al. IL-21 induces differen- of recombinant tumor necrosis factor in disseminated malignant
tiation of human naive and memory B cells into antibody-secret- melanoma. Am J Clin Oncol 1992; 15: 256–259.
ing plasma cells. J Immunol 2005; 175: 7867–7879. 523. Feng SY, Guo Y, Factor VM, et al. The Fn14 intermediate-early
502. Eugster H, Muller M, Karrer U, et al. Multiple immune abnor- response gene is induced during liver regeneration and highly
malities in tumor necrosis factor and lymphotoxin-α double defi- expressed in both human and murine hepatocellular carcinomas.
cient mice. Int Immunol 1996; 8: 23. Am J Pathol 2000; 156: 1253–1261.
503. Eustace D, Han X, Gooding R, Rowbottom A, Riches P, 524. Feng Y, Broder CC, Kennedy PE, Berger EA. HIV entry cofactor:
Heyderman E. Interleukin-6 functions as an autocrine growth fac- functional cDNA cloning of a seven-transmembrane, G protein-
tor in cervical carcinomas in vitro. Gynecol Oncol 1993, 50: 15. coupled receptor. Science 1996; 272: 872.
504. Evans CH, Watkins SC, Stefanovic-Racic M. Nitric oxide and 525. Fenton MJ, Buras JA, Donnelly RP. IL-4 reciprocally regulates
cartilage metabolism. Methods Enzymol 1996; 269: 75. IL-1 and IL-1 receptor antagonist expression in human mono-
505. Evans CH. Lymphotoxin – An immunologic hormone with cytes. J Immunol 1992; 149: 1283.
anti-carcinogenic and anti-tumor activity. Cancer Immunol 526. Fernandez Botran R, Sanders VM, Oliver KG, Vitetta ES.
Immunother 1982; 12: 181. Evidence that soluble interleukin-4 receptors may act as transport
506. Facon T, Harousseau JL, Maloisel F, et al. Stem cell factor in proteins. J Exp Me 1991; 174: 673–681.
combination with filgastim after chemotherapy improves periph- 527. Fernandez-Botran R. Soluble cytokine receptors and their role in
eral blood progenitor cell yield and reduces apheresis require- immunoregulation. FASEB 1991; 5: 2567.
ments in multiple myeloma patients: a ramdomized controlled 528. Ferrari D, Chiozzi P, Falzoni S, et al. Extracellular ATP triggers
trial. Blood 1999; 94: 1218–1225. IL-1β release by activating the purinergic P2Z receptor of human
507. Falcone M, Rajan AJ, Bloom BR, Brosman CF. A critical role for macrophages. J Immunol 1997; 159: 1451.
IL-4 in regulating disease severity in experimental allergic 529. Ferrari D, Chiozzi P, Falzoni S, et al. Purinergic modulation of
encephalitis as demonstrated in IL-4-deficient C57BL/6 mice and IL-1β release from microglial cells stimulated with bacterial
BALB/c mice. J Immunol 1998; 160: 4822–4830. endotoxin. J Exp Med 1997; 185: 579.
508. Fallon PG, Ballantyne SJ, Mangan NE, et al. Identification of an 530. Fibbe WE, van der Meer JWM, et al. A single low dose of human
Interleukin-25-dependent cell population that provides IL-4, recombinant interleukin 1 accelerates the recovery of neutrophils
IL-5, and IL-13 at the onset of helminth expulsion. J Exp Med in mice with cyclophosphamide-induced neutropenia. Exp
2006; 203: 1105–1116. Hematol 1989; 17: 805.
509. Fan Z, Yu P, Wang Y, et al. NK-cell activation by LIGHT triggers 531. Fichtner-Feigl S, Strober W, Kawakami K, et al. IL-13 signaling
tumor specific CD8+ T cell immunity to reject established through the IL-13α(2) receptor is involved in induction of trans-
tumors. Blood 2006; 107: 1342–1351. forming growth factor β(1) production and fibrosis. Nature Med
510. Fanger NA, Maliszewski CR, Schooley K, Griffith TS. Human 2006; 12: 99–106.
dendritic cells mediate apoptosis via tumor necrosis factor-re- 532. Fickenscher H, Pirzer H. Interleukin-26. Int Immunopharmacol
lated apoptosis inducing ligand (TRAIL). J Exp Med 1999; 2004; 4: 609–613.
190: 1155. 533. Finbloom DS and Winestock KD. IL-10 induces the tyrosine
511. Fanslow WC, Clifford KN, Seaman M, et al. Recombinant CD40 phosphorylation of tyk2 and Jak1 and the differential assembly of
ligand exerts potent biologic effects on T cells. J Immunol 1994; STAT1α and STAT3 complexes in human T cells and monocytes.
152: 4262. J Immunol 1995; 155: 1079–1090.
512. Fantuzzi G, Reed DA, Dinarello CA. IL-12-induced IFNγ is 534. Finke JH, Rayman P, Hart L, et al. Characterization of tumor
dependent on caspase-1 processing of the IL-18 precursor. J Clin infiltrating lymphocyte subsets from human renal cell carcinoma:
Invest 1999; 104: 761. specific reactivity defined by cytotoxicity interferon γ secretion
513. Faour WH, Mancini A, He OW Di Batista JA. T-cell derived IL-17 and proliferation. J Immunother with Emphasis on Tumor
regulates the levels and stability of cyclooxygenase-2 (COX-2) Immunology 1994; 15: 91–104.
mRNA through restricted activation of the p38 mitogen-activated 535. Fiorentino DF, Bond MW, Mosmann TR. Two types of mouse T
protein kinase cascade. J Biol Chem 2003; 278: 26897–26907. helper IV: Th2 clones secrete a factor that inhibits cytokine pro-
514. Farrar J, Hilfiker M, Johnson B, et al. Identification of a T-cell duction by Th1 clones. J Exp Med 1989; 170: 2081–2095.
derived B-cell growth factor distinct from interleukin-2. J Exp 536. Fiorentino DF, Zlotnik A, Mosmann TR, et al. IL-10 inhibits
Med 1982; 155: 914–923. cytokine production by activated macrophages. J Immunol 1991;
515. Faulkner H, Humphries N, Renauld J-C, Van Snick J, Grencis R. 147: 3815.
Interleukin-9 is involved in host protective immunity to intestinal 537. Fiorentino DF, Zlotnik A, Viera P, et al. IL-10 acts on the antigen
nematode infection. Eur J Immunol 1997; 27: 2536. presenting cell to inhibit cytokine production by TH1 cells. J
516. Faure E, Thomas L, Xu H, et al. Bacterial lipopolysaccharide and Immunol 1991; 146: 3444–3451.
IFN-γ induce Toll-like receptor 2 and Toll-like receptor 4 expres- 538. Firestein GS, Boyle DL, Yu C, et al. Synovial interleukin-1
sion in endothelial cells: role of NF-κB activation. J Immunol receptor antagonist and interleukin-1 balance in rheumatoid
2001; 166: 2018–2024. arthritis. Arthritis Rheum 1994; 37: 644–652.
230 Cytokines

539. Fischer M, Harvima IT, Carvalho RF, et al. Mast cell CD30 560. Franco AV, Zhang XD, Van Berkel E, et al. The role of NF-κB
ligand is upregulated in cutaneous inflammation and mediates in TNF-related apoptosis-inducing ligand (TRAIL)-induced
degranulation-independent chemokine secretion. J Clin Invest apoptosis of melanoma cells. J Immunol 2001; 166:
2006; 116: 2748–2756. 5337–5345.
540. Fisher PB. Is mda-7/IL-24 a “magic bullet” for cancer? Cancer 561. Frank MB, Watson J, Gillis S. Biochemical and biologic charac-
Res 2005; 65: 10128–10138. terization of lymphocyte regulatory molecules. VIII. Purification
541. Fisher RI, Coltman CA, Doroshow JH, et al. Metastatic renal of interleukin-2 from a human T-cell leukemia. J Immunol 1981;
cancer treated with interleukin-2 and lymphokine activated killer 127: 2361.
(LAK) cells. A phase II clinical trial. Ann Intern Med 1988; 108: 562. Franklin WA, Glaspy J, Pflaumer SM, et al. Incidence of tumor
518–523. cell contamination in leukapherisis products of breast cancer
542. Fitzpatrick LA, Severson A, Edwards WD, et al. Diffuse calcifi- patients mobilized with stem cell factor and granulocyte colony
cation in human coronary arteries. Association of osteopontin stimulating factor (G-CSF) or G-CSF alone. Blood 1999; 94:
with atherosclerosis. J Clin Invest 1994; 94: 1597–1604. 340–347.
543. Flanagan JG, Chan DC, Leder P. Transmembrane form of the kit 563. Fransen L, Van de Hayden J, Ruysshaert R, et al. Recombinant
growth factor is determined by alternative splicing and is missing tumor necrosis factor: its effects and its synergism with inter-
in the S1d mutant. Cell 1991; 64: 1025–1035. feron γ on a variety of normal and transformed human cell lines.
544. Flanagan WM, Corthesy B, Bram RJ, Crabtree GR. Nuclear Eur J Cancer and Clin Oncol 1986; 22: 419–426.
association of a T cell transcription factor blocked by FK-506 and 564. Fraser CC, Thacker JD, Hogge DE, et al. Alterations in lym-
cyclosporin A. Nature 1991; 352: 803. phopoiesis after hematopoietic reconstitution with IL-7 virus
545. Flesch IEA and Kaufman SHE. Activation of bacteriostatic mac- infected bone marrow. J Immunol 1993; 151: 2409–2418.
rophage functions by γ interferon, interleukin-4 and tumor necro- 565. Freeman GJ, Boussiotis VA, Anunmanthan A, et al. B7–1 and
sis factor. Infect Immun 1990; 58: 2675. B7–2 do not deliver identical costimulatory signals, since B7–2
546. Fluckiger A-C, Durand I, Banchereau J. Interleukin-10 induces but not B7–1 preferentially costimulates initial production of
apoptotic death of B-chronic lymphocytic leukemia cells. J Exp IL-4. Immunity 1995; 2: 523.
Med 1994; 179: 91. 566. Freisen C, Herr I, Krammer PH, Debatin KM. Involvement of
547. Fluckiger A-C, Garrone P, Durand I, Galizi J-P, Banchereau J. CD95 (APO-1/Fas) receptor/ligand system in drug-induced
Interleukin-10 (IL-10) upregulates functional high affinity IL-2 apoptosis in leukemia cells. Nature Med 1996.
receptors on normal and leukemic B lymphocytes. J Exp Med 567. French RR, Chan HTC, Tutt AL, Glennie MJ. CD40 antibody
1993; 178: 1473. evokes a cytotoxic T-cell response that eradicates lymphoma and
548. Foey AD, Parry SL, Williams LM, et al. Regulation of monocyte bypasses T-cell help. Nat Med 1999; 5: 548.
IL-10 synthesis by endogenous IL-1 and TNF-α: Role of the p38 568. Friend SL, Hosier S, Nelson A, Foxworthe D, Williams DE, Farr
and p42/44 mitogen-activated protein kinases. J Immunol 1998; A. A thymic stromal cell line supports in vitro development of
160: 920–928. surface IgM+ B cells and produces a novel growth factor affect-
549. Fong L, Hou Y, Rivas A, Benike C, et al. Altered peptide ligand ing B and T lineage cells. Exp Hematol 1994; 22: 321.
vaccination with Flt3 ligand expanded cells for tumor immuno- 569. Frishman J, Long B, Knospe W, et al. Genes for interleukin 7 are
therapy. Proc Natl Acad Sci USA 2001; 98: 8809–8814. translated in leukemia cell subsets of individuals with chronic
550. Fong Y, Tracey KJ, Moldawer LL, et al. Antibodies to cachectin/ lymphocytic leukemia. J Exp Med 1993; 177: 955–964.
tumor necrosis factor reduce interleukin-1B and interleukin 6 570. Frohlich A, Marsland BJ, Sonderegger I, et al. IL-2 receptor sig-
appearance during lethal bacteremia. J Exp Med 1989; 170: 1627. naling is integral to the development of Th2 effector responses in
551. Fontana A, Kristensen F, Dubs R, et al. Production of prostaglan- vitro. Blood 2006.
din E and interleukin-1 like factor by cultured astrocytes and CG 571. Fu Y-X, Huang G, Wang Y, Chaplin DD. Lymphotoxin-α depen-
glioma cells. J Immunol 1982; 129: 2413–2419. dent spleen microenvironment supports the generation of mem-
552. Ford R, Tamayo A, Martin B, et al. Identification of B-cell growth ory B cells and is required for their subsequent antigen-induced
factors (interleukin-14; high molecular weight-B-cell growth fac- activation. J Immunol 2000; 164: 2508–2514.
tors) in patients with aggressive B-cell lymphomas. Blood 1995; 572. Fujita T, Matsumoto Y, Hirai I, et al. Immunosuppressive effect
86: 283–293. on T cell activation by interleukin 16-cDNA-transfected human
553. Ford RJ, Tamayo A, Marker B, Ambrus JL. IL-14 and growth squamous cell line. Cell Immunol 2000; 202: 54–60.
control in human B cell lymphomas. FASEB J 1992; 8: A1021. 573. Fukao T, Matsuda S, Koyasu S. Synergistic effects of IL-4 and
554. Foss FM, Koc Y, Stetler-Stevenson MA, et al. Costimulation of IL-18 on IL-12-dependent IFN-γ production by dendritic cells. J
cutaneous T cell lymphoma cells by Interleukin 7 and interleukin Immunol 2000; 164: 64.
2: potential autocrine and paracrine effectors in the Sezary syn- 574. Fukushi J, Ono M, Morikawa W, Iwamoto Y, Kuwano M. The
drome. J Clin Oncol 1994; 12: 326–335. activity of soluble VCAM-1 in angiogenesis stimulated by IL-4
555. Fossiez F, Djossou O, Chomarat P, et al. T cell interleukin-17 and IL-13. J Immunol 2000; 165: 2818–2823.
induces stromal cells to produce proinflammatory and hematopoi- 575. Funakoshi S, Longo DL, Beckwith M, et al. Inhibition of
etic cytokines. J Exp Med 1996; 183: 2593. human B cell lymphoma growth by CD40 stimulation. Blood
556. Foxwell BM, Beadling C, Gushin D, Kerr I, Cantrell D. 1994; 83: 2787.
Interleukin-7 can induce the activation of Jak1, Jak3, and STAT5 576. Furman WL, Luo X, Fairclough D, et al. Phase I trial of subcuta-
proteins in murine T cells. Eur J Immunol 1995; 25: 3041–3046. neous interleukin-1α in children with malignant solid tumors.
557. Foxwell BM, Willcocks JL, Taylor-Fishwick DA, et al. Inhibition Med Pediatr Oncol 1997; 28: 444–450.
of activation induced changed in the structure of the T cell inter- 577. Furukawa J, Hara I, Nagai H, et al. Interleukin-21 gene transfec-
leukin-7 receptor by cyclosporin A and FK506. Eur J Immunol tion into mouse bladder cancer cells, results in tumor rejection
1993; 23: 85–89. through the cytotoxic T lymphocyte response. J Urol 2006; 176:
558. Foxwell BML, Taylor-Fishwick DA, Simon JL, et al. Activation 1198–1203.
induced changes in expression and structure of the IL-7 receptor 578. Gabbianelli M, Pelosi E, Montesoro E, et al. Multilevel effects of
in human T cells. Int Immunol 1992; 4: 277. Flt-3 ligand on human hematopoiesis: expansion of putative stem
559. Fraker DL, Alexander HR, Andrich M, Rosenberg SA. Treatment cells and proliferation of granulomonocytic progenitors/mono-
of patients with melanoma of the extremity using hyperthermic cytic precursors. Blood 1995; 86: 1661–1670.
isolated limb perfusion with melphalan, tumor necrosis factor, 579. Gadient R, Patterson PH. Leukemia inhibitory factor, interleu-
and interferon γ: results of a tumor necrosis factor dose escalation kin-6 and other cytokines using the GP130 transducing receptor:
study. J Clin Oncol 1996; 26: 404–410. roles in inflammation and injury. Stem Cells 1999; 17: 127.
Walter M. Lewko and Robert K. Oldham 231

580. Gagari E, Tsai M, Lantz CS, Fox LG, Galli SJ. Differential release 602. Gearing DP, Comeau MR, Friend DJ, et al. The IL-6 signal trans-
of mast cell interleukin-6 via c-kit. Blood 1997; 89: 2654. ducer gp130: an oncostatin M receptor and an affinity converter
581. Gajewski TF, Fallarino F, Ashikari A, Sherman M. Immunization for the LIF receptor. Science 1992; 225: 1434.
of HLA-A2+ patients with MAGE-3 or Melan A peptide-pulsed 603. Gearing DP, Cosman D. Homology of the p40 subunit of natural
autologous peripheral blood mononuclear cells plus recombinant killer cell stimulatory factor (NKSF) with the extracellular
human interleukin 12. Clin Cancer Res 2001; 7: 895s–901s. domain of the interleukin-6 receptor. Cell 1991; 66: 9.
582. Gallagher G, Dickensheets H, Eskdale J, et al. Cloning, expression 604. Gearing DP, Gough NM, King JA, et al. Molecular cloning and
and initial characterization of interleukin-19, a novel homologue of expression of cDNA encoding a murine myeloid leukemia inhibi-
human interleukin-10 (IL-10). Genes Immun 2000; 1: 442–450. tory factor (LIF). EMBO 1987: 6: 3995–4002.
583. Gallagher G, Eskdale J, Jordan W, et al. Human interleukin-19 605. Gedrich RW, Gilfillan MC, Druckett CS, Van Dongen LW, Thompson
and its receptor: a potential role in the induction of Th2 responses. CB. CD30 contains two binding sites with different specificities for
Int Immunopharmacol 2004; 4: 615–626. members of the tumor necrosis factor receptor-associated factor
584. Gallagher G, Stimson WH, Findlay J, et al. Interleukin 6 enhances family of signal transducing proteins. Biol Chem 1996; 271: 12852.
the induction of human lymphokine activated killer cells. Cancer 606. Gee MS, Koch CJ, Evans SM, et al. Hypoxia-mediated apoptosis
Immunol Immunother 1990; 31: 49–52. from angiogenesis inhibition underlies tumor control by recom-
585. Galy AHM and Spits H. CD40 is functionally expressed on thy- binant interleukin 12. Cancer Res 1999; 59: 4882.
mic epithelial cells. J Immunol 1992; 149: 775. 607. Geiger T, Tobin H, Consenti-Vargas A, et al. Neutralization of
586. Gamble JR, Harlan JM, Klebamoff SJ, Vadas MA. Stimulation of interleukin-1β in vivo with a monoclonal antibody alleviates
the adherence of neutrophils to umbilical vein endothelium by collagen-induced arthritis in DBA/1 mice and prevents the asso-
human recombinant tumor necrosis factor. Proc Natl Acad Sci ciated acute-phase response. Clin Exp Rheumatol 1993; 11: 515.
USA 1985; 82: 8667. 608. Geissinger E, Weisser C, Fisher P, et al. Autocrine stimulation by
587. Gansbacher B, Bannerji R, Daniels B, et al. Retroviral vector osteopontin contributes to antiapoptotic signaling of melanocytes
mediated γ-interferon gene transfer into tumor cells generates in dermal collagen. Cancer Res 2002; 62: 4802–4828.
potent and long lasting antitumor immunity. Cancer Res 1990; 609. Geissler EN, Ryan MA, Housman DE. The dominant-white spot-
50: 7820–7825. ting (W) locus of the mouse encodes the c-kit protooncogene.
588. Gansbacher B, Zier K, Daniels B, et al. Interleukin 2 gene trans- Cell 1988; 55: 185.
fer into tumor cells abrogates tumorigenicity and induces protec- 610. Geisterfer M, Richards C, Baumann M, et al. Regulation of IL-6
tive immunity. Exp Med 1990; 172: 1217–1224. and the hepatic IL-6 receptor in acute inflammation in vivo.
589. Ganser A, Lindemann A, Spielt G, et al. Effects of recombinant Cytokine 1993; 5: 1–7.
human interleukin-3 in patients with normal hematopoiesis and 611. Gemlo BT, Palladino MA Jr, Jaffe HS, et al. Circulating cytok-
in patients with bone marrow failure. Blood 1990; 76: 666–676. ines in patients with metastatic cancer treated with recombinant
590. Gao JJ, Xue Q, Papasian CJ, Morrison DC. Bacterial DNA and interleukin 2 and lymphokine-activated killer cells. Cancer Res
lipopolysaccharide induce synergistic production of TNF-α 1988; 48: 5864.
through a post-transcriptional mechanism. J Immunol 2001; 166: 612. Georganas C, Liu H, Perlman H, et al. Regulation of IL-6 and
6855–6860. IL-8 expression in rheumatoid arthritis synovial fibroblasts: the
591. Garcia VE, Jullien ED, Song M, et al. IL-15 enhances the dominant role for NF-κB but not C/EBPβ or c-Jun. J Immunol
response of human γδ T cells to non-peptide microbial antigens. 2000; 165: 7199–7206.
J Immunol 1998; 160: 4322. 613. Gerard C, Bruyns C, Marchant A, et al. Interleukin-10 reduces
592. Garvy BA Riley RL. IFN γ abrogates IL-7 dependent prolifera- the release of tumor necrosis factor and prevents lethality in
tion in per-B cells coinciding with onset of apoptosis. Immunology experimental endotoxemia. J Exp Med 1993; 177: 547.
1994; 81: 381–388. 614. Gerli R, Pitzalis C, Bistoni O, et al. CD30+ T cells in rheuma-
593. Gaspal FM, Kim MY, McConnell FM, et al. Mice deficient in toid synovitis: mechanisms of recruitment and functional role.
OX40 and CD30 signals lack memory antibody responses J Immunol 2000; 164: 4399.
because of deficient CD4 T cell memory. J Immunol 2005; 174: 615. Gerritse K, Laman JD, Noelle RJ, et al. CD40–CD40 ligand
3891–3896. interactions in allergic encephalomyelitis and multiple sclerosis.
594. Gasparetto C, Laver J, Abboud M, et al. Effect of interleukin-1 on Proc Natl Acad Sci USA 1996; 93: 2499.
hematopoietic progenitors: evidence of stimulatory and inhibi- 616. Gershenwald JE, Fong YM, Fahey TJ, et al. Interleukin 1 recep-
tory activities in a primate model. Blood 1989; 74: 547. tor blockade attenuates the host inflammatory response. Proc
595. Gately MK, Caravajal DM, Connaughton SE, et al. Interleukin-12 Natl Acad Sci USA 1990; 87: 4966.
antagonist activity of mouse interleukin-12 p40 homodimer in 617. Gery I, Gershon RK, Waksman BH. Potentiation of the
vitro and in vivo. Ann NY Acad Sci 1996; 795: 1. T-lymphocyte response to mitogens. II. The cellular source of
596. Gately MK, Wilson DE, Wong HL. Synergy between recombi- potentiating mediators. J Exp Med 1972; 136: 143.
nant Interleukin 2 and interleukin 2 depleted lymphokine-con- 618. Ghayur T, Banerjee S, Hugunin M, et al. Caspase-1 processes
taining supernatants in facilitating allogeneic human cytolytic T IFN-γ inducing factor and regulates LPS-induced IFN γ produc-
lymphocyte responses in vitro. J Immunol 1986; 136: 1274. tion. Nature 1997: 386: 619.
597. Gately MK, Wolitzky AG, Quinn PM, Chizzonite R. Regulation 619. Ghezzi P, Saccardo B, Villa P, et al. Role of interleukin-1 in the
of human cytolytic lymphocyte responses by interleukin 12. depression of liver drug metabolism by endotoxin. Infect Immun
Cellular Immunology 1992; 143: 127–142. 1986; 54: 837–840.
598. Gattei V, Aldinucci D, Rossi FM, et al. CD30 ligand (CD30L)- 620. Giavazzi R, Garofalo A, Bani MR, et al. Interleukin 1-induced
expressing acute myeloid leukemias: a new model of paracrine augmentation of experimental metastases from a human mela-
interactions for the regulation of blast cell proliferation. Leuk noma in nude mice. Cancer Res 1991; 50: 4771.
Lymphoma 1999; 35: 21. 621. Gilfillan MC, Noel PJ, Podack ER, Reiner SL, Thompson CB.
599. Gattei V, Degan M, Glogini A, et al. CD30 ligand is frequently Expression of the costimulatory receptor CD30 is regulated by
expressed in hematopoietic malignancies of myeloid and lym- both CD28 and cytokines. J Immunol 1998; 160: 2180–2187.
phoid origin. Blood 1997; 89: 2048–2059. 622. Gimbaud R, Abitbol V, Bertrand V, et al. Leukemia inhibitory
600. Gazzinelli RT, Oswald IP, James SL, Sher A. IL-10 inhibits para- factor involvement in human ulcerative colitis and its potential
site killing and nitrogen oxide production by IFNγ activated mac- role in malignant course. Eur Cytokine Netw 1998; 9: 607–612.
rophages. J Immunol 1992; 148: 1792. 623. Gimbrone MA, Obin MS, Brock AF, et al. Endothelial interleu-
601. Gearing DP, Bruce AG. Oncostatin M binds the high affinity leu- kin 8. a novel inhibitor of leukocyte endothelial interactions.
kemia inhibitory factor receptor. New Biol 1992; 4: 61–65. Science 1989; 246: 1601.
232 Cytokines

624. Giovarelli M, Musiani P, Modesti A, et al. Local release of IL-10 644. Gordon JR, Galli SJ. Promotion of mouse collagen gene expres-
by transfected mouse mammary adenocarcinoma cells does not sion by mast cells stimulated via the Fc epsilon RI: role of mast
suppress but enhances antitumor reaction and elicits a strong cell derived transforming growth factor β and tumor necrosis
cytotoxic lymphocyte and antibody dependent immune memory. factor α. J Exp Med 1994; 180: 2027–2037.
J Immunol 1995; 155: 3112–3123. 645. Gordon MS, McCaskill-Stevens WJ, et al. A phase I trial of
625. Girasole G, Passeri G, Jilka RL, Manolagas SC. Interleukin-11: a recombinant human interleukin-11 (neumega rhIL-11 growth
new cytokine critical for osteoclast development. J Clin Invest factor) in women with breast cancer receiving chemotherapy.
1994; 93: 1516. Blood 1996; 87: 3615–3624.
626. Giri JG, Adhieh M, Eisenman J, et al. Utilization of the β and γ 646. Gorierly S, Vincart B, Stordeur P, et al. Deficient IL-12 (p35)
chains of the IL-2 receptor by the novel cytokine IL-15. EMBO J gene expression by dendritic cells derived from neonate mono-
1994; 13: 2822–2830. cytes. J Immunol 2001; 166: 2141–2146.
627. Giri JG, Kumaki S, Ahdieh M, et al. Identification and cloning of 647. Grabstein K, Eisenman J, Shanebeck K, et al. Cloning of a T cell
a novel IL-15 binding protein that is structurally related to the α growth factor that interacts with the B-chain of the interleukin 2
chain of the IL-2 receptor. EMBO J 1995; 14: 2354–3663. receptor. Science 1994; 264: 965–968.
628. Giri JG, Newton RC, Horuk R. Identification of a soluble inter- 648. Grabstein KH, Namen AE, Shanebeck K, et al. Regulation of
leukin-1 binding protein in cell free supernatants: evidence for human T cell proliferation by IL-7. J Immunol 1990; 144:
soluble interleukin-1 receptor. J Biol Chem 1990; 265: 17416. 3015–3020.
629. Go NF, Castle BE, Barrett R, et al. Interleukin 10 (IL-10) a novel 649. Grabstein KH, Park LS, Morrissey JP, et al. Regulation of T cell
B cell stimulatory factor: unresponsiveness of X chromosome proliferation by T cell stimulating factor-1.J Immunol 1987; 139:
linked immunodeficiency B cells. J Exp Med 1990; 172: 1148–1153.
1625–1631. 650. Grabstein KH, Waldschmidt TJ, Finkelman FD, et al. Inhibition
630. Goda C, Kanaji T, Kanaji S, et al. Involvement of IL-32 in acti- of murine B and T lymphopoiesis in vivo by an anti-interleukin 7
vation-induced cells death in T cells. Int Immunol 2006; 18: monoclonal antibody. J Exp Med 1993; 178: 257.
233–240. 651. Graca L, Honye K, Adams E, Cobbold SP, Waldmann H. Anti-
631. Godfraind C, Louahed J, Faulkner H, et al. Intraepithelial infiltra- CD154 therapeutic antibodies induce infectious transplant toler-
tion by mast cells with both connective tissue-type and mucosal- ance. J Immunol 2000; 165: 4783–4786.
type characteristics in gut, trachea, and kidneys of IL-9 transgenic 652. Gracie JA, Forsey RJ, Chan WL, et al. A proinflammatory role
mice. J Immunol 1998; 160: 3989. for IL-18 in rheumatoid arthritis. J Clin Invest 1999; 104: 1393.
632. Godfrey WR, Fagnoni FF, Harara MA, et al. Identification of a 653. Graf D, Muller S, Korthauer U, et al. A soluble form of TRAP
human Ox-40 ligand, a costimulator of CD4+ T cells with homol- (CD40 ligand) is rapidly released after T cell activation. Eur J
ogy to tumor necrosis factor. J Exp Med 1994; 180: 757. Immunol 1995; 25: 1749.
633. Goebeler M, Schnarr A, Toksoy A, et al. Interleukin-13 selec- 654. Gramaglia I, Jember A, Pippig SD, et al. Ox40 costimulatory
tively induces monocyte chemoattractant protein-1 synthesis and receptor determines the development of CD4 memory by regulat-
secretion by human endothelial cells: involvement of IL-4Rα and ing primary clonal expansion. J Immunol 2000; 165: 3043–3050.
stat6 phosphorylation. Immunobiology 1997; 91: 450. 655. Gramaglia I, Weinberg AD, Lemon M, Croft M. OX-40 ligand: a
634. Goldfeld AE, McCaffrey PG, Strominger JL, Rao A. Identification potent costimulatory molecule for sustaining primary CD4 T cell
of a novel cyclosporin-sensitive element in the human tumor responses. J Immunol 1998; 161: 6510–6517.
necrosis factor α gene promoter. J Exp Med 1993; 178: 1365. 656. Granelli-Piperno A, Nolan P Inaba K, Steinman RM. The effect
635. Gollob JA, Mier JW, Veenstra K, et al. Phase I trial of twice- of immunosuppressive agents on the induction of nuclear factors
weekly intravenous interleukin 12 in patients with metastatic that bind to sites on the interleukin-2 promoter. J Exp Med 1990;
renal cell cancer or malignant melanoma: ability to maintain 172: 1869.
IFN-γ induction is associated with clinical response. Clin Cancer 657. Granger GA, Williams TW. Lymphocyte cytotoxicity in vitro:
Res 2000; 6: 1678–1692. Activation and release of a cytotoxic factor. Nature 1968;
636. Golumbek PT, Lazenby AJ, Levitsky HI, et al. Treatment of 218: 1253.
established renal caner by tumor cells engineered to secrete inter- 658. Granowitz EV, Clark BD, Mancilla J, Dinarello CA. Interleukin-1
leukin-4. Science 1991; 254: 713–716. receptor antagonist competitively inhibits the binding of interleu-
637. Gonzalez-Hernandez JA, Ehrhart-Bornstein M, et al. Human kin-1 to type II interleukin-1 receptor. J Biol Chem 1991; 266:
adrenal cells express tumor necrosis factor messenger ribonucleic 14147–14150.
acid: evidence for paracrine control of adrenal function. J Clin 659. Gray PW, Barret K, Chantry D, Turner M, Feldman M. Cloning
Endo Metab 1996; 81: 807. of human tumor necrosis factor (TNF) receptor cDNA: expres-
638. Goodwin RG, Alderson MR, Smith CA, et al. Molecular and bio- sion of recombinant soluble TNF-binding protein. Proc Natl
chemical characterization of a ligand for CD27 defines a new Acad Sci USA 1990; 87: 7380–7384.
family of cytokines with homology to tumor necrosis factor. Cell 660. Green PM, Noel PJ, Sperling AI, et al. Absence of B7-dependent
1993; 73: 447. responses in CD28-deficient mice. Immunity1994; 1: 501.
639. Goodwin RG, Din WS, Davis-Smith T, et al. Molecular cloning 661. Green S, Dobrjansky A, Chiasson MA, et al. Corynebacterium
of a ligand for the inducible T cell gene 4-1BB: a member of an parvum as the priming agent in the production of tumor necrosis
emerging family of cytokines with homology to tumor necrosis factor in the mouse. J Natl Cancer Inst 1977; 59: 1519.
factor. Eur J Immunol 1993; 23: 2631–2641. 662. Greene CM, Meachery G, Taggart CC, et al. Role of IL-18 in
640. Goodwin RG, Friend D, Ziegler SF, et al. Cloning of the human CD4+ T lymphocyte activation in sarcoidosis. J Immunol 2000,
and murine interleukin-7 receptors: Demonstration of a soluble 165; 4718.
form and homology to a new receptor superfamily. Cell 1990; 663. Greenfelder S, Nunes AP, Kwee L, Labow M, Chizzonite RA, Ju G.
60: 941. Molecular cloning and characterization of a second subunit of the
641. Goodwin RG, Lupton GS, Schmierer A, et al. Human interleukin interleukin-1 receptor complex. J Biol Chem 1995; 270: 13757.
7: molecular cloning and growth factor activity on human and 664. Greil J, Gramatzki M, Burger R, et al. The acute lymphoblastic
murine B-lineage cells. Proc Nat Acad Sci USA 1989; 86: 302. leukemia cell line SEM with t(4;11) chromosomal rearrangement
642. Gordon C, Ranges GE, Greenspan JS, Wolfsy D. Chronic therapy is biphenotypic and responsive to interleukin-7. British J
with tumor necrosis factor-α in autoimmune NZB/NZW F1 mice. Haematology 1994; 86: 275–283.
Clin Immunol Immunopathol 1989; 52: 421. 665. Grell M, Douni E, Wajant H, Lohden M, et al. The transmem-
643. Gordon J, MacLean LD. A lymphocyte-stimulating factor pro- brane form of tumor necrosis factor is the prime activating ligand
duced in vitro. Nature 1965; 208: 795–796. of the 80 kDa tumor necrosis factor receptor. Cell 1995; 83: 793.
Walter M. Lewko and Robert K. Oldham 233

666. Grenier A, Dehoux M, Boutten A, et al. Oncostatin M production 689. Gu ZJ, Costes V, Lu ZY, Zhang XG, et al. Interleukin-10 is a
and regulation by human polymorphonuclear neutrophils. Blood growth factor for human myeloma cells by induction of an
1999; 93: 1413. oncostatin autocrine loop. Blood 1996; 88: 3972–3986.
667. Grewal IS, Foellmer HG, Grewell KD, et al. Requirement for CD40 690. Gu ZJ, Zhang XG, Hallet MM, et al. A ciliary neurotrophic fac-
ligation in costimulation induction, T cell activation and experimen- tor-sensitive human myeloma cell line. Exp Hematol 1996; 24:
tal allergic encephalomyelitis. Science 1996; 273: 1864. 1195–1200.
668. Grewal IS, Grewell KD, Wong FS, et al. Local expression of trans- 691. Guesdon F, Freshney N, Waller RJ, Rawlinson L, Saklatvala J.
gene encoded TNF α in islets prevents autoimmune diabetes in Interleukin-1 and tumor necrosis factor stimulate two novel pro-
nonobese diabetic (NOD) mice by preventing the development of tein kinases that phosphorylate hsp27 and β-casein. J Biol Chem
auto reactive islet-specific T cells. J Exp Med 1996; 184: 1963. 1993; 268: 4236.
669. Gri G, Gallo E, Di Carlo E, et al. OX40-transduced tumor cell 692. Guimbaud R, Abitbol V, Bertrand V, et al. Leukemia inhibitory
vaccine synergizes with GM-CSF and requires CD40-APC sig- factor involvement in human ulcerative colitis and its potential
naling to boost the host T cell antitumor response. J Immunol role in malignant course. Eur Cytokine Netw 1998; 9: 607–612.
2003; 170: 99–106. 693. Guinn BA, Bertram EM, DeBenedette MA, et al. 4–1 BBL
670. Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA. enhances anti-tumor responses in the presence or absence of
Fas ligand-induced apoptosis as a mechanism of immune CD28 but CD28 is required for protective immunity against
privilege. Science 1995; 270: 1189. parental tumors. Cell Immunol 2001; 210: 56–65.
671. Griffith TS, Chin WA, Jackson GC, et al. Intracellular regulation 694. Guo H, Cai CQ, Schroeder RA, Kuo PC. Osteopontin is a nega-
of TRAIL-induced apoptosis in human melanoma cells. J tive feedback inhibitor of nitrogen oxide synthetase in murine
Immunol 1998; 161: 2833. macrophages. J Immunol 2001; 166: 1079–1086.
672. Griffith TS, Wiley SR, Kubin MZ, et al. Monocyte-mediated 695. Gurunathan S, Irvine KR, Wu C-Y, et al. CD40 ligand/trimer
tumoricidal activity via the tumor necrosis factor-related cytokine, DNA enhances both humoral and cellular immune responses and
TRAIL. J Exp Med 1999; 189: 1343. induces protective immunity to infectious and immune challenge.
673. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine J Immunol 1998; 161: 4563.
activated killer cell phenomenon. J Exp Med 1982; 155: 1823. 696. Gustavson LE, Nadeau RW, Oldfield NF. Pharmacokinetics of
674. Grimm EA, Smid CM, Lee JJ, et al. Unexpected cytokines in Teceleukin (recombinant human interleukin-2) after intravenous
serum of malignant melanoma patients during sequential or subcutaneous administration to patients with cancer. J Biol
biochemotherapy. Clin Cancer Res 2000; 6: 3895–3903. Response Modif 1989; 8: 440.
675. Grohmann U, Bianchi R, Belladonna ML, et al. IL-12 acts selec- 697. Guthridge MA, Stomski FC, Thomas D, et al. Mechanism of acti-
tively on CD8α-dendritic cells to enhance presentation of a tumor vation of GM-CSF, IL-3, IL-5 family of receptors. Stem Cells
peptide. J Immunol 1999; 163: 3100. 1998; 16: 301–313.
676. Gross V, Andrus T, Caesar I, Roth M, Scholmerich J. Evidence 698. Guy GR, Chua SP, Wong NS, et al. Interleukin 1 and tumor
for continuous stimulation of interleukin-6 production in Crohn’s necrosis factor activate common multiple protein kinases in
disease. Gastroenterology 1992; 102: 514. human fibroblasts. J Biol Chem 1991; 266: 14343–14352.
677. Groux H, Bigler M, de Vries JE, Roncarolo M-G. Inhibitory and 699. Habib T, Senadheera S, Weinberg K, et al. The common γ chain
stimulatory effects of IL-10 on human CD8+ T cells. J Immunol (γC) is a required signaling component of the IL-21 receptor and
1998; 160: 3188. supports IL-21-induced cell proliferation via JAK3. Biochemistry
678. Groux H, Cottrez F, Rouleau M, et al. A transgenic model to ana- 2002; 41: 8725–8731.
lyze the immunoregulatory role of IL-10 secreted by antigen- 700. Hahne M, Rimoldi D, Schroter M, et al. Melanoma cell expres-
presenting cells. J Immunol 1999; 162: 1723–1729. sion of FAS (Apo-1/CD95) ligand: implications for tumor
679. Grove RI, Eberhardy C, Abid S, et al. Oncostatin M is a mitogen immune escape. Science 1996; 274: 1363–1366.
for vascular smooth muscle cells. Proc Natl Acad Sci USA 1993; 701. Halfter H, Kremerskothen, Weber J, et al. Growth inhibition of
90: 823–827. newly established human glioma cell lines by leukemia inhibi-
680. Grubler U, Chua AO, Schoenhaut DS, et al. Coexpression of two tory factor. J Neurooncol 1998; 39: 1–18.
distinct genes required to generate secreted bioactive cytotoxic 702. Halfter H, Stogbauer F, Friedrich M, et al. Oncostatin M-mediated
lymphocyte maturation factor. Proc Nat Acad Sci USA 1991; 88: growth inhibition of human glioblastoma cells does not depend
4143–4147. on stat 3 or on mitogen-activated protein kinase activation. J
681. Grunig G, Warnock M, Wakil AE, et al. Requirement for IL-13 Neurochem 2000; 75: 973–981.
independently of IL-4 in experimental asthma. Science 1998; 703. Halloran MM, Haskel CJ, Woods JM, et al. Interleukin-13 is an
282: 2261–2263. endothelial chemotactin. Pathobiology 1997; 65: 287.
682. Gruol DL, Nelson TE. Physiological and pathological roles of 704. Hamann D, Hilkens CMU, Grogan JL, et al. CD30 expression
interleukin-6 in the central nervous system. Mol Neurobiol does not discriminate between human Th1- and Th2-type T cells.
1997; 15: 307. J Immunol 1996; 156: 1387–1391.
683. Gruss H-J, Boiani N, Williams DE, et al. Pleiotrophic effects of 705. Hamilton JA, Leizer T, Piccoli DS, et al. Oncostatin M stimulates
the CD30 ligand on CD30-expressing cells and lymphoma cell urokinase-type plasminogen activator activity in human synovial
lines. Blood 1994; 83: 2045. fibroblasts. Biochem Biophys Res Commun 1991; 180: 652.
684. Gruss HJ, Brach MA, Drexler HG, Bross KJ, Herrmann F. 706. Hammonds P, Beggs M, Beresford G, et al. Insulin secreting β
Interleukin 9 is expressed by primary and cultured Hodgkin and cells possess specific receptors for interleukin-1β. FEBS Lett
Reed-Sternberg cells. Cancer Res 1992; 52: 1026. 1990; 261: 97. Biochem Biophys Res Commun 1991; 180: 652.
685. Gruss HJ, Dower SK. Tumor necrosis factor ligand superfamily. 707. Han X, Itoh K, Balch CM, Pellis JH. Recombinant interluekin-4
Blood 1995; 85: 3378–3404. (RIL-4) inhibits interleukin 2 induced activation of peripheral
686. Grzegorzewski K, Komschlies KL, et al. Administration of blood lymphocytes. Lymphokine Cytokine Res 1988; 7:
recombinant human interleukin-7 to mice induces the exportation 227–234.
of myeloid progenitor cells from the bone marrow to peripheral 708. Hanavadi S, Martin TA, Watkins G, et al. Expression of interleu-
sites. Blood 1994; 83: 377. kin 11 and its receptor and their prognostic value in breast cancer.
687. Gu L, Tseng S, Horner RM, Tam C, Loda M, Rollins BJ. Control Ann Surg Oncol 2006; 13: 802–808.
of TH2 by the chemokine monocyte chemoattractant protein-1. 709. Hanna N, Hanna I, Hleb M, et al. Gestational age dependent
Nature 2000; 404: 407. expression of IL-10 and its receptor in human placental tissues
688. Gu Y, Kuida K, Tsutui H, et al. Activation of interferon γ inducing and isolated cytotrophoblasts. J Immunol 2000; 164: 5721–5728.
factor mediated by interleukin-1β converting enzyme. Science 710. Hannum C, Culpepper J, Campbell D, et al. Ligand for FLT3/
1997; 275: 206. FLT2 receptor tyrosine kinase regulates growth of hematopoietic
234 Cytokines

stem cells and is coded by variant RNAs. Nature 1994; 368: 730. Hedrick SM, Cohen D, Nieson E, et al. Isolation of cDNA clones
643–648. encoding T-cell specific membrane association proteins. Nature
711. Hansen HP, Diedrich Sebastian, et al. CD30 shedding from 1984; 308: 149.
Karpas 299 lymphoma cells is mediated by TNF-α-converting 731. Heinzel FP, Hujer AM, Ahmed FN, Rerko RM. In vivo produc-
enzyme. J Immunol 2000; 165: 6703–6709. tion and function of IL-12 p40 homodimers. J Immunol 1997;
712. Happel KI, Zheng M, Young E. Roles of Toll-like receptor 4 and 158: 4381.
IL-23 in IL-17 expression in response to Klebsiella pneumonia 732. Held-Feindt J, Mentlein R. CD70/CD27 Ligand, a member of the
infection. J Immunol 2003; 170: 4432–4436. TNF family, is expressed in brain tumors. Int J Cancer 2002; 98:
713. Harada N, Nakayama N, Nakano H, et al. Pro-inflammatory 352–356.
effect of TWEAK/Fn14 interaction on human umbilical vein 733. Hellerstein MK, Meydani SN, Meydani M, Wu K, Dinarello CA.
endothelial cells. Biochem Biophys Res Commun 2002; 299: Interleukin-1-induced anorexia in rat. Influence of prostaglan-
488–493. dins. J Clin Invest 1989; 84: 228.
714. Harashima S, Horiuchi T, Hatta N, et al. Outside to inside signal 734. Henney CS, Kuribayashi K, Kern DE, Gillis S. Interleukin-2 aug-
through the membrane TNF-α induces E selectin (CD62E) ments natural killer cell activity. Nature 1981; 291: 335.
expression on activated human CD4+ T cells. J Immunol 2001; 735. Herbelin A, Machzvoine F, Schneider E, et al. IL-7 is requisite
166: 130–136. for IL-1 induced thymocyte proliferation. Involvement of IL-7 in
715. Harding FA, McArthur JG, Gross JA, Gaulet DH, Allison JP. the synergistic effects of granulocyte macrophage colony stimu-
CD-28-mediated signaling costimulates murine T cells and lating factor or tumor necrosis factor with IL-1. J Immunol 1992;
prevents induction of anergy in T cell clones. Nature 1992; 148: 99–105.
356: 607. 736. Herbert JM, Savi P, Laplace M-C, et al. IL-4 and IL-13 exhibit
716. Hargreaves PG, Wang F, Antcliff J, et al. Human myeloma cells comparable abilities to produce pyrogen induced expression of
shed the interleukin-6 receptor: inhibition of tissue inhibitor of procoagulant activity in endothelial cells and monocytes. FEBS
metalloproteinase-3 and a hydroxamate-bases metalloproteinase Lett 1993; 328: 268.
inhibitor. Br J Haematol 1998; 101: 694. 737. Hercend T, Meuer SC, Reinherz EL, et al. Generation of a cloned
717. Harlan JM, Killen PD, Harker LA, Striker GE, Wright DG. NK cell line from the null cell fraction of human peripheral
Neutrophil-mediated endothelial injury in vitro mechanisms of blood. J Immunol 1982; 129: 1299.
cellular detachment. J Clin Invest 1981; 68: 1394. 738. Herman C, Chernajovsky Y. Mutation of proline 211 reduces
718. Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-pro- shedding of the human 75 TNF receptor. J Immunol 1998; 160:
ducing CD4+ effector T cells develop via a lineage distinct from 2478–2487.
the T helper type 1 and 2 lineages. Nat Immunol 2005; 6: 739. Hermann E, Darcissac E, Idziorek T, Capron A, Bahr GM.
1123–1132. Recombinant interleukin-16 selectively modulates surface recep-
719. Harrop JA, McDonnell PC, Brigham-Burke M, et al. Herpes tor expression and cytokine release in macrophages and dendritic
virus entry mediator ligand (HVEM-L), a novel ligand for cells. Immunology 1999; 97: 241–248.
HVEM/TR2, stimulates proliferation of T cells and inhibits HT29 740. Hermans IF, Ritchie DS, Daisch A, et al. Impaired ability of
cell growth. J Biol Chem 1998; 273: 27548. MHC class II−/− dendritic cells to provide protection is rescued
720. Harrop JA, Reddy M, Dede K, et al. Antibodies to TR2 (herpes- by CD40 ligation. J Immunol 999; 163: 77.
virus entry mediator), a new member of the TNF receptor super- 741. Hershey GK. IL-13 receptors and signaling pathways: an evolv-
family, block T cell proliferation, expression of activation ing web. J Allergy Clin Immunol 2003; 111: 677–691.
markers, and the production of cytokines. J Immunol 1998; 161: 742. Herx L, Rivest S, Yong VW. Central nervous system-initiated
1786–1794. neurotrophism in trauma: IL-1β is required for production of cili-
721. Hartmann F, Renner C, Jung W, Pfreundschuh M. Anti-CD16/ ary neurotrophic factor. J Immunol 2000; 165: 2232–2239.
CD30 bispecific antibodies as possible treatment for refactory 743. Hess SA, Rensing-Ehl A, Schwabe R, Bufler P, Engelman H.
Hodgkin’s disease. Leuk Lymphoma 1998; 31: 385–392. CD40 functions in non-hematopoietic cells: mobilization of
722. Haselmayer P, Tenzer S, Kwon BS, et al. Herpes virus entry NF-κB and induction of IL-6 production. J Immunol 1995;
mediator synergizes with toll-like receptor mediated neutrophil 155: 4588.
inflammatory responses. Immunology 2006; 119: 404–411. 744. Heufler C, Koch F, Schuler G. Granulocyte macrophage colony
723. Haskill S, Martin M, Van Le L, et al. cDNA cloning of an intrac- stimulating factor and interleukin 1 mediate the maturation of
ellular form of the interleukin-1 receptor antagonist associated murine epidermal Langerhans cells into potent immunostimula-
with epithelium. Proc Natl Acad Sci USA 1991; 88: 3681. tory dendritic cells. J Exp Med 1988; 167: 700.
724. Hawley RG, Berger LC. Growth control mechanisms in multiple 745. Heufler C, Topar G, Grasseger A, et al. Interleukin 7 is produced
myeloma. Leuk Lymphoma 1998; 29: 465–475. by murine and human keratinocytes. J Exp Med 1993; 178:
725. Hayakawa F, Towatari M, Kiyoi H, et al. Tandem-duplicated Flt3 1109–1114.
constitutively activates STAT5 and MAP kinase and introduces 746. Heuze-Vourc’h N, Liu M, Dalwadi H, et al. IL-20, an anti-angio-
autonomous cell growth in IL-3-dependent cell lines. Oncogene genic cytokine that inhibits COX-2 expression. Biochem Biophys
2000; 19: 624–631. Res Commun 2005; 333: 470–475.
726. Hayashida K, Kitamura T, Gorman DM, et al. Molecular cloning 747. Hibbert L, Pflanz S, De Waal Malefyt R, et al. IL-27 and INF-α
of a second subunit for the receptor for human granulocyte-mac- signal via Stat 1 and Stat 3 and induce T-bet and IL-12Rβ2 in
rophage colony stimulating factor GM-CSF: reconstitution of a naive T cells. J Interferon Cytokine Res 2003; 23: 513–522.
high affinity GM-CSF receptor. Proc Natl Acad Sci USA 1990; 748. Hibi K, Takahashe T, Sekido Y, et al. Coexpression of the stem
87: 9655–9659. cell factor and the c-kit genes in small cell lung cancer. Oncogene
727. Hayward AR, Levy J, Facchetti F, et al. Cholangiopathy and 1991; 6: 2291–2296.
tumors of the pancreas liver, and biliary tree in boys with X-linked 749. Hickman CJ, Crim JA, Mostowski HS, Siegel JP. Regulation of
immunodeficiency with hyper-IgM. J Immunol 1997; 158: 977. human cytotoxic T lymphocyte development by interleukin-7. J
728. He Y-W, Malek TR. Interleukin-7 receptor α is essential for the Immunol 1990; 145: 2415–2420.
development of γδ+ T cells, but not natural killer cells. J Exp 750. Hidi R, Riches V, Al-Ali M, et al. Role of B7-CD28/CTLA-4
Med 1996; 184: 289. costimulation and KF-kappa B in allergen-induced T cell chemot-
729. Heath WR, Kurts C, Caminschi I, Carbone FR, Miller JF. CD30 axis by IL-16 and RANTES. J Immunol 2000; 164: 266–271.
prevents T cell responses to non-lymphoid tissues. Immunol Rev 751. Higgins LM, McDonald SA, et al. Regulation of T cell activation
1999; 169: 23. in vitro and in vivo by targeting the OX40–OX40ligand interaction:
Walter M. Lewko and Robert K. Oldham 235

amelioration of ongoing inflammatory bowel disease with an 773. Ho AS, Liu Y, Khan TA, et al. A receptor for interleukin-10 is
OX40-IgG fusion protein. J Immunol 1999; 162: 486. structurally related to interferon receptors. Proc Natl Acad Sci
752. Higuchi CM, Thompson JA, Lindgren CG, et al. Induction of USA 1993; 90: 11267–11271.
lymphokine-activated killer activity by interleukin 4 in human 774. Ho DH, Vu H, Brown SAN, et al. Soluble tumor necrosis factor-
lymphocytes preactivated by interleukin 2 in vitro or in vivo. like weak inducer of apoptosis overexpression in HEK293 cells
Cancer Res 1989; 49: 6487–6492. promotes tumor growth and angiogenesis in athymic nude mice.
753. Higuchi M, Nagasawa K, Horiuchi T, et al. Membrane tumor Cancer Res 2004; 64: 8968–8972.
necrosis factor-α (TNF-α) expressed on HTLV-I infected T cells 775. Hoch H, Dorsch M, Diamantstein T, Blankenstien T. Interleukin
mediates a costimulatory signal for B cell activation: character- 7 induces CD4+ T cell dependent tumor rejection. J Exp Med
ization of membrane TNF-α. Clin Immunol Immunopathol 1997, 1991; 174: 1291–1298.
82: 133. 776. Hoch H, Dorsch M, Kunzendorf U, et al. Mechanism of the rejec-
754. Hilbert DM, Kopf M, Mock BA, Kohler G, Rudikoff S. tion induced by tumor cell targeted gene transfer of interleukin 2,
Interleukin-6 is essential for in vivo development of B lineage interleukin 4, interleukin 7 tumor necrosis factor or interferon γ.
neoplasms. J Exp Med 1995; 182: 243–248. Proc Nat Acad Sci USA 1993; 90: 2774–2778.
755. Hill RJ, Warren MK, Levin J. Stimulation of thrombopoiesis in 777. Hochman PS, Majeau GR, Mackey F, Browning JL. Proinflammatory
mice by human recombinant interleukin 6. J Clin Invest 1990; responses are efficiently induced by homotrimeric but not heterotri-
85: 1242. meric lymphotoxin. J Inflamm 1996; 46: 220.
756. Hill S, Youde SJ, Man S, et al. Activation of CD40 in cervical 778. Hofbauer LC, Khosla S, Dunstan CR, et al. Estrogen stimulates
carcinoma cells facilitates CTL responses and augments chemo- gene expression and protein production of osteoprotegerin in
therapy-induced apoptosis. J Immunol 2005; 174: 41–50. human osteoblastic cells. Endocrinology 1999; 140: 4367.
757. Hilliard B, Wilmen A, Seidel C, et al. Roles of TNF-related apop- 779. Hoffman TK, Meidenbauer N, Dworacki G, Kanaya H, Whiteside
tosis-inducing ligand in experimental autoimmune encephalitis. TL. Generation of tumor specific T lymphocytes by cross prim-
J Immunol 2001; 166: 1314–1319. ing with human dendritic cells ingesting apoptotic tumor cells.
758. Hilton DJ, Zhang J-G, Metcalf D, et al. Cloning and characteriza- Cancer Res 2000; 60: 3542.
tion of a binding subunit of the interleukin-13 receptor that is also 780. Hogaboam C, Kunkel SL, Strieter RM, et al. Novel role of trans-
a component of the interleukin-4 receptor. Proc Natl Acad Sci membrane stem cell factor for mast cell activation and eotaxin
USA 1996; 93: 497. production in mast cell–fibroblast interactions. J Immunol 1998;
759. Hilton DJ. LIF: lots of interesting functions. Trends Biochem Sci 160: 6166.
1991: 17: 72. 781. Hogge DE, Yalcintepe L, Wong SH, et al. Variant diphtheria
760. Hinrichs CS, Spolski R, Paulos CM, et al. IL-2 and IL-21 confer toxin-interleukin-3 fusion proteins with increased receptor affin-
opposing differentiation programs too CD8+ T cells for adoptive ity have enhanced cytotoxicity against acute myeloid leukemia
immunotherapy. Blood; 00: 000–000. progenitors. Clin Cancer Res 2006; 12 1284–1291.
761. Hintzen RQ, de Jong R, Hack CE, et al. A soluble from of the 782. Hogquist KA, Unanue ER, Chaplin DD. Release of IL-1 from
human T cell differentiation antigen CD27 is released after trig- mononuclear phagocytes. J Immunol 1991; 147: 2181.
gering of the TCR/CD3 complex. J Immunol 1991; 147: 29–35. 783. Hoidal JR. The eosinophils and acute lung injury. Am Rev Respir
762. Hintzen RQ, Lens SM, Lammers K, et al. Engagement of CD27 Dis 1990; 142: 1245.
with its ligand CD70 provides a second signal for T cell activa- 784. Holbrook ST, Ohls RK, Schibler KR, et al. Effect of interleukin 9
tion. J Immunol 1995; 154: 2612–2623. on clonogenic maturation and cell cycle status of fetal and adult
763. Hintzen RQ, Lens SMA, Koopman G, Paris ST, Spitz H, van Lier hematopoietic progenitors. Blood 1991; 77: 2129–2134.
RAW. CD70 represents the human ligand for CD27. Int Immunol 785. Holen I, Croucher PI, Hamdy FC, et al. Osteoprotegerin (OPG) is
1994; 6: 477. a survival factor for human prostate cancer cells. Cancer Res
764. Hirahara N, Nio Y, Sasaki S, Takamura M, et al. Reduced inva- 2002; 62: 1619–1623.
siveness and metastasis of Chinese hamster ovary cells trans- 786. Hollenbaugh D, Mischel-Petty N, Edwards CP, et al. Expression
fected with human interleukin-17 gene. Anticancer Res 2000; 20: of functional CD40 by vascular endothelial cells. J Exp Med
3137. 1995; 182: 33.
765. Hirano A, Longo DL, Taub DD, et al. Inhibition of human breast 787. Holmes WE, Lee J, Kuang WJ, et al. Structure and functional
carcinoma growth by a soluble recombinant human CD40 ligand. expression of a human interleukin-8 receptor. Science 1991;
Blood 1999; 93: 2999. 253: 1278.
766. Hirano T, Yasukawa K, Harada H, et al. Complementary DNA for 788. Holtmann H, Wallach D. Downregulation of the receptors for
a novel interleukin (BSF-2) that induces B lymphocytes to pro- tumor necrosis factor by interleukin 1 and 4 β-phorbol-12
duce immunoglobulin. Nature 1986; 324: 73–76. myristate-13-acetate. J Immunol 1987; 139: 1161.
767. Hirayama F, Clark SC, Ogawa M. Negative regulation of early B 789. Hong HJ, Lee JW, Park SS, et al. Humanized anti-4-1BB monoclo-
lymphopoiesis by interleukin 3 and interleukin 1a. Proc Natl nal antibody suppresses antigen-induced humoral immune reaponse
Acad Sci 1994; 91: 469. in nonhuman primates. J Immunother 2000; 23: 613–621.
768. Hirayama F, Lyman SD, Clark SC, Ogawa M. The flt3 ligand 790. Hoon DSB, Banex M, Okun E, et al. Modulation of human mela-
supports proliferation of lymphohematopoietic progenitors and noma cells by interleukin-4 and in combination with γ-interferon
early B-lymphoid progenitors. Blood 1995; 85: 1762. or α-tumor necrosis factor. Cancer Res 1991; 51: 2002–2008.
769. Hirota H, Kiyama H, Kishimoto T, Taga T. Accelerated nerve 791. Hoon DSB, Edward O, Banez M, et al. Interleukin 4 alone and
regeneration in mice by upregulated expression of interleukin (IL) with γ-interferon or tumor necrosis factor inhibits the growth and
6 and IL-6 receptor after trauma. J Exp Med 1996; 183: 2627. modulates cell surface antigens on human renal cell carcinoma.
770. Hirota S, Ito A, Nagoshi J, et al. Expression of bone protein mes- Cancer Res 1991; 51: 5687–5693.
senger ribonucleic acids in human breast cancers. Possible 792. Hor S, Pirzer H, Dumoutier L, et al. The T cell lymphokine inter-
involvement of osteopontin in development of calcifying foci. leukin-26 targets epithelial cells through the interleukin-20
Lab Invest 1995; 72: 64–69. receptor1 and interleukin-10 receptor 2 chains. J Biol Chem
771. Hirte HW, Miller D, Tonkin K, et al. A randomized trial of para- 2004; 279: 33343–33351.
centesis plus intraperitoneal tumor necrosis factor-α versus para- 793. Horn D, Fitzpatrick WC, Gompper PT, et al. Regulation of cell
centesis alone in patients with symptomatic ascites from recurrent growth by recombinant oncostatin M. Growth Factors 1990; 2: 157.
ovarian carcinoma. Gynecol Oncol 1997; 64: 80–87. 794. Horton SA, Oldham RK, Yannelli JR. Generation of human lym-
772. Hisada M, Kamiya S, Fujita K, et al. Potent antitumor activity of phokine-activated killer cells following brief exposure to high
interleukin-27. Cancer Res 2004; 64: 1152–1156. dose interleukin-2. Cancer Res 1990; 50: 1686–1692.
236 Cytokines

795. Hoshino K, Tsutsui H, Kawai T, et al. Generation of IL-18 defi- 816. Hudkins KL, Giacelli CM, Eitner F, Couser WG, Johnson RJ,
cient mice: evidence for IL-1 receptor-related protein as an Alpers CE. Osteopontin expression in human cresentic glomeru-
essential IL-18 binding receptor. J Immunol 1999; 162: 5041. lonephritis. Kid Int 2000; 57: 105–116.
796. Hoshino T, Kawase Y, Okamoto M, et al. IL-18 transgenic mice: 817. Hui W, Bell M, Carrol G. Detection of oncostatin M in synovial
In vivo evidence of a broad role for IL-18 in modulating immune fluid from patients with rheumatoid arthritis. Ann Rheu Dis 1997,
function. J Immunol 2001; 166: 7014–7018. 56: 184.
797. Hoshino T, Wiltrout RH, Young HA. IL-18 is a potent coinducer 818. Huland E, Huland H, Heinzer H. Interleukin-2 by inhalation:
of IL-13 in NK and T cells: a new potential role for IL-18 in mod- Local therapy for metastatic renal cell carcinoma. J Urology
ulating the immune response. J Immunol 1999; 162: 5070–5077. 1992; 147: 344–348.
798. Hosken NA, Shibuya K, Heath AW, Murphy KM, O’Garra A. 819. Hultner L, Druez C, Moeller J, et al. Mast cell growth enhancing
The effect of antigen dose on CD4+ T helper cell phenotype activity (MEA) is structurally related and functionally identical
development in a T cell receptor-transgenic model. J Exp Med to the novel mouse T cell growth factor P40/TCGF1II. Eur J
1995; 182: 1579. Immunol 1990; 20: 1413–1416.
799. Hosoi T, Wada S, Suzuki S, et al. Bacterial endotoxin induces 820. Hultner L, Kolsch S, Stassen M, et al. In activated mast cells,
IL-20 expression in the glial cells. Brain Res Mol Brain Res IL-1 upregulates the production of several Th2 related cytokines
2004; 130: 23–29. including IL-9.J Immunol 2000; 164: 5556–5563.
800. Hossiau FA, Renauld JC, Stevens M, et al. Human T cell lines 821. Humphreys NE, Xu D, Hepworth MR, et al. IL-33, a potent
and clones respond to IL-9. J Immunol 1993; 150: 2634–2640. inducer of adaptive immunity to intestinal nematodes. I Immunol
801. Howard M, Hilfiker M, Johnson B, et al. Identification of a T cell 2008; 180: 2443–2449.
derived B cell growth factor distinct from Interleukin-2. J Exp 822. Hunninghake GW, Glazier MM, Monick MM, Dinarello CR.
Med 1982; 155: 914–923. Interleukin-1 is a chemotactic factor for T lymphocytes. Am Rev
802. Howard M, Muchamual S, Andrade S, Menon S. Interleukin Respir Dis 1987; 135: 66.
10 protects mice from lethal endotoxemia. J Exp Med 1993; 823. Hunte BE, Hudak D, Campbell D, Xu Y, Rennick D. flk2/flt3
177: 1205. ligand is a potent cofactor for the growth of primitive B cell
803. Howells G, Pham P, Taylor D, Foxwell B, Feldman M. Interleukin progenitors. J Immunol 1995; 156: 486.
4 induces interleukin 6 production by endothelial cells: synergy 824. Hunter CA, Chizzonite R, Remmington JS. IL-1β is required for
with interferon-γ. Eur J Immunol 1991; 21: 97. IL-12 to induce production of IFNγ by NK cells. J Immunol
804. Hsieh CS, Macatonia SE, Tripp CS, et al. Development of TH1 1995; 155: 4347–4354.
CD4+ T cells through IL-12 produced by Listerial-induced mac- 825. Hurst SD, Muchamuel T, Gorman DM, et al. New IL-17 family
rophages. Science 1993; 260: 547–549. members promote Th1 and Th2 responses in the lung: in vivo
805. Hsieh MY, Chen WY, Jiang MJ, et al. Interleukin-20 promotes function of the novel cytokine IL-25. J Immunol 2002; 169:
angiogenesis in a direct and indirect manner. Genes Immun 2006; 443–453.
7: 234–242. 826. Hurst, SM, McLoughlin RM, Monslow J, et al. Secretion of
806. Hsu D-H, de Waal Malefyt R, Fiorentino DF, et al. Expression of oncostatin M by infiltrating neutrophils: regulation of IL-6 and
interleukin 10 activity by Epstein Barr virus protein BCRFI. chemokine expression in mesothelial cells. J Immunol 2002; 169:
Science 1990; 250: 830–832. 5244–5251.
807. Hsu H, Lacey DL, Dunstan C R, et al. Tumor necrosis factor 827. Hurtado JC, Kim SH, Pollok KE, Kwon BS. Potential role of
receptor family member RANK mediates osteoclast differentia- 4-1BB in T cell activation: comparison with costimulatory mol-
tion and activation induced by osteoprotegerin ligand. Proc Natl ecule CD28. J Immunol 1995; 155: 3360.
Acad Sci USA 1999; 96: 3540. 828. Hurtado JC, Kim Y-J, Kwon BS. Signals through 4-1BB are
808. Hsu H, Solovyev I, Colombero A, Elliott G, Kelley JM, Boyle costimulatory to previously activated splenic T cells and
WJ. ATAR, a novel TNF receptor family member, signals through inhibit activation-induced cell death. J Immunol 1997; 158:
TRAF2 and TRAF5. J Biol Chem 1997; 272: 13471–13474. 2600–2609.
809. Hsu YH, Li HH, Hsieh MY, et al. Function of interleukin-20 as a 829. Hurteau JA, Blessing JA, DeCesare SL, Creasman WT.
proinflammatory molecule in rheumatoid and experimental Evaluation of recombinant human interleukin-12 in patients with
arthritis. Arthritis Rheum 2006; 54: 2722–2733. recurrent or refactory ovarian cancer: a gynecologic cncology
810. Huang EY, Madireddi MT, Gopalkrishnan RV, et al. Genomic group study. Gynecol Oncol 2001; 82: 7–10.
structure, chromosomal localization and expression profile of a 830. Husain SR, Puri RK. Interleukin-13 receptor-directed cytotoxin
novel melanoma differentiation associated (mda-7) gene with for malignant glioma therapy: from bench to bedside. J
cancer specific growth suppressing and apoptosis inducing prop- Neurooncol 2330; 65: 37–48.
erties. Oncogene 2001; 20: 7051–7063. 831. Hutchins D, Steel CM. Regulation of ICAM-1 (CD54) expres-
811. Huang J, Kerstan KW, Ahmadzadeh M, et al. Modulation by IL-2 sion in human breast cancer cell lines by interleukin 6 and fibro-
of CD70 and CD27 expression on CD8+ T cells: Importance for blast derived factors. Int J Cancer 1994; 58: 80–84.
the therapeutic effectiveness of cell transfer immunotherapy. 832. Hutson TE, Mekhail T, Molto L, et al. Phase I trial of subcutane-
J Immunol 2006; 176: 7726–7735. ously administered rhuIL-12 and rhuIFN-α2b in patients with
812. Huang S, Mills L, Mian B. Fully humanized neutralizing anti- metastatic renal cell carcinoma or malignant melanoma. Proc
bodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, tumor ASCO 2001; 20: 1030.
growth, and metastasis of human melanoma. Am J Pathol 2002; 833. Hymowitz SG, Filvaroff EH, Yin JP, et al. IL-17s adopt a cystine
161: 125–134. knot fold: structure and activity of a novel cytokine, IL-17F, and
813. Huang SK, Xiao H-Q, Kleine-Tebbe J, et al. IL-13 expression at implications for receptor binding. The EMBO Journal 2001; 20:
the sites of allergic challenge in patients with asthma. J Immunol 5332–5341.
1995; 155: 2688–2694. 834. Iemura A, Tsai M, Ando A, Wershil BK, Galli SJ. The c-kit
814. Huang SM, Lopez CA, Heck DE, et al. Osteopontin inhibits ligand, stem cell factor, promotes mast cell survival by suppress-
induction of nitric oxide synthase gene expression by inflamma- ing apoptosis. Am J Pathol 1994, 144: 321.
tion mediators in mouse kidney epithelial cells. J Biol Chem 835. Ihle JN, Keller J, Oroszlan S, et al. Biological properties of inter-
1994; 2712: 5414. leukin-3. I. Demonstration of WEHI-3 growth factor activity,
815. Huber AR, Kunkel SL, Todd RF, Weiss SJ. Regulation of transen- mast cell growth factor activity, P cell-stimulating activity, colony-
dothelial neutrophil migration by endogenous interleukin-8. stimulating activity, and histamine producing cell-stimulating
Science 1991; 254: 99. factor activity. J Immunol 1983; 131: 282.
Walter M. Lewko and Robert K. Oldham 237

836. Ihn H, LeRoy EC, Trojanowska M. Oncostatin M stimulates tran- 856. Irmler M, Thome M, Hahne M, et al. Inhibition of death receptor
scription of the human α2(I) collagen gene via the Sp1/Sp3- signals by cellular FLIP. Nature 1997; 388: 190.
binding site. J Biol Chem 1997; 272: 24666. 857. Iscove NN. Erythropoietin independent stimulation of early
837. Ihn H, Tamaki K. Oncostatin M stimulates the growth of dermal erythropoiesis in adult marrow cultures by conditioned medium
fibroblasts via a mitogen-activated protein kinase-dependent of lectin stimulated mouse spleen cells. In: Golde D, Kline MJ,
pathway. J Immunol 2000; 165: 2149–2155. Metcalf D, Fox CF, eds. Hematopoietic Cell Differentiation.
838. Iho S, Shau HY, Golub SH. Characteristics of Interleukin 6 Academic Press, New York, 1978; 37–52.
enhanced lymphokine activated killer cell function. Cell Immunol 858. Ishida T, Kobayashi N, Tojo T, et al. CD40 signaling-medi-
1991; 135: 66–77. ated induction of Bcl-XL, Cdk4 and Cdk6. J Immunol 1995;
839. Iikura M, Suto H, Kajiwara N, et al. IL-33 can promote survival, 155: 5527.
adhesion, and cytokine production in human mast cells. Lab. 859. Ishimi Y, Miyaura C, Jin CH, et al. IL-6 is produced by osteo-
Invest 2007; 87: 971–978. blasts and induces bone resorption. J Immunol 1990; 145:
840. Iinuma H, Okinaga K, Fukushima R, et al. Superior protection 3297–3303.
and therapeutic effect of IL-12 and IL-18 gene-transduced den- 860. Ishimitsu R, Nishimura H, Yajima T, et al. Overexpression of
dritic neuroblastoma fusion cells on liver metastasis of murine interleukin 15 in vivo enhances Tc1 response, which inhibits
neuroblastoma. J Immunol 2006, 176: 3461–3469. allergic inflammation in a murine model of asthma. J Immunol
841. Iizuka K, Chaplin DD, Wang Y, Wu Q, et al. Requirement for 2001; 166: 1991–2001.
membrane lymphotoxin in natural killer cell development. Proc 861. Ito D, Back TC, Shakov AN, Wiltrout RH, Nedospasov SA. Mice
Natl Acad Sci USA 1999; 96: 6336. with targeted mutation in lymphotoxin-α exhibit enhanced tumor
842. Ikebuchi K, Clark SC, Ihle JN, Souza L, Ogawa M. Granulocyte- growth and metastasis: impaired NK development and recruit-
colony stimulating factor enhances interleukin-3 dependent pro- ment. J Immunol 1999; 163: 2809.
liferation of multipotential hematopoietic progenitors. Proc Natl 862. Ito F, Li O, Shreiner AB, et al. Anti-CD137 monoclonal antibody
Acad Sci USA 1988; 85: 2445. administration augments the antitumor efficacy of dendritic-
843. Ikebuchi K, Wong GG, Clark SC, et al. Interleukin-6 enhance- based vaccines. Cancer Res 2004; 64: 8411–8419.
ment of interleukin-3 dependent proliferation of multipotential 863. Ito H, Kurtz J, Shaffer J, Sykes M. CD4 T cell-mediated allore-
hematopoietic progenitors. Proc Natl Acad Sci USA 1987; 84: sistance to fully MHC-mismatched allogeneic bone marrow
9035. engraftment is dependent on CD40–CD40 ligand interactions,
844. Ikeda K, Nakajima H, Suzuki K, et al. Mast cells produce inter- and lasting T cell tolerance is induced by bone marrow transplan-
leukin-25 upon FcεRI-mediated activation. Blood 2003; 101: tation with initial blockade of this pathway. J Immunol 2001;
3594–3596. 166: 2970–2981.
845. Ikejima T, Okusawa S, Ghezzi P, van der Meer JWM, Dinarello 864. Ito R, Kitadai Y, Kyo E, et al. Interleukin 1 acts as an autocrine
CA. IL-1 induces TNF in human PBMC in vitro and a circulating growth stimulator for human gastric carcinoma cells. Cancer Res
TNF-like activity in rabbits. J Inf Dis 1990; 162: 215. 1994; 54: 4102–41206.
846. Ikejima T, Okusawa S, van der Meer JW, Dinarello CA. Induction 865. Ito T, Wang YH, Duramad O, et al. OX40 ligand shuts down
by toxic shock syndrome toxin-1 of a circulating tumor necrosis IL-10-producing regulatory T cells. Proc Natl Acad Sci USA
factor-like substance in rabbits and of immunoreactive tumor 2006; 103: 13138–13143.
necrosis factor and interleukin-1 from human mononuclear cells. 866. Itoh K, Hirohata S. The role of IL-10 in human B cell activation,
J Infect Dis 1988; 158: 1017. proliferation and differentiation. J Immunol 1995; 154:
847. Ikeuchi H, Kuroiwa T, Hiramatsu N, et al. Expression of interleu- 4341–4350.
kin-22 in rheumatoid arthritis: potential role as a proinflamma- 867. Itoh K, Inoue T, Ito K, Hirohata S. The interplay of IL-10 and
tory cytokine. Arthritis Rheum 2005; 52: 1037–1046. IL-2 in humoral immune responses: IL-10 synergizes with IL-2
848. Im D-H, Barchan D, Maiti PK, Fuchs S, Souroujon MC. Blockade to enhance responses of human B lymphocytes in a mechanism
of CD40 ligand suppresses chronic experimental myasthenia which is different from up-regulation of CD25 expression. Cell
gravis by down-regulation of Th1 differentiation and up-regula- Immunol 1994; 157: 478.
tion of CTLA-4. J Immunol 2001; 166: 6893–6898. 868. Iwagaki H, Hizuta A, Koha H, et al. Circulating levels of soluble
849. Imai T, Hieshima K, Haskell C, et al. Identification and molecular CD30 and other markers in colorectal cancer. J Med 1999; 30:
characterization of fractalkine receptor CX3CR-1, which medi- 111–121.
ates both leukocyte migration and adhesion. Cell 1997; 91: 521. 869. Iwakura Y, Ishigame H. The IL-23/IL-17 axis of inflammation. J
850. Imai Y, Nara N, Tohda S, et al. Antiproliferative and differentia- Clin Invest 2006; 116: 1218–1222.
tive effects of recombinant interleukin-4 on a G-CSF-stimulating- 870. Jacob CO, Aiso S, Michie SA, McDevitt HO, Acha-Orbea H.
dependent myeloblastic leukemic cell line. Blood 1991; 78: Prevention of diabetes in nonobese diabetic mice by tumor necro-
471–478. sis factor (TNF): similarities between TNF-α and interleukin-1.
851. Imura A, Hori T, Imada K, et al. The human OX40/gp34 system Proc Natl Acad Sci USA 1990; 87: 968.
directly mediates adhesion of activated T cells to vascular 871. Jacobsen FW, Veiby OP Jacobsen SE. IL-7 stimulates CSF-
endothelial cells. J Exp Med 1996; 183: 2185–2195. induced proliferation of murine bone marrow macrophages and
852. Infante-Duarte C, Horton HF, Byrne MC, Kamradt T. Microbial Mac-1+ myeloid progenitors in vitro. J Immunol 1994; 153:
lipopeptides induce the production of IL-17 in Th Cells. J 270–276.
Immunol 2000; 165: 6107–6115. 872. Jacobsen SEW, Okkenhaug C, Myklebust J, Veiby OP, Lyman
853. Inoue M, Kyo S, Fujita M, et al. Coexpression of the c-kit recep- SD. The flt3 ligand potently and directly stimulates the growth
tor and the stem cell factor in gynecological tumors. Cancer Res and expansion of primitive murine bone marrow progenitor cells
1994; 54: 3049–3053. in vitro: synergistic interaction with interleukin (IL)-11, IL-12,
854. Inoue S, Shanker M, Miyahara R. MDA-7/IL-24-based cancer and the other hematopoietic growth factors. J Exp Med 1995;
gene therapy: translation from the laboratory to the clinic. Curr 181: 1357–1363.
Gene Ther 2006; 6: 73–91. 873. Jacobson NG, Szabo SJ, Weber-Nordt RM, et al. Interleukin-12
855. Ip WK, Wong CK, Li ML, et al. Interleukin-31 induces cytokine signaling in the T helper Th1 cells involves tyrosine phosphoryla-
and chemokine production from human bronchial epithelial cells tion of signal transducer and activator of transcription STAT3 and
through activation of mitogen-activated protein kinase signaling STAT4. J Exp Med 1995; 181: 1755–1762.
pathways: implications for allergic response. Immunology 2007; 874. Jadus MR, Good RW, Crumpacker DB, Yannelli JR. Effects of
122: 532–541. Interleukin 4 upon human tumoricidal cells obtained from
238 Cytokines

patients bearing solid tumors. J Leukocyte Biol 1991; 49: 895. Jinquan T, Larsen CG, Gesser B, Matsushima K, Thestrup-
139–151. Pedersen K. Human IL-10 is a chemoattractant for CD8+ T lym-
875. Jadus MR, Parkman R. The selective growth of murine newborn- phocytes and an inhibitor of IL-8-induced CD4+ T lymphocyte
derived suppressor cells and their probable mode of action. J migration. J Immunol 1993; 151: 4545–4551.
Immunol 1986; 136: 783–792. 896. Jo M, Kim T-M, Soel D-W, Esplen J, et al. Apoptosis induced in
876. Jadus MR, Schmunk G, Djeu JY, Parkman RJ. Morphology and normal human hepatocytes by tumor necrosis factor-related
lytic mechanism of interleukin-3-dependent natural cytotoxic apoptosis-inducing ligand. Nat Med 2000; 6: 564.
cells: tumor necrosis factor as a possible mediator. Immunol 897. Johnson CS, Chang MJ, Braunschweiger PG, Furmanski P. Acute
1986; 137: 2774–2783. hemorrhagic necrosis of tumors induced by interleukin-1α:
877. Jain S, Chakraborty G, Kundu GC. The crucial role of cyclooxy- effects independent of tumor necrosis factor. J Natl Cancer Inst
genase-2 in osteopontin-induced protein kinase c α/c-src/IκB 1991; 83: 842–848.
kinase α/β-dependent prostate tumor progression and angiogen- 898. Johnson CS, Keckler DJ, Topper MI, Braunschweiger PG,
esis. Cancer Res 2006; 66: 6638–6648. Furmanski P. In vivo hematopoietic effects of recombinant IL-1α
878. Janovic DV, Di Battista JA, Martel-Pellitier J, et al. IL-17 stimu- in mice: stimulation of granulocytic, monocytic, megakaryocytic
lates production and expression of proinflammatory and and early erythroid progenitors, suppression of late stage erythro-
hematopoietic cytokines, IL-1β, and TNF-α, by human mac- poiesis and reversal of erythroid suppression with erythropoietin.
rophages. J Immunol 1998; 160: 3513. Blood 1989; 73: 678.
879. Janovic DV, Martel-Pellitier J, et al. Stimulation of a 92-kd 899. Jones SE, Khandelwal P, McIntyre K, et al. Randomized, double
gelatinase (matrix metalloproteinase 9) production by interleu- blind, placebo controlled trial to evaluate the hematopoietic
kin-17 in human monocyte/macrophages: a possible role in rheu- growth factor PIXY321 after moderate-dose fluorouracil, doxo-
matoid arthritis. Arthritis Rheum 2000; 43: 1134. rubicin, and cyclophosphamide in stage II and III breast cancer.
880. Jansen PM, de Jong IW, Hart M, et al. Release of Leukemia J Clin Oncol 1999; 17: 3025–3032.
inhibitory factor in primate sepsis. Analysis of the role of TNF-α. 900. Joosten LA, Netea MG, Kim SH, et al. IL-32, a proinflammatory
J Immunol 1996; 156: 4401–4407. cytokine in rheumatoid arthritis. Proc Natl Acad Sci USA 2006;
881. Jansson M, Panoutsakopoulou V, Baker J, et al. Attenuated 103: 3298–3303.
experimental autoimmune encephalitis in eta-1/osteopontin-defi- 901. Joosten LAB, Helen MMA, Van de Loo FAJ, Van den Berg WB.
cient mice. J Immunol 2002; 168: 2096–2099. Anti-cytokine treatment of established type II collagen-induced
882. Jarnicki AG, Lysaght J, Todryk S, et al. Suppression of antitumor arthritis in DBA/1 mice: a comparative study using anti-TNFα,
immunity by IL-10 and TGF-β-producing T cells infiltrating the anti-IL-1 α/β, and IL-1Ra. Arthritis Rheum 1996; 39: 797.
growing tumor: influence of tumor environment on the induction 902. Jordan WJ, Eskdale J, Lemmon GP, et al. Human IL-19 regulates
of CD4+ and CD8+ regulatory T cells. J Immunol 2006; 177: immunity through auto-induction of IL-19 and production of
896–904. IL-10. Eur J Immunol 2005, 35: 1576–1582.
883. Jarsdorfer B, Blackwell SE, Wolldridge JE, et al. B-chronic lym- 903. Josien R, Wong BR, Li HL, Steinman RM, Choi Y. TRANCE, a
phocytic leukemia cells and other B cells can produce granzyme TNF family member, is differentially expressed on T cell subsets
B and gain cytotoxic potential after IL-21-based activation. and induced cytokine production in dendritic cells. J Immunol
Blood 2006; 108: 2712–2719. 1999; 162: 2562.
884. Jaye M, Howk R, Burgess W, et al. Human endothelial cell 904. Josimovic-Alasevic O, Durkop H, Schwarting R, et al. Ki-1
growth factor: cloning, nucleotide sequence, and chromosome (CD30) antigen is released by Ki-1 positive tumor cells in vitro
location. Science 1986; 233: 541. and in vivo. I. Partial characterization of soluble Ki-1 antigen and
885. Jelinek DF and Braten JK. Role of IL-12 in human B lymphocyte detection of the antigen in cell culture supernatants and in serum
proliferation and differentiation. J Immunol 1995; 154: 1606. by an enzyme linked immunosorbent assay. Eur J Immunol 1989;
886. Jelinek DF, Splawski JB, Lipsky PE. The roles of IL-2 and IFNγ 19: 157.
in human B cell activation, growth and differentiation. Eur J 905. Joyce DA, Gibbons DP, Green P, Steer JH, Feldman M, Brennan
Immunol 1986; 16: 925. FM. Two inhibitors of cytokine release, interleukin-10 and inter-
887. Jenkins MK, Taylor PS, Norton SD, Urdahl KB. CD28 delivers a leukin-4, have contrasting effects on release of soluble p75 tumor
costimulatory signal involved in antigen specific IL-2 production necrosis factor receptor by cultured monocytes. Eur J Immunol
by human T cells. J Immunol 1991; 147: 2461. 1994; 24: 2699.
888. Jeremias I, Herr I, Boehler T, Debatin KM. TRAIL/Apo-2 ligand 906. Ju SK, Lee SC, Kwon TH, et al. Immunity to melanoma mediated
in activated mouse T and B cells. Eur J Immunol 1998; 28: 143. by 4-1BB is associated with enhanced activity of tumor infiltrat-
889. Jiang H, Lin JJ, Su ZZ, et al. Subtraction hybridization identifies ing lymphocytes. Immunol Cell Biol 2005; 83: 344–351.
a novel melanoma differentiation associated gene, mda-7, modu- 907. Jung HC, Eckmann L, Yang SK, et al. A distinct array of proin-
lated during human melanoma differentiation, growth and pro- flammatory cytokines is expressed in human colon epithelial
gression. Oncogene 1995; 11; 2477–2486. cells in response to bacterial invasion. J Clin Invest 1995; 95:
890. Jiang H, Su ZZ, Lin JJ, et al. Melanoma differentiation associated 55–65.
gene mda-7 suppresses cancer cell growth. Proc Natl Acad Sci 908. Jung T, Widjdeness J, Neumann C, de Vries JE, Yssl H.
USA 1996; 93: 9160–9165. Interleukin-13 is produced by activated CD45RA+ and CD45RO+
891. Jin L, Nakao A, Nakayama M, et al. Induction of RANTES by T cells: modulation by interleukin-4 and interleukin-12. E J
TWEAK/Fn14 interaction in human keratinocytes. J Invest Immunol 1996; 26: 571.
Dermatol 2004; 122: 1175–1179. 909. Junn E, Lee Ju HR, Han SH, et al. Requirement for hydrogen
892. Jin L, Raymond DP, Crabtree TD, Pelletier SJ, Houlgrave CW, peroxide generation in TGF-β1 signal transduction in human
Pruett TL, Sawyer RG. Enhanced murine macrophage TNF lung fibroblast cells: Involvement of hydrogen peroxide and Ca+
receptor shedding by cytosine guanine sequences in oligodeoxy- 2 in TGF-β1-induced IL-6 expression. J Immunol 2000; 165:
nucleotides. J Immunol 2000; 165: 5153–5160. 2190–2197.
893. Jinquan T, Deleuran B, Gesser B, et al. Regulation of human T 910. Kaashoek JG, Mout JR, Falkenburg JHF, et al. Cytokine produc-
lymphocyte chemotaxis in vitro by T cell derived cytokines IL-2, tion by the bladder carcinoma cell line 5637. Lymphokine
IFN-λ, IL-4, IL-10, and IL-13. J Immunol 1995; 154: Cytokine Res 1991; 10: 231.
3742–3752. 911. Kalinski P, Smits HH, Schuitemaker JHN, et al. IL-4 is a media-
894. Jinquan T, Jing C, Jacobi HH, et al. CXCR3 expression and acti- tor of IL-12p70 induction by human Th2 cells: reversal of polar-
vation of eosinophils: role of IFN-γ-inducible protein-10 and ized Th2 phenotype by dendritic cells. J Immunol 2000; 165:
monokine-induced by IFN-γ. J Immunol 2000; 165: 1548–1566. 1877–1881.
Walter M. Lewko and Robert K. Oldham 239

912. Kamanaka M, Yu P, Yasui T, et al. Protective role of CD40 in tors interleukin-2 and interleukin-4. J Immunotherapy 1993; 14;
Leishmania major infected at two distinct phases of cell mediated 336–347.
immunity. Immunity 1996; 4: 275. 934. Kawakami Y, Rosenberg SA, Lotze MT. Interleukin 4 promotes
913. Kaneda T, Nojima T, Nakagawa M, et al. Endogenous production the growth of tumor infiltrating lymphocytes cytotoxic for human
of TGF-β is essential for osteoclastogenesis induced by a combi- autologous melanoma. J Exp Med 1988; 168: 2183–2191.
nation of receptor activator of NF-κB ligand and macrophage- 935. Kawano M, Matsushima K, Masuda A, et al. A major 50 kDa
colony stimulating factor. J Immunol 2000; 165: 4254–4263. human B cell growth factor II induces both Tac antigen expres-
914. Kaneko H, Hori T, Yamagita S, et al. Introduction of OX40 ligand sion and proliferation by several lymphocytes. Cell Immunol
into lymphoma cells elicits anti-lymphoma immunity in vivo. 1988; 111: 273–286.
Exp Hematol 2005; 33: 336–343. 936. Kawasakura S, Lowenstein L. A factor stimulating DNA synthe-
915. Kang CM, Jang AS, Ahn MH, et al. Interleukin-25 and interleu- sis derived from the medium of leucocyte cultures. Nature 1965;
kin-13 production by alveolar macrophages in response to parti- 208: 794–795.
cles. Am J Respir Cell Mol Biol 2005; 33: 290–296. 937. Kawashima I, Ohsumi J, Mita-Honjo K, et al. Molecular cloning
916. Kang SM, Le D, Springer ML, Stock PG, Blau HM. Immune of the cDNA encoding adipogenesis inhibitory factor and identity
response and myoblasts that express Fas ligand. Science 1997; with interleukin 11. FEBS Lett 1991; 283: 199–202.
278: 1322. 938. Kay AB. Inflammatory blood cells in bronchial asthma. J Asthma
917. Kang WK, Park C, Yoon HL, et al. Interleukin 12 gene therapy of 1989; 26: 335.
cancer by peritumoral injection of transduced autologous fibroblasts: 939. Kayagaki N, Yamiguchi N, Nakayama M, et al. Type I interferons
outcome of a phase I study. Hum Gene Ther 2001; 10: 671–684. (IFNs) regulate tumor necrosis factor related apoptosis-inducing
918. Kang Y, He W, Tully S. Breast cancer bone metastasis mediated ligand (TRAIL) expression on human T cells: a novel mechanism
by the Smad pathway. Proc Natl Acad Sci USA 2005; 102: for the antitumor effects of type I interferons. J Exp Med 1999;
13909–13914. 189: 1451.
919. Kasaian MT, Whitters MJ, Carter LL, et al. IL-21 limits NK cell 940. Kaye J, Gillis S, Mizel SB, et al. Growth of a cloned helper T cell
responses and promotes antigen-specific T cell activation: a line induced by a monoclonal antibody specific for the antigen
mediator of the transition form innate to adaptive immunity. receptor: interleukin 1 is required for the expression of receptors
Immunity 2002; 16: 559. for interleukin 2. J Immunol 1984; 133: 1339–1345.
920. Kasama T, Strieter RM, Lukacs NW, Burdick MD, Kunkel SL. 941. Keane J, Nicoll J, Kim S, et al. Conservation of structure and
Regulation of neutrophil-derived chemokine expression by function between human and murine IL-16. J Immunol 1998;
IL-10. J Immunol 994; 152: 3559. 160: 5945–5954.
921. Kaser A, Dunzendorfer S, Offner FA, et al. A role of IL-16 in the 942. Keates AC, Castagliuolo I, Cruickshank WW, et al. Interleukin-16
cross-talk between dendritic and T cells. J Immunol 1999; 163: is upregulated in Crohn’s disease and participates in TNBS colitis
3232–3238. in mice. Gastroenterology 2000; 119: 972–982.
922. Kaser A, Dunzendorfer S, Offner FA, et al. B-lymphocyte- 943. Keegan AD, Johnson JA, Tortolani PJ, et al. Similarities and dif-
derived IL-16 attracts dendritic cells and Th cells. J Immunol ferences in signal transduction by interleukin-4 and interleu-
2000; 165: 2474–2480. kin-13: analysis of Janus kinase activation. Proc Natl Acad Sci
923. Kaspenberg ML, Wierenga EA, Bos JD, Jansen HM. Functional USA 1995; 92: 7681–7685.
subsets of allergen-reactive human CD4+ T cells. Immunol Today 944. Keegan AD, Pierce JH. The interleukin-4 receptor: signal transduc-
1991; 12: 392. tion by a hematopoietic receptor. J Leuk Biol 1994; 5: 272–279.
924. Katayama N, Clark SC, Ogawa M. Growth factor requirement 945. Kehri JH, Dukovich M, Whalen G, et al. Novel interleukin 2
for survival in cell-cycle dormancy of primitive murine lympho- receptor appears to mediate IL-2-induced activation of natural
hematopoietic progenitors. Blood 1993; 81: 610. killer cells. J Clin Invest 1988; 81: 779.
925. Katsura K, Park M, Gatanaga M, et al. Identification of the prote- 946. Kehry MR. CD40-mediated signaling in B cells: balancing cell
olytic enzyme which cleaves human p75 TNF receptor in vitro. survival, growth and death. J Immunol 1996; 156: 2345–2348.
Biochem Biophys Res Commun 1996; 222: 298–302. 947. Kelleher K, Bean K, Clark SC, et al. Human intertleukin-9:
926. Katz A, Shulman LM, Porgador A, et al. Abrogation of B16 mel- genomic sequence, chromosomal location, sequences essential
anoma metastases by long term low dose interleukin 6 therapy. J for its expression in human T-cell leukemia virus HTLV-1 trans-
Immunother Emphasis Tumor Immunol 1993; 13: 98–109. formed human T cells. Blood 1991; 77: 1436–1441.
927. Katz A, Shulman LM, Revel M, et al. Combined therapy with 948. Kellokumpu-Lehtinen P, Talpaz M, et al. Leukemia inhibitory
IL-6 and inactivated tumor cell suppressed metastasis in mice factor stimulates breast, kidney and prostate cancer cell prolifera-
bearing 3LL lung carcinomas. Int J Cancer 1993; 53: 812–818. tion by paracrine and autocrine pathways. Int J Cancer 1996; 66:
928. Kaufmann Y, Armiglio N, Rosenthal E, et al. Proliferation 515–519.
response of leukemic cells to CD70 ligation oscillates with recur- 949. Kelner GS, Kennedy SJ, Bacon KB, et al. Lymphotactin: a
rent remission and relapse in a low-grade lymphoma. J Immunol cytokine that represents a new class of chemokine. Science 1994;
2005; 175: 6940–6947. 266: 1395.
929. Kawaguchi M, Kokubu F, Odaka M, et al. Induction of granulo- 950. Kemeny L, Ruzicka T, Dobozy A, Michel G. Role of Interleukin
cyte-macrophage colony stimulating factor by a new cytokine, 8 receptor in skin. International Arch Allergy and Immunology
ML-1 (IL-17F), via Raf I-MEK-ERK pathway. J Allergy Clin 1994; 104: 317–322.
Immunol 2004; 114: 444–450. 951. Kennedy J, Rossi DL, Zuraski SM, et al. Mouse IL-17: a cytokine
930. Kawaguchi M, Onuchic LF, Li X-D, et al. Identification of a preferentially expressed by αβTCR+ CD4−CD8− T cells. J
novel cytokine, ML-1, and its expression in subjects with asthma. Interferon Cyto Res 1996; 16: 611–617.
J Immunol 2001; 167: 4430–4435. 952. Kennedy MK, Mohler KM, Shanebeck KD, et al. Induction of B
931. Kawaguchi M, Takahashi D, Hizawa N, et al. IL-17F sequence cell costimulatory function of recombinant murine CD40 ligand.
variant (His161Arg) is associated with protection against asthma E J Immunol 1994, 24: 116.
and antagonizes wild-type IL-17 activity. J Allergy Clin Immunol 953. Kenyon BM, Browne F, D’Amato RJ. Effects of thalidomide and
2006: 117: 795–801. related metabolites on a mouse corneal model for neovascular-
932. Kawakami Y, Custer M, Rosenberg SA, Lotze M. Interleukin-4 ization. Exp Eye Res 1997; 64: 971–978.
regulates Interleukin-2 induction of lymphokine activated killer 954. Kern PA Saghizadeh M, Ong JM, et al. The expression of tumor
activity from human leukocytes. J Immunol 1989; 142: 3452. necrosis factor in human adipose tissue. Regulation by obesity,
933. Kawakami Y, Haas, GP, Lotze MT. Expansion of tumor infiltrat- weight loss and relationship to lipoprotein lipase. J Clin Invest
ing lymphocytes from human tumors using the T cell growth fac- 1995; 95: 2111–2119.
240 Cytokines

955. Khan SY, Kelher MR, Heal JM, et al. Soluble CD40L accumu- 977. Kitamura T, Sato N, Arai K, Miyajima A. Expression cloning of
lates in stored blood components, primes neutrophils through the human IL-3 receptor cDNA reveals a shared β subunit for
CD40, and is a potential cofactor in the development of transfu- human IL-3, and GM-CSF receptors. Cell 1991; 66: 1165–1174.
sion-related acute lung injury. Blood 2006; 108: 2455–2562. 978. Kitamura Y, Go S, Hatanaka K. Decrease of mast cells in W/Wv
956. Khapli SM, Mangashetti LS, Yogesha SD, et al. IL-3 acts directly mice and their increase by bone marrow transplantation. Blood
on osteoclast precursors and irreversibly receptor activation of 1978; 52: 447.
NF-κB ligand-induced osteoclast differentiation by diverting the 979. Kitamura Y, Go S. Decreased production of mast cells in S1/S1d
cells to macrophage lineage. J Immunol 2003; 171: 142–151. anemic mice. Blood 1979; 53: 492.
957. Khatri VP, Caligiuri MA. A review of the association between 980. Kitazawa H, Muegge K, Badolato R, et al. IL-7 activates α4β1
interleukin-10 and human B-cell malignancies. Cancer Immunol integrin in murine lymphocytes. J Immunol 1997; 159:
Immunother 1998; 46: 239–244. 2259–2264.
958. Kieth JC, Albert L, Sonis ST, Pfieffer CJ, Schaub RG. IL-11, a 981. Kituchi T, Joki T, Saitoh S, et al. Antitumor activity of interleu-
plieotropic cytokine: exciting new effects on gastrointestinal kin-2-producing tumor cells and recombinant interleukin-12
mucosal biology. Stem Cells 1994; 12: (Suppl 1): 79–89. against mouse glioma cells located in the central nervous system.
959. Kilian PL, Kaffka KL, Biondi KA, et al. Antiproliferative effect Int J Cancer 1999; 425–430.
of interleukin-1 on human ovarian carcinoma cell line (NIH: 982.Kivisakk P, Matusevicius B, He B, et al. IL-15 mRNA expression is
OVCAR-3). Cancer Res 1991; 51: 1823–1828. up-regulated in blood and cerebrospinal fluid mononuclear cells in
960. Kilinc MO, Aulakh KS, Nair RE, et al. Reversing tumor immune multiple sclerosis (MS). Clin Exp Immunol 1998; 111: 193.
suppression with intratumoral IL-12: Activation of memory asso- 983. Klebanoff SJ, Vadas MA, Harlam JM, et al. Stimulation of neu-
ciated T effector/memory cells, induction of T suppressor apop- trophils by tumor necrosis factor. J Immunol 1986; 136: 4220.
tosis, and infiltration of CD8+ T effectors. J Immunol 2006; 177: 984. Klein U, Rajewsky K, Kuppers R. Human immunoglobulin (Ig)
6962–6973. M+ IgD+ peripheral blood B cells expressing the CD27 cell sur-
961. Kim J, Sanders SP, Sierkierski ES, Casolaro V, Proud D. Role of face antigen carry somatically mutated variable region genes:
NF-κB in cytokine production induced from human airway epithelial CD27 as a general marker for somatically mutated (memory) B
cells by rhinovirus infection. J Im-munol 2000; 165: 3384–3392. cells. J Exp Med 1998; 188: 1679.
962. Kim KE, Song H, Kim TS, et al. Interleukin-18 is a critical factor 985. Kleinschek MA, Owyang AM, Joyce-Shaikh B, et al. IL-25 regu-
for vascular endothelial growth factor-induced migration in gas- lates Th17 function in autoimmune inflammation. J Exp Med
tric cancer cell lines. Oncogene 2006; 9: 1480–1486. 2007; 204: 161–170.
963. Kim S-H, Eisenstein M, Resnikov L, et al. Structural require- 986. Klimiuk PA, Goronzy JJ, Weyland CM. IL-16 as an anti-inflam-
ments of six naturally occurring isoforms of the interleukin 18 matory cytokine in rheumatoid arthritis. J Immunol 1999; 162:
binding protein to inhibit interleukin 18. Proc Natl Acad Sci USA 4293–4299.
2000; 97: 1190. 987. Klouche M, Bhakdi S, Hemmes M, Rose-John S. Novel path of
964. Kim SH, Han SY, Azam T, et al. Interleukin-32: a cytokine and activation of primary smooth muscle cells: up-regulation of
inducer of TNFα. Immunity 2005; 22: 131–142. gp130 creates an autocrine activation loop by IL-6 and its soluble
965. Kim SH, Reznikov LL, Stuyt RJL, et al. Functional reconstitu- receptor. J Immunol 1999; 163: 4583.
tion and regulation of IL-18 activity by the IL-18Rβ Chain. J 988. Knappe A, Hor S, Wittmann S, Flickenscher H. Induction of a
Immunol 2001; 166: 148–154. normal cellular homolog of IL-10, AK155, by transformation
966. Kim SW, Hayashi M, Lo JF, et al. Tid 1 negatively regulates the of T lymphocytes with herpesvirus saimiri. J Virol 2000; 74:
migratory potential of cancer cells by inhibiting the production of 3881–3887.
interleukin-8. Cancer Res 2005; 65: 8784–8791. 989. Knapper S, Burnett AK, Littlewood T, et al. A phase 2 trial of the
967. Kim SW, Kang YJ, Kim WJ, et al. TWEAK can induce proin- flt3 inhibitor lestaurtinib (CEP701) as first-line treatment for
flammatory cytokines and matrix metalloproteinase-9 in mac- older patients with acute myeloid leukemia not considered fit for
rophages. Circ J 2004; 68: 396–399. intensive chemotherapy. Blood 2006; 108: 3262–3270.
968. Kimura Y, Takeshita T, Kondo M, et al. Sharing of the IL-2 recep- 990. Kneitz B, Herrmann T, Yonehara S, Schimpl A. Normal clonal
tor γ chain with the functional IL-9 receptor complex. Int expansion but impaired Fas-mediated cell death and anergy induc-
Immunol 1995; 7: 115–120. tion in interleukin-2 deficient mice. Eur J Immunol 1995; 25: 2572.
969. Kinashi T, Harada N, Severinson E, et al. Cloning of complemen- 991. Knopf M, Baumann H, Freer G, et al. Impaired immune and
tary DNA encoding T-cell replacing factor and the identity of acute phase responses in interleukin-6-deficient mice. Nature
B-cell growth factor II. Nature 1986; 324: 70–73. 1994; 368: 339–342.
970. Kincaid PW, Medina KL, Smithson G. Sex hormones are nega- 992. Kobata T, Agematsu K, Kameoka J, Schlossman SF, Morimoto
tive regulators of lymphopoiesis. Immunol Rev 1994; 137: 119. C. CD27 is a signal transducing molecule involved in CD45RA+
971. Kirkwood JM, Wilson J, Whiteside TL, Donnelly S, Herberman naive T cell costimulation. J Immunol 1994; 153: 5422.
RB. Phase IB trial of picibanil (OK-432) as an immunomodulator 993. Kobayashi M, Fitz L, Ryan M, et al. Identification and purifica-
in patients with resected high risk melanoma. Cancer Immunol tion of natural killer cell stimulatory factor (NKSF), a cytokine
Immunother 1997; 44: 137–149. with multiple biological effects on human lymphocytes. J Exp
972. Kirman I, Nielsen OH. Increased numbers of interleukin-15-ex- Med 1989; 170: 827–845.
pressing cells in active ulcerative colitis. Am J Gastroenterology 994. Koch AE, Polverini PK, Kunkel SL, et al. Interleukin 8 as a mac-
1996; 91: 1789. rophage-derived mediator of angiogenesis. Science 1992; 258:
973. Kirnbauer R, Charvat B, Schauer E, et al. Modulation of intracel- 1798–1801.
lular adhesion molecule 1 expression on human melanocytes and 995. Kodama T, Matsuyama T, Kuribashi K, et al. IL-18 deficiency
melanoma cells: evidence for a regulatory role of IL-6, IL-7 selectively enhances allergy induced eosinophilia in mice. J
TNFβ, and UVB light. J Invest Dermat 1992; 98: 320–326. Allergy Clin Immunol 2000; 105: 45.
974. Kishimoto T, Akira S, Taga T. Interleukin 6 and its receptor: a 996. Kohase M, May LM, Tamm I, Vilcek J, Sehgal PB. A cytokine
paradigm for cytokines. Science 1992; 258: 593–597. network in human diploid fibroblasts: interactions of β interfer-
975. Kishimoto T, Hirano T. Molecular regulation of B lymphocyte ons, tumor necrosis factor, platelet-derived growth factor and
response. Ann Rev Immunol 1988; 6: 485–512. interleukin 1. Mol Cell Biol 1987; 7: 273.
976. Kitadai Y, Haruma K, Sumii K, et al. Expression of interleukin-8 997. Kohn DB, Weinberg KI, Nolta JA, et al. Engraftment of gene-
correlates with vascularity in human gastric carcinomas. Am J modified umbilical cord blood cells in neonates with adenosine
Pathol 1998; 152: 93. deaminase deficiency. Nat Med 1995; 1: 1017.
Walter M. Lewko and Robert K. Oldham 241

998. Kohno K, Kataoka T, Ohtsuki Y, et al. IFN-γ-inducing factor activated killer cells: early induction of interleukin 1β by inter-
(IGIF) is a costimulatory factor on the activation of Th1 but not leukin 2. Cancer Res 1989; 49: 940.
Th2 cells and exerts its effect independently of IL-12. J Immunol 1017. Kovacs JA, Vogel S, Albert JM, et al. Controlled trial of inter-
1997; 158: 1541. leukin-2 infusions in patients infected with the human immuno-
999. Koi A. The role of interleukin 6 as the hepatocyte-stimulating deficiency virus. N Eng J Med 1996; 335: 1350–1356.
factor in the network of inflammatory cytokines. Ann NY Acad 1018. Kradin RL, Kurnick JT, Lazarus DS, et al. Tumor-infiltrating
Sci 1989; 557: 1–8. lymphocytes and interleukin-2 in treatment of advanced cancer.
1000. Kojima H, Takeuchi M, Ohta T, et al. Interleukin-18 activates Lancet 1989; 1: 577–580.
the IRAK-TRAF6 pathway in mouse EL-4 cells. Biochem 1019. Kramer PH. CD90 (Apo/Fas)-mediated apoptosis: live and let
Biophys Res Communic 1998; 244: 183. die. Adv Immunol 1999; 71: 163.
1001. Komschlies KL, Back TT, Gregorio TA, et al. Effects of rhIL-7 1020. Kratz A, Campos-Neto A, Hanson MS, Ruddle NH. Chronic
on leukocyte subsets in mice: implications for antitumor activ- inflammation caused by lymphotoxin is lymphoid neogenesis. J
ity. Immunology 1994; 61: 95–104. Exp Med 1996; 183: 1461.
1002. Kondo M, Takeshita T, Higuchi M, et al. Functional participa- 1021. Krautwald S. IL-16 activates the SPAK signaling pathway in
tion of the IL-2 receptor γ chain in IL-7 receptor complexes. CD4+ macrophages. J Immunol 1998; 160: 5874–5879.
Science 1994; 263: 1453–1454. 1022. Krenger W, Snyder K, Smith S, Ferrara JL. Effects of exogenous
1003. Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of IL-10 in a murine model of graft-versus-host disease to minor
osteoclastogenesis, lymphnode development and lymph node histocompatibility antigens. Transplantation 1994; 58: 1251.
organogenesis. Nature 1999; 397: 315. 1023. Kreuger J, Ray A, Tamm I, et al. Expression and function of
1004. Koni PA, Sacca R, Lawton P, et al. Distinct roles of lymphoid IL-6 in epithelial cells. J Cell Biol 1991; 45: 327.
organogenesis for lymphotoxins α and β revealed in lymphoto- 1024. Kroncke R, Loppnow H, Flad HD, Gerdes J. Human follicular
xin β-deficient mice. Immunity 1997; 6: 491. dendritic cells and vascular cells produce interleukin 7: a poten-
1005. Koo AS, Armstrong C, Bochner B, et al. Interleukin-6 and renal tial role for interleukin-7 in the germinal center reaction. Eur J
cell cancer: production, regulation, and growth effects. Cancer Immunol 1996; 26: 2541–2544.
Immunol Immunother 1992; 35: 97–105. 1025. Kryczek I, Wei S, Zou L, et al. Th17 and regulatory T cell
1006. Kopf M, Ruedl C, Schmitz M, et al. OX40-deficient mice are dynamics and the regulation by IL-2 in the tumor microenviron-
defective in Th cell proliferation but are competent in generat- ment. J Immunol 2007; 178: 6730–6733.
ing B cell and CTL responses after virus infection. Immunity 1026. Ku C, Murakami M, Sakamoto A, Kappler J, Marrack P. Control
1999; 11: 699. of homeostasis of CD8+ memory T cells by opposing cytokines.
1007. Kopp WC, Urba WJ, Rager HC, et al. Induction of IL-1R antag- Science 2000; 288: 675.
onist after interleukin 1 therapy in patients with cancer. Clin 1027. Ku H, Yonemura Y, Kaushansky K, Ogawa M. Thrombopoietin,
Cancer Res 1996; 2: 501–506. the ligand for the Mpl receptor, synergizes with steel factor and
1008. Korherr C, Hofmeister R, Wesche H, Falk W. A critical role for other early acting cytokines in supporting proliferation of primi-
the interleukin-1 receptor accessory protein in interleukin-1 tive hematopoietic progenitors of mice. Blood 1996; 87: 4544.
signaling. Eur J Immunol 1997; 27: 262. 1028. Kubin M, Kamoun M, Trinchieri G. Interleukin 12 synergizes
1009. Korzus E, Nagase H, Rydell R, Travis J. The mitogen-activated with B7/CD28 interaction in inducing efficient proliferation
protein kinase and JAK-STAT signaling pathways are required and cytokine production of human T cells. J Exp Med 1994;
for an oncostatin M-responsive element-mediated activation of 180: 211–222.
matrix metalloproteinase 1 gene expression. J Biol Chem 1997; 1029. Kudo-Sato C, Hodge JW, Kwak H, et al. 4-1BB ligand enhances
272: 1188. tumor specific immunity of poxvirus vaccines. Vaccine 2006;
1010. Kosugi H, Nakagawa Y, Hotta T, et al. Structure of the gene 24: 4975–4986.
encoding the α subunit of the human interleukin-3 receptor. 1030. Kuestner RE, Taft DW, Haran A, et al. Identification of the
Biochem Biophys Res Commun 1995; 208: 360. IL-17 receptor related molecule IL-17RC as a receptor for
1011. Kotake S, Udagawa N, Takahashi N, et al. IL-17 in synovial IL-17F. J Immunol 2007; 179: 5462–5473.
fluid from patients with rheumatoid arthritis is a potent stimula- 1031. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W.
tor of osteoclastogenesis. J Clin Invest 1999; 103: 1345. Interleukin-10 deficient mice develop chronic enterocolitis. Cell
1012. Kotenko SV, Izotova LS, et al. Identification of the functional 1993; 75: 263–274.
IL-TIF (IL-22) receptor complexes: the IL-10R2 chain 1032. Kuhn R, Rajewski K, Muller W. Generation and analysis of
(IL-10Rβ) is a shared component of both IL-10 and IL-TIF Interleukin-4 deficient mice. Science 1991; 254: 707–709.
(IL-22) receptor complexes. J Biol Chem 2000; 276: 2725. 1033. Kumano K, Nakao A, Nakajima H, et al. Interleukin-18
1013. Kotenko SV, Izotova LS, Mirochnitchenko OV, et al. enhances antigen induced eosinophilia recruitment into the
Identification, cloning and characterization of a novel soluble mouse airways. Am J Respir Care Med 1999; 160: 873.
receptor that binds IL-22 and neutralizes its activity. J Immunol 1034. Kumanogoh A, Wang X, Lee I, et al. Increased T-cell autoreactivity
2001; 166: 7096–7103. in the absence of CD40–CD40 ligand interactions: A role of CD40
1014. Kotenko SV, Krause CD, et al. Identification and functional in regulatory T cell development. J Immunol 2001; 166: 353–360.
characterization of a second chain of the interleukin-10 receptor 1035. Kumaraguru U, Davis IA, Deshpande S, Tevethia SS, Rouse
complex. EMBO J 1997; 16: 5894. BT. Lymphotoxin α−/−mice develop functionally impaired CD8+
1015. Kotowicz K, Callard RE. Human immunoglobulin class and IgG T cell responses and fail to contain virus infection of the central
subclass regulation: dual action of interleukin 4. Eur J Immunol nervous system. J Immunol 2001; 166: 1066–1077.
1993; 23: 2250. 1036. Kupper T, Horowitz M, Lee F, et al. Autocrine growth of T cells
1016. Kovacs EJ, Beckner SK, Longo DL, et al. Cytokine gene independent of growth factor for a cloned antigen specific
expression during the generation of human lymphokine- helper T cell. J Immunol 1987; 138: 4280.
242 Cytokines

1037. Kurek B, Austin L, Cheema SS, Bartlett PF, Murphy M. 1055. Lane P. Brocker T, Hubele S, et al. Soluble CD40 ligand can
Upregulation of leukemia inhibitory factor and interleukin-6 in replace the normal T cell-derived CD40 ligand to signal to B
transected sciatic nerve and muscle following-denervation. cells in T cell-dependent activation. J Exp Med 1993; 177:
Neuromusc Disord 1996; 6: 105. 1209.
1038. Kurschner C and Yusaki M. Neuronal interleukin-16: A dual 1056. Lang D, Knop J, Wesche H, et al. The type II IL-1 receptor
function PZP domain protein. J Neuroscience 1999; 19: interacts with the IL-1 receptor accessory protein: a novel
7770–7780. mechanism of regulation of IL-1 responsiveness. J Immunol
1039. Kurt-Jones EA, Beller DI, Mizel SB, Unanui ER. Identification 1998; 161: 6871.
of a membrane-associated interleukin 1. Proc Natl Acad Sci 1057. Langowski JL, Zhang X, Wu L, et al. IL-23 promotes tumor
(USA) 1985; 82: 1204–1208. incidence and growth. Nature 2006; 442: 461–465.
1040. Kurt-Jones EA, Hamburg S, Ohara J, et al. Heterogeneity of 1058. Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a
helper/inducer T lymphocytes. J Exp Med 1987; 166: pathogenic T cell population that induces autoimmune inflam-
1774–1787. mation. J Exp Med 2005; 210: 233–240.
1041. Kurts C, Carbone FR, Krummel MF, et al. Signaling through 1059. Langstein J, Michel J, Fritsche J, et al. CD137 (ILA/4-1BB), a
CD30 protects against autoimmune diabetes mediated by CD8 member of the TNF receptor family, induces monocyte activa-
T cells. Nature 1999; 398: 341. tion via bidirectional signaling. J Immunol 1998; 160:
1042. Kurzrock RZ, Estrov M, Wetzler M, Gutterman JU, Talpaz M. 2488–2494.
LIF: not just a leukemia inhibitory factor. Endocrinol Rev 1991; 1060. Lantz M, Gullberg U, Nilsson E, Olsson I. Characterization in
12: 208. vitro of a human tumor necrosis factor binding protein, a soluble
1043. Kuwana M, Medsger TA, Wright TM. Analysis of soluble and form of the tumor necrosis factor receptor. J Clin Invest 1990;
cell surface factors regulating anti-DNA topoisomerase I 86: 1396.
autoantibody production demonstrates synergy between Th1 1061. Lanz CS, Boesiger J, Song CH, et al. Role of interleukin-3 in
and Th2 autoreactive cells. J Immunol 2000; 164: 6138–6146. mast cell and basophil development and in immunity to para-
1044. Kwon BS, Tan KB, Ni J, Lee NO, et al. A newly identified mem- sites. Nature 1998; 392: 90.
ber of the tumor necrosis factor superfamily with a wide tissue 1062. Larmann K, Hughes CCW, Schechner J, Fanslow WC, Pober
distribution and involvement in lymphocyte activation. J Biol JS. CD40 on human endothelial cells: inducibility by cytokines
Chem 1997; 272: 14272. and functional regulation of adhesion molecule expression.
1045. Kwon BS, Weissman SM. cDNA sequences of two inducible Proc Natl Acad Sci USA 1995; 92: 4342.
T-cell genes. Proc Natl Acad Sci USA 1989; 86: 1993–1993. 1063. Larsen CG, Anderson AO, Oppenheim JJ, Matsushima K.
1046. Laan M, Cui Z-H, Hoshino H, et al. Neutrophil recruitment by Production of Interleukin 8 by human dermal fibroblasts and
human IL-17 via C-X-C chemokine release in the airways. J keratinocytes in response to interleukin 1 or tumor necrosis fac-
Immunol 1999; 162: 2347–2352. tor. Immunology 1989; 68: 31.
1047. Labarge S, Cruikshank WW, Beer DJ, Center DM. Secretion of 1064. Larsen CP, Alexander DZ, Hollenbaugh D, et al. CD40–gp39
IL-16 (Lymphocyte chemoattractant factor) from serotonin- interactions play a critical role during allograft rejection.
stimulated CD8+ T cells in vitro. J Immunol 1996; 156: Transplantation 1996; 61: 4.
310–315. 1065. Lasfar A, Lewis-Antes A, Smirnov SV, et al. Characterization of
1048. Labarge S, Cruikshank WW, Kornfeld H, Center DM. Histamine the mouse interferon-λ Ligand–receptor system: IFN-λs exhibit
induced secretion of lymphocyte chemoattractant factor from antitumor activity against B16 melanoma. Cancer Res 2006; 66:
CD8+ T cells is independent of transcription and translation: 4468–4477.
evidence for constitutive protein synthesis and storage. J 1066. Lauw FN, Pajkrt D, Hack CE, et al. Proinflammatory effect of
Immunol 1995; 155: 2902–2910. IL-10 during human endotoxemia. J Immunol 2000; 165:
1049. Laberge S, Pinsonneault S, Varga EM, et al. Increased expres- 2783–2789.
sion of IL-16 immunoreactivity in bronchial mucosa after seg- 1067. Le PT, Lazorick S, Whichard LP, et al. Human thymic epithelial
mental allergen challenge in patients with asthma. J Allergy cells produce IL-6, granulocyte-monocyte-CSF and leukemia
Clin Immunol 2000; 106: 293–301. inhibitory factor. J Immunol 1990; 145: 3310.
1050. Lacey DL, Tims E, Tan H-L, et al. Osteoprotegerin ligand is a 1068. Leali D, Dell’Era P, Stabile H, et al. Osteopontin (Eta-1) and
cytokine that regulates osteoclast differentiation and activation. fibroblast growth factor-2 cross-talk in angiogenesis. J Immunol
Cell 1998; 93: 165. 2003; 171: 1085–1093.
1051. Lachman LB, Dinarello CA, Llanska ND, Fidler IJ. Natural and 1069. Lean JM, Fuller K, Chambers TJ. FLT3 ligand can substitute for
recombinant human interleukin-1β is cytotoxic for human mel- macrophage-colony stimulating factor in support of osteoclast
anoma cells. J Immunol 1986; 136: 3098–3103. differentiation and function. Blood 2001; 98: 2707–2713.
1052. Lacraz S, Nicod LP, Chicheportiche R, Welgus HG, Dayer JM. 1070. Lebedeva IV, Sauane M, Gopalkrishnan RV, et al. mda-7/IL-24:
IL-10 inhibits metalloproteinase and stimulates TIMP-1 pro- exploiting cancer’s Achilles’ heal. Mol Ther 2005; 11: 4–18.
duction in human mononuclear phagocytes. J Clin Invest 1995; 1071. Lebedeva IV, Su ZZ, Chang Y, et al. The cancer suppressor gene
96: 2304. mda-7 induces apoptosis in human melanoma cells. Oncogene
1053. Lahn M, Kalaharadi H, Mittlestadt P, et al. Early preferential 2002; 21: 708–718.
stimulation of γδ T cells by TNF-α. J Immunol 1998; 160: 1072. Lechan RM, Toni R, Clark BC, et al. Immunoreactive
5221–5230. interleukin-1β localization in the rat forebrain. Brain Res 1990;
1054. Laky K, Lefrancois L, Puddington L. Age-dependent intestinal 514: 135.
lymphoproliferative disorder due to stem cell factor receptor 1073. Lee J, Ho W-H, Maruoka M, et al. IL-17E, a novel proinflam-
deficiency: Parameters in small and large intestine. J Immunol matory ligand for the IL-17 receptor homolog IL-17Rh1. J Biol
1997; 158: 1417–1427. Chem 2001; 276: 1660–1664.
Walter M. Lewko and Robert K. Oldham 243

1074. Lee J, Horuk R, Rice GC, et al. Characterization of two high 1091. Lembersky B, Baldisseri M, Kunschner A, et al. Phase I-II study
affinity human interleukin-8 receptors. J Biol Chem 1992; 267: of IP low dose interleukin-2 in refractory stage III ovarian can-
16283. cer (meeting abstract). Proc Annu Meet Am Soc Clin Oncol
1075. Lee P, Wang F, Kuniyoshi J, et al. Effects of interleukin-12 on 1989; 8: A636.
the immune response to a multipeptide vaccine for resected 1092. Lenardo M. Interleukin-2 programs mouse αβ T lymphocytes
metastatic melanoma. J Clin Oncol 2001, 19: 3836–3847. for apoptosis. Nature 1991; 353: 858.
1076. Lee S, Kaneko H, Sekigawa I, et al. Circulating interleukin-16 1093. Lens SM, Drillenburg P, den Drijver BF, et al. Aberrant expres-
in systemic lupus erythematosus. Br J Rheumatol 1998; 37: sion and reverse signaling of CD70 on malignant B cells. Brit J
1334–1337. Haematol 1999; 106: 491–503.
1077. Lee SC, Liu W, Dickson DW, Bronson CF, Berman JW. 1094. Lens SMA, de Jong R, Hooibrink B, et al. Phenotype and func-
Cytokine production by human microglia and astrocytes. tion of human B cells expressing CD70 (CD27 ligand). Eur J
Differential induction by lipopolysaccharide and IL-1 β. J Immunol 1996; 26: 2964–2971.
Immunol 1993; 150: 2659–2667. 1095. Lentsch A, Crouch LD, Jordan JA, et al. Regulatory effects of
1078. Lee SH, Aggarwal BB, Rinderknecht E, et al. The synergistic interleukin-11 during acute lung inflammatory injury. J
antiproliferative effect of γ-interferon and human lymphotoxin. Leukocyte Biol 1999; 66: 151.
J Immunol 1984; 133: 1083. 1096. Lenzi R, Kudelka MP, Verschraegen C, Nash M, et al.
1079. Lee SJ, Drabik K, Van Wagner NJ, et al. ICAM-induced expres- Recombinant human interleukin-12 (RHIL-12) in patients with
sion of pro-inflammatory cytokines in astrocytes: Involvement ovarian and gastrointestinal cancers. Evidence of biological
of extracellular signal-regulated kinase and p38 mitogen-acti- activity and lack of significant toxicity with low-dose intraperi-
vated protein kinase pathways. J Immunol 2000, 165: toneal (IP) administration. Proc ASCO 1999; 18: 1721.
4658–4666. 1097. Leonard WJ, Noguchi M, Russel SM, McBride OW. The molec-
1080. Lee SW, Tsou AP, Chan H, et al. Glucocorticoids selectively ular basis of severe combined immunodeficiency: the role of the
inhibit the transcription of the interleukin 1β gene and decrease interleukin-2 receptor γ chain as a common γ chain, γc.
the stability of interleukin 1 beat mRNA. Proc Natl Acad Sci Immunological Reviews 1994; 138: 61–86.
(USA) 1988; 85: 1204–1208. 1098. Lesback ME, McKenna HJ, Hoek JA, et al. Safety of FLT3
1081. Lee SY, Park CG, Choi Y. T cell receptor-dependent cell death ligand in healthy volunteers. Blood 1997; 90: 751a.
of T cell hybridomas mediated by the CD30 cytoplasmic domain 1099. Lesser SS, Lo D. CNTFII, I presume? Nat Neurosci 2000; 3: 851.
in association with tumor necrosis factor reeptor-associated fac- 1100. Lesslauer W, Tabuchi H, Gentz R, et al. Recombinant soluble
tors. J Exp Med 1996; 183: 669. tumor necrosis factor receptor protects mice from lipopolysac-
1082. Lee TY, Koo AS, Peyret C, et al. The effect of IL-6 on TIL charide lethality. Eur J Immunol 1991; 21: 2883.
derived from human renal cell carcinoma. J Urol 1991; 145: 1101. Lester MR, Hofer, MF, Gately M, Trumble A, Leung DYM.
663–667. Down-regulating effects of IL-4 and IL-10 on the IFN-γ response
1083. Leeuenberg JF, Dententer MA, Buurman WA. Lipopolysaccharide in atopic dermatitis. J Immunol 1995; 154: 6174–6181.
LPS-mediated soluble TNF release and TNF receptor expres- 1102. Leung BP, Culshaw S, Gracie JA, et al. A role for IL-18 in neu-
sion by monocytes: role of CD14, LPS binding protein and trophil activation. J Immunol 2001; 167: 2879–2886.
bactericidal/permeability increasing protein. J Immunol 1994; 1103. Leung BP, McInnes IB, Esfandiari E, Wei X-Q, Liew FY.
152: 5070. Combined effects of IL-12 and IL-18 on the induction of colla-
1084. Leibovich SJ, Polverini PJ, Shepard HM, et al. Macrophage gen-induced arthritis. J Immunol 2000: 164: 6495–6502.
induced angiogenesis is mediated by tumor necrosis factor-α. 1104. Leung SX, Elias JA. Interleukin-11 inhibits macrophage inter-
Nature 1987; 329: 630. leukin-12 production. J Immunol 1997; 159: 2161.
1085. Leite-de-Moraes MC, Hameg A, et al. IL-18 enhances IL-4 pro- 1105. Levi-Schaffer F, Rubinchik E. Activated mast cells are fibro-
duction by ligand-activated NKT lymphocytes: a pro-Th2 effect genic for 3T3 fibroblasts. J Invest Dermatol 1995; 104: 999.
of IL-18 exerted through NK cells. J Immunol 2001: 166: 1106. Levitsky JW, Simons H, Vogelstein B, Frost P. Interleukin 2
945–951. produced by tumor cell bypasses T helper function in the gen-
1086. Leite-de-Moraes MC, Herbelin A, et al. MHC class I selected eration of an antitumor response. Cell 1990; 60: 397–403.
CD4-CD8-TCRαβ+ T cells are a potential source of IL-4 during 1107. Levy Y, Brouet JC. Interleukin-10 prevents spontaneous death
primary immune response. J Immunol 1995; 155: 4544. of germinal center B cells by induction of the Bcl-2 protein. J
1087. Leite-de-Moraes MC, Moreau G, et al. The IL-4 producing Clin Invest 1994; 93: 424.
NKT cells are biased towards IFN-γ production by IL-12: influ- 1108. Lewko WM, Good, RW, Bowman D, et al. Growth of tumor
ence of the microenvironment on the functional capacities of derived activated T cells for the treatment of cancer. Cancer
NKT cells. Eur J Immunol 1998; 28: 1507. Biotherapy 1994; 9(3): 211–224.
1088. Leleivre E, Plun-Favreau H, Chevalier S, et al. Signaling path- 1109. Lewko WM, Hall PB, Oldham RK. Growth of tumor derived
ways recruited by the cardiotrophin-like cytokine/cytokine-like activated T cells for the treatment of advanced cancer. Cancer
factor-1composite cytokine. J Biol Chem 2001; 276: 22476. Biotherapy and Radiopharmaceuticals 2000; 15: 357–366.
1089. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. 1110. Lewko WM, Smalley RV, Oldham RK. Lymphokines and Cytokines.
The dorsoventral regulatory gene cassette spatzle/Toll/cactus In: Oldham RK, ed. Principles of Cancer Biotherapy. Kluwer
controls the potent antifungal response in Drosophila adults. Academic Press, Dordrecht, The Netherlands, 1998: 211–265.
Cell 1996; 86: 973. 1111. Lewko WM, Smith TL, Bowman DJ, et al. Interleukin-15 and
1090. LeMay RG, Otterness IG, Vander AJ, Kluger MJ. In vivo evi- the growth of tumor derived activated T-cells. Cancer Biotherapy
dence that the rise in plasma IL-6 following injection of a 1995; 10: 13–20.
fever-inducing dose of LPS is mediated by IL-1β. Cytokine 1112. Li C-B, Gray PW, Lin PF, et al. Cloning and expression of
1990; 2: 199. lymphotoxin cDNA. J Immunol 1987; 138: 4496–4501.
244 Cytokines

1113. Li H, Chen J, Hung A, Stinson J, et al. Cloning and character- 1131. Lind M, Deleuran B, Yssl H, Fink-Eriksen E, Thestrup-Peterson
ization of IL-17B and IL-17C, two new members of the IL-17 K. IL-4 and IL-13 but not IL-10 are chemotactic factors for
cytokine family. Proc Natl Acad Sci USA 2000; 97: 773–778. human osteoblasts. Cytokine 1995; 7: 78.
1114. Li HH, Lin YC, Chen PJ, et al. Interleukin-19 upregulates kera- 1132. Lindgren CG, Thompson JA, Higuchi CM, Fefer A. Growth and
tinocyte growth factor and its association with psoriasis. Brit J autologous tumor lysis by tumor infiltrating lymphocytes from
Dermatol 2005; 153: 591–595. metastatic melanoma expanded in interleukin 2 or interleukin 2
1115. Li L, Elliott J, Mosmann T. IL-10 inhibits cytokine production, plus Interleukin 4.J Immunother 1993; 14: 322–328.
vascular leakage and swelling during T helper 1 cell induced 1133. Lindqvist C, Ostman AL, Okerblom C, Akerman K. Decreased
delayed-type hypersensensitivity. J Immunol 1994; 153: 3967. interleukin-2β chain receptor expression by interleukin-4 on
1116. Li Q, Carr A, Ito F, et al. Polarization effects of 4-1BB during LGL: influence on the IL-2 induced cytotoxicity and prolifera-
CD28 costimulation in generating tumor-reactive T cells for tion. Cancer Lett 1992; 64: 43–49.
cancer immunotherapy. Cancer Res 2003; 63: 2546–2552. 1134. Linsley PS, Ledbetter JA. The role of the CD28 receptor during
1117. Li Q, Carr AL, Donald EJ, et al. Synergistic effects of IL-12 and T cell responses to antigen. Annu Rev Immunol 1993; 11: 191.
IL-18 in skewing tumor-reactive T-cell responses towards a type 1135. Lisby S, Muller KM, Jongeneel CV, Saurat JH, Hauser C.
1 pattern. Cancer Res 2005; 65: 1063–1070. Nickle and skin irritants upregulate tumor necrosis factor
1118. Li WY, Lusheng S, Kanbour A, et al. Lymphocytes infiltrating mRNA in keratinocytes by differential but potentially synergis-
human ovarian tumors: synergy between tumor necrosis factor tic mechanisms. Int Immunol 1995; 7: 343–352.
and interleukin 2 in the generation of CD8+ effectors from 1136. Liu C, Hart RP, Liu X-J, et al. Cloning and characterization of
tumor infiltrating lymphocytes. Cancer Res 1989; 49: 5979. an alternately processed human type II interleukin-1 receptor
1119. Li XC, Roy-Chaudhury P, Hancock WW, et al. IL-2 and IL-4 mRNA. J Biol Chem 1996; 271: 20965.
double knockout mice reject islet allografts: a role for novel T 1137. Liu J, Hadjokas N, Mosey B, et al. Oncostatin M-specific recep-
cell growth factors in allograft rejection. J Immunol 1998; 161: tor expression and function in the regulation of cell proliferation
890–896. of normal and malignant mammary epithelial cells. Cytokine
1120. Li Y, Bleakley M, Yee C. IL-21 influences the frequency, pheno- 1998; 10: 295–302.
type and affinity of the antigen-specific CD8 T cell response. J 1138. Liu J, Modrell B, Aruffo A, et al. Interaction between oncostatin
Immunol 2005; 175: 2261–2269. M and the signal transducer, gp130. Cytokine 1994; 6: 272–278.
1121. Li Y, Yee C. IL-21 mediated Foxp3 suppression leads to 1139. Liu L, Ding C, Heuer JG, et al. Selective enhancement of multi-
enhanced generation of antigen-specific CD8+ T lymphocytes. potential hematopoietic progenitors in vito and in vivo by IL-20.
Blood 2008; 111: 229–235. Blood 2005; 117: 3206–3209.
1122. Liang SC, Long AJ, Bennett F, et al. An IL-17F/A heterodimer 1140. Liu X, Yue P, Schonthal AH, et al. Cellular FLICE-inhibitory
protein is produced by mouse Th17 cells and induces airway protein down regulation contributes to celecoxib-induced apop-
neutrophil recruitment. J Immunol 2007; 179: 7791–7799. tosis in human lung cancer cells. Cancer Res 2006; 66:
1123. Liang SC, Tan X-Y, Luxenberg DP, et al. Interleukin (IL)-22 11115–11119.
and IL-17 are coexpressed by Th17 cells and cooperatively 1141. Liu Y, Wu Y, Ramarathinam L, et al. Gene-targeted B-deficient
enhance expression of antimicrobial peptides. J Exp Med 2006; mice reveal a critical role for B cells in the CD4 cell response.
203: 2271–2279. Int Immunol 1995; 7: 1353.
1124. Liao Y-C, Cheng Y-C, Wang Y-C, et al. IL-19 induces Th2 1142. Llorente L, Richard-Patin Y, Garcia-Padilla C, et al. Clinical
cytokines and was upregulated in asthma patients. J Immunol and biological effects of anti-interleukin-10 monoclonal anti-
2004; 173: 6712–6718. body administration in systemic lupus erythematosis. Arthritis
1125. Liao Y-C, Liang W-G, Chen F-W, et al. IL-19 induces produc- Rheu 2000; 43: 1790–1800.
tion of IL-6 and TNF-α and results in cell apoptosis through 1143. Lo CH, Lee SC, Wu PY, et al. Antitumor and antimetastatic
TNF-α. J Immunol 2002; 169: 4288–4297. activity of IL-23. J Immunol 2003; 171: 600–607.
1126. Libby P. Warner SJ, Friedman GB, Interleukin1: a mitogen for 1144. Lockhart E, Green AM, Flnn JL. IL-17 production is dominated
vascular human smooth muscle cells that induces the release of by γδ T cells rather than CD4 T cells during Mycobacterium
growth-inhibitory prostanoids. J Clin Invest 1988; 81: tuberculosis infection. J Immunol 2006; 177: 4002–4009.
487–498. 1145. Lodolce JP, Boone DL, Chai S, et al. IL-15 receptor maintains
1127. Lichtman A, Kurt-Jones E, Abbas A. B cell stimulatory factor 1 lymphoid homeostasis by supporting lymphocyte homing and
and not interleukin 2 is and autocrine growth factor for some proliferation. Immunity 1998; 9: 669.
helper T lymphocytes. Proc Nat Acad Sci 1987; 84: 824. 1146. Loenen WAM, De Vries E, Gravestein LA, et al. The CD27
1128. Lienard D, Eggermont AM, Koops HS, et al. Isolated limb per- membrane receptor, a lymphocyte specific member of the nerve
fusion with tumor necrosis factor-α and melphalan with or with- growth factor family, gives rise to a soluble form by protein
out interferon-γ for the treatment of in-transit melanoma processing that does not involve endocytosis. Eur J Immunol
metastases: a multicentre ramdomized phase II study. Melanoma 1992; 22: 447–455.
Res 1999; 9: 491–502. 1147. Loetscher M, Gerber B, Loetscher P, et al. Chemokine receptor
1129. Lim KG, Wan HC, Bozza PT, et al. Human eosinophils elabo- specific for IP-10 and Mig: structure, function and expression in
rate the lymphocyte chemoattractants IL-16 (lymphocyte activated T-lymphocytes. J Exp Med 1996; 184: 963.
chemoattractant factor) and RANTES. J Immunol 1996; 156: 1148. Londei M, Verhoef A, Hawrylowicz C, et al. Interleukin 7 is a
2566–2570. growth factor for mature human T cells. Eur J Immunol 1990;
1130. Lin T-J, Issekutz TB, Marshall JS. Human mast cells transmi- 20: 425–428.
grate through human umbilical vein endothelial monolayers and 1149. London L, Perussiia B, Trinchieri G. Induction and proliferation
selectively produce IL-8 in response to stromal cell-derived in vitro of resting human natural killer cells: expression of sur-
factor-1α. J Immunol 2000; 165: 211–220. face activation antigens. J Immunol 1985; 134: 718.
Walter M. Lewko and Robert K. Oldham 245

1150. Lonnemann G, Shapiro L, Engler-Blum G, et al. Cytokines in 1168. Lubbert M, Mantovani L, Lindemann A, Mertlesmann R,
human renal interstitial fibrosis. I. Interleukin-1 is an autocrine Herrmann F. Expression of leukemia inhibitory factor is regu-
factor for cultured fibrosis-derived kidney fibroblasts. Kidney lated in human mesenchymal cells. Leukemia 1991; 5: 361.
Int 1995; 47: 845–854. 1169. Lubberts E, Joosten LAB, Opers B, et al. IL-1-independent role
1151. Lopez AF, Sanderson CJ, Gamble JR, et al. Recombinant human of IL-17 in synovial inflammation and joint destruction during
interleukin 5 is a selective activator of human eosinophil func- collagen-induced arthritis. J Immunol 2001; 167: 1004–1013.
tion. J Exp Med 1988; 167: 219. 1170. Luger TZ, Stadler BM, Katz SI, et al. Epidermal cell
1152. Lopez MV, Adris SK, Bravo AI, et al. IL-12 and IL-10 expres- (keratinocyte)-derived thymocyte-activating factor (ETAF). J
sion synergize to induce immune-mediated eradication of estab- Immunol 1981; 127: 1493–1498.
lished colon and mammary tumors and lung metastases. J 1171. Lundin J, Kimby E, Bergman L, et al. Interleukin 4 therapy for
Immunol 2005; 175: 5885–5894. patients with chronic lymphocytic leukemia: a phase I/II study.
1153. Lorenz MG, Kantor JA, Schlom J, et al. Antitumor immunity Br J Haematol 2001: 112: 155–160.
elicited by a recombinant vaccinia virus expressing CD70 1172. Lundqvist A, Abrams SI, Schrump DS, et al. Bortezomib and
(CD27L). Human Gene Ther 1999; 10: 1095–1103. depsipeptide sensitize tumors to tumor necrosis factor-related
1154. Lorenzen J, Lewis CE, McCracken D, et al. Human tumor asso- apoptosis-inducing ligand: a novel way to potentiate natural
ciated NK cells secrete increased amounts of interferon-γ and killer cell tumor cytotoxicity. Cancer Res 2006; 66: 7317–7325.
interleukin-4. British J Cancer 1991; 64: 457–462. 1173. Luscher U, Filgueira L, Juretic A, et al. The pattern of cytokine
1155. Lotze M, Jirik F, Kabouridis P, et al. B cell stimulating factor-2/ gene expression in freshly excised human metastatic melanoma
interleukin 6 is a co-stimulant for human thymocytes and T lym- suggests a state of reversible anergy of tumor infiltrating lym-
phocytes. J Exp Med 1988; 167: 1253–1258. phocytes. Int J Cancer 1994; 57: 612.
1156. Lotze M. Blanco FJ, Von Kempis J, et al. Cytokine regulation of 1174. Luzza F, Parello T, Monteleone G, et al. Up-regulation of IL-17
chondrocyte functions. J Rheumatol 1995; 22: 104. is associated with bioactive IL-8 expression in Helicobacter
1157. Lotze M. Role of IL-4 in the antitumor response. In: Spits H, ed. pylori-infected human gastric mucosa. J Immunol 2000; 165:
IL-4: Structure and Function. CRC Press, Boca Raton, Florida, 5332–5337.
1992; 111: 274–280. 1175. Lyman SD, Jacobsen SEW. c-kit ligand and Flt-3 ligand: stem/
1158. Lotze MT, Frana LW, Sharrow SO, et al. In vivo administration progenitor cell factors with overlapping yet distinct activities.
of purified human interleukin 2. I. Half-life and immunologic Blood 1998; 91: 1101–1134.
effects of the Jurkat cell line-derived interleukin 2. J Immunol 1176. Lyman SD, James L, Escobar S, et al. Identification of soluble
1985; 134: 157. and membrane-bound isoforms of murine Flt-3 ligand gener-
1159. Lotze MT, Hellerstedt B, Stolinski L, et al. The role of IL-2, ated by alternative splicing of mRNAs. Oncogene 1995; 10:
IL-12 and dendritic cells in cancer therapy. Cancer J Sci Am 149–157.
1997 3: 1: S109–114. 1177. Lyman SD, James L, Vanden Bos T, et al. Molecular cloning of
1160. Lotze MT, Matory YL, Ettinghausen SE, et al. In vivo adminis- a ligand for primitive hematopoietic cells. Cell 1993; 75:
tration of purified human interleukin 2. II. Half life, immuno- 1157–1167.
logic effects, and expansion of peripheral lymphoid cells in vivo 1178. Lyman SD, James L, Zappone J, et al. Characterization of the
with recombinant interleukin 2. J Immunol 1985; 135: protein encoded by the Flt3 (FLK2) receptor-like tyrosine
2865–2875. kinase gene. Oncogene 1993; 6: 815–822.
1161. Louahed J, Kermouni A, Van Snick J, Renauld J-C. IL-9 induces 1179. Lyman SD, Seaberg M, Hanna R, et al. Plasma/serum levels of
expression of granzymes and high affinity IgE receptor in Flt3 ligand are low in normal individuals and highly elevated in
murine T helper clones. J Immunol 1995; 154: 5061–5070. patients with Fanconi anemia and acquired aplastic anemia.
1162. Louahed J, Renauld J-C, Demoulin J-B, et al. Differential activ- Blood 1995; 86: 4091–4096.
ity of dexamethasone on IL-2, IL-4, or IL-9-induced prolifera- 1180. Lynch CN, Wang YC, Lund JK, et al. TWEAK induces angio-
tion of murine factor-dependent T cell lines. J Immunol 1996; genesis and proliferation of endothelial cells. J Biol Chem 1999;
156: 3704–3710. 274: 8455–8459.
1163. Lowenthal JW, MacDonald HR. Binding and internalization of 1181. Lynch DH and Miller RE. Induction of murine lymphokine acti-
interleukin 1 by T cells. Direct evidence for high and low affinity vated killer cells by recombinant IL-7. J Immunol 1990; 145: 198.
classes of interleukin-1 receptors. J Exp Med 1986; 164: 1060. 1182. Lynch DH and Miller RE. Interleukin 7 promotes Long term in
1164. Lu C, Kerbel RS. Interleukin 6: undergoes transition from para- vitro growth and antitumor cytotoxic T lymphocytes with immu-
crine growth inhibitor to autocrine growth stimulator during human notherapeutic efficacy in vivo. J Exp Med 1994; 179: 31–42.
melanoma progression. J Cell Biol 1993; 120: 1281–1288. 1183. Lynch DH, Andreasen A, Maraskovsky E, et al. Flt-3 ligand
1165. Lu C, Vickers MF, Kerbel RS. Interleukin 6: a fibroblast derived induces tumor regression and antitumor immune responses in
growth inhibitor of human melanoma cells from early but not vivo. Nat Med 1997; 3: 625–631.
advanced stages of tumor progression. Proc Nat Acad Sci USA 1184. Lynch DH. The role of FasL and TNF in the homeostatic regula-
1992; 89: 9215–9219. tion of immune response. Adv Exp Med Biol 1996; 406: 135.
1166. Lu G, Janjic BM, Janjic J, et al. Innate direct anticancer effector 1185. Ma A, Datta M, Margosian E, Chen J, Horak I. T cells but not
function of human immature dendritic cells. II. Role of TNF, B-cells are required for bowel inflammation in Interleukin-2-
lymphotoxin-α1β2, Fas ligand and TNF-relalted apoptosis-in- deficient mice. J Exp Med 1995; 182: 1567.
ducing ligand. J Immunol 2002; 168: 1831–1839. 1186. Ma H-L, Liang S, Li J, et al. IL-22 is required for Th17 cell-
1167. Luan J, Shattuck-Brandt R, Hagnegahdar H, et al. Mechanism mediated pathology in a mouse model of psoriasis-like skin
and biological significance of constitutive expression of MGSA/ inflammation. J Clin Invest 2008; 118: 597–607.
GRO chemokines in malignant melanoma tumor progression. 1187. Ma HL, Whitters MJ, Konz R, et al. IL-21 activates both innate
J Leukocyte Biol 1997; 62: 588. and adaptive immunity to generate potent antitumor responses
246 Cytokines

that require perforin but are independent of IFN-γ. J Immunol 1207. Maleckar JR, Fridell CS, Lewko WM, et al. Tumor derived acti-
2003; 171: 608–615. vated cells: cell culture conditions and characterization. In:
1188. Macatonia SE, Doherty TM, Knight SC, et al. Differential effect Stevenson HC, ed. Adoptive cellular immunotherapy of cancer.
of IL-10 on dendritic cell induced T cell proliferation and IFN-γ Marcel Dekker, New York, 1989; 159–173.
production. J Immunol 1993; 150: 3755. 1208. Maleckar JR, Fridell CS, Sferruzza A, et al. Activation and
1189. Macatonia SE, Hosken NA, Litton M, et al. Dendritic cells pro- Expansion of tumor derived activated cells (TDAC). J Nat
duce IL-12 and direct the development of Th1 cells form naive Cancer Inst 1989; 1: 1655–1660.
CD4+ T cells. J Immunol 1995; 154: 5071–5079. 1209. Malik N, Haugen HS, Mondrell B, Shoyab M, Clegg CH.
1190. MacGlashan D Jr, White JM, Huang S, et al. Secretion of IL-4 Developmental abnormalities in mice transgenic for bovine
from human basophils. J Immunol 1994; 152: 3006. oncostatin M. Mol Cell Biol 1995; 15: 2349.
1191. Maciaszek JW, Parada NA, et al. IL-16 represses HIV-1 pro- 1210. Malik N, Kallestad JC, Gunderson NL, et al. Molecular cloning,
moter activity. J Immunol 1997; 158: 5–8. sequence analysis, and functional expression of a novel growth
1192. Mackarehtschian K, Hardin JD, Moore KA, et al. Targeted dis- regulator, oncostatin M. Mol Cell Biol 1989, 9: 2847.
ruption of the flk2/flt3 gene leads to deficiencies in primitive 1211. Maling HM, Webster ME. Williams WA, Saul W, Anderson W.
hematopoietic progenitors. Immunity 1995; 3: 147–161. Inflammation induced by histamine, serotonin, bradykinin and
1193. Mackey F, Loetscher H, Steuber D, Gher G, Lesslauer W. Tumor compound 48/80 in the rat: antagonists and mechanism of
necrosis factor-α (TNF-α)-induced cell adhesion to endothelial action. J Pharmacol Exp Ther 1973; 191: 300.
cells is under dominant control of one TNF receptor type, 1212. Maliszewski CR, Sato TA, Vanden Bos T, et al. Cytokine
TNF-R55. J Exp Med 1993; 177: 1277. Receptors and B cell functions. I. Recombinant soluble recep-
1194. Mackey F, Majeau GR, Hochman PS, Browning JL. tors specifically inhibit IL-4 and IL-4 induced B cell activities in
Lymphotoxin β receptor triggering induces activation of the vitro. J Immunol 1990; 144: 3028–3033.
nuclear factor κB transcription factor in some cell types. J Biol 1213. Mallett S, Fossum S, Barclay AN. Characterization of the
Chem 1996; 271: 24934–24938. MRC OX40 antigen of activated CD4 positive T lymphocytes:
1195. Mackey MF, Gunn JR, Ting PP, et al. Dendritic cells require a molecule related to nerve growth factor receptor. EMBO J
maturation via CD40 to generate protective anticancer immu- 1990; 9: 1063.
nity. J Immunol 1998; 161: 2094. 1214. Manabe A, Cousan-Smith E, Kumagai M, et al. Interleukin-4
1196. Mackey MF, Gunn JR, Ting PP, et al. Protective immunity induces programmed cell death (apoptosis) in cases of high-risk
induced by tumor vaccines requires interaction between CD40 acute lymphoblastic leukemia. Blood 1994; 83: 1731–1737.
and its ligand CD154. Cancer Res 1994; 57: 2569. 1215. Manabe N, Kawaguchi H, Chicuda H, et al. Connection between
1197. Mackiewicz A, Schooltink H, Heinrich PC, Rose-John S. B-lymphocyte and osteoclast differentiation pathways. J
Complex of soluble human IL-6-receptor/IL-6 up regulates Immunol 2001; 167: 2625–2653.
expression of acute phase proteins. J Immunol 1992; 149: 2021. 1216. Mandrup-Poulsen T, Bendtzen K, Dinarello CA, Nerup J.
1198. MacNeil IA, Suda T, Moore KW, et al. IL-10, a novel growth Human tumor necrosis factor potentiates human interleukin
factor for mature and immature T cells. J Immunol 1990; 1-mediated rat pancreatic β-cell cytotoxicity. J Immunol 1987;
145: 4167. 139: 4077.
1199. Maeker HT, Hansen G, Walter DM, DeKruyff RH, Levy S, 1217. Manfield L, Jang D, Murrell AC. Nitric oxide enhances cyclooxy-
Umetsu DT. Vaccination with allergen-IL-18 fusion DNA pro- genase activity in articular cartilage. Inflamm Res 1996; 45: 254.
tects against and reverses established airway hyperreactivity in 1218. Mangan PR, Harrington LE, O’Quinn DH, et al. Transforming
a murine asthma model. J Immunol 2001; 166: 959–965. growth factor-β induces development of the TH17 lineage.
1200. Magazin M, Guillemot JC, Vita N, Ferra P. Interleukin13 is Nature 2006; 41: 231–234.
a monocyte chemoattractant. Eur Cytokine Network 1994; 1219. Manganini M, Maier JA. Transforming growth factor β2 inhibi-
5: 397. tion of hepatocyte growth factor-induced endothelial prolifera-
1201. Mahasreshti PJ, Kataram M, Wu H. Ovarian cancer targeted tion and migration. Oncogene 2000, 19: 124.
adenoviral-mediated mda-7/IL-24 gene therapy. Gynecol Oncol 1220. Manolagas SC, Bellido T, Jilka PL. New insights into the cel-
2006; 100: 521–532. lular, biochemical and molecular basis of postmenopausal and
1202. Maier R, Ganu V, Lotz M. Interleukin 11, an inducible cytokine senile osteoporosis: roles of IL-6 and pg130. Int J Immunopharm
in human articular chondrocytes and synoviocytes, stimulates 1996; 17: 109.
the production of tissue inhibitor of metalloproteinases. J Biol 1221. Marakowsky E, Daro E, Roux E, et al. In vivo generation of
Chem 1993; 268: 21527. human dendritic cell subsets by FLT3 ligand. Blood 2000;
1203. Maini R, Clair EW, Breedveld F, et al. Infliximab (chimeric 96: 878.
anti-tumour necrosis factor monoclonal antibody) versus pla- 1222. Maraskovsky E, Brasel K, Teepe M, et al. Dramatic increase in
cebo in rheumatoid arthritis patients receiving concomitant the numbers of functionally mature dendritic cells in FLT3
methotrexate: a randomized phase III trial. ATTRACT study ligand-treated mice: multiple dendritic cell populations identi-
group. Lancet 1999; 354: 1932–1939. fied. J Exp Med 1996; 184: 1953.
1204. Maione TE, Gray GS, Petro J, et al. Inhibition of angiogenesis 1223. Maraskovsky E, O’Reilly LA, Teepe M, et al. Bcl-2 can rescue
by recombinant human platelet factor-4 and related peptides. T lymphocyte development in interleukin-7 receptor-deficient
Science 1990; 247: 77. mice but not in mutant rag-1−/−mice. Cell 1997; 89: 1011.
1205. Maki K, Sunaga S, Ikuta K. The V-J recombination of T cell 1224. Maraskovsky E, Teepe M, Morrissey PJ, et al. Impaired survival
receptor-γ genes is blocked in interleukin-7 receptor deficient and proliferation in IL-7 receptor deficient peripheral blood T
mice. J Exp Med 1996; 184: 2423. cells. J Immunol 1996; 157: 5315.
1206. Maldonado-Lopez R, De Smedt T, et al. CD8α+ and 1225. March CJ, Mosely B, Larsen A, et al. Cloning, sequence and
CD8α−subclasses of dendritic cells direct the development of expression of two distinct human interleukin-1 complementary
distinct T helper cells in vivo. J Exp Med 1999; 189: 587. cDNA’s. Nature 1985; 315: 641–647.
Walter M. Lewko and Robert K. Oldham 247

1226. Margolin K, Aronson FR, Sznol M, et al. Phase II studies of 1245. Matsue H, Bergstresser PR, Takashima A. Keratinocyte derived
recombinant human IL-4 in advanced renal cancer and malig- IL-7 serves as a growth factor for dendritic epidermal T cells in
nant melanoma. J Immunother 1994; 15: 147–153. mice. J Immunol 1993; 151: 6012–6019.
1227. Mariner JM, Lantz V, Waldmann TA, Azimi N. Human T cell 1246. Matsue H, Bergstresser PR, Takashima A. Reciprocal cytokine
lymphotrophic virus type I Tax activates IL-15Rα gene expres- mediated cellular interactions in mouse epidermis: promotion of
sion through NF-κB site. J Immunol 2001; 166: 2602–2609. γδ T cell growth by IL-7 and TNFα and inhibition of keratino-
1228. Marshall DJ, Rudnick KA, McCarthy SG, et al. Interleukin-18 cyte growth by γ interferon. J Invest Dermatol 1993b; 101: 543.
enhances Th1 immunity and tumor protection of a DNA vac- 1247. Matsuguchi T, Musikacharoroen T, Ogawara T, Yoshikai Y.
cine. Vaccine 2006; 24: 244–253. Gene expressions of Toll-like receptor 2, but not Toll-like recep-
1229. Marshall E, Chakir J, Laviolette M, et al. IL-11 expression is tor 4. is induced by LPS and inflammatory cytokines in mouse
increased in severe asthma: association with epithelial cells and macrophages. J Immunol 2000; 165: 5767–5772.
eosinophils. J Allergy Clin Immunol 2000; 105: 232. 1248. Matsui Y, Zsebo KM, Hogan LM. Embryonic expression of
1230. Marshall JS, and Bienenstock J. The role of mast cells in inflam- hematopoietic growth factor encoded by Sl locus and the ligand
matory reactions of the airways, skin and intestine. Curr Opin for c kit. Nature 1990; 343: 667–669.
Immunol 1990; 6: 853. 1249. Matsumoto M, Mariathyasan S, Nahm HM, et al. Role of lym-
1231. Marsters SA, Ayres TM, Skubatch M, et al. Herpesvirus entry photoxin and the type 1 TNF receptor in the formation of germi-
mediator, a member of the tumor necrosis factor receptor nal centers. Science 1996; 271: 1289.
(TNFR) family, interacts with members of the TNFR-associated 1250. Matsushima K, Oppenheimer JJ. Interleukin 8 and MCAF:
factor family and activates the transcription factors NF-κB and Novel inflammatory cytokines inducible by IL-1 and TNF∞.
AP-1. J Biol Chem 1997; 272: 14029. Cytokine 1989; 1: 2–13.
1232. Marsters SA, Sheridan JP, Pitti RM, et al. A novel receptor for 1251. Matthews W, Jordan CT, Weigand GW, Pardoll D, Lemischka
Apo2L/TRAIL contains a truncated death domain. Current Biol IR. A receptor tyrosine kinase specific to hematopoietic stem
1997; 7: 1003. and progenitor cell-enriched populations. Cell 1991; 65:
1233. Martinet O, Divino CM, Zang Y, et al. T cell activation with 1143–1152.
systemic agonistic antibody versus 4-1BB ligand gene delivery 1252. Mattner F, Fisher S, Gluckes S, et al. The interleukin-12 subunit
combined with interleukin-12 eradicate liver metastases of p40 specifically inhibits effects of the interleukin-12 heterodi-
breast cancer. Gene Ther 2002; 9: 786–792. mer. Eur J Immunol 1993; 23: 2203–2208.
1234. Marz P, Cheng J-C, Gadient RA, et al. Sympathetic neurons can 1253. Mauer D, Holter W, Majdic O, Fisher GF, Knapp W. CD27
produce and respond to interleukin-6. Proc Natl Acad Sci USA expression by a distinct subpopulation of human B lympho-
1998; 95: 3251. cytes. Eur J Immunol 1999; 20: 2679.
1235. Mashikian MV, Ryan TC, Seman A, et al. Reciprocal desensiti- 1254. Mauri DN, Ebner R, Montgomery RI, et al. LIGHT, a new
zation of CCR5 and CD4 is mediated by IL-16 and macrophage- member of the TNF superfamily, and lymphotoxin α are ligands
inflammatory protein-1β, respectively. J Immunol 1999; 163: for herpesvirus entry mediator. Immunity 1998; 8: 21.
3123–3130. 1255. Mavalia C, Scaletti C, Romagnani P, et al. Type 2 helper T cell
1236. Mashikian MV, Tarpy RE, Saukkonen JJ, et al. Identification of predominance and high CD30 expression in systemic sclerosis.
IL-16 as the lymphocyte chemotactic activity in the bronchoal- Am J Pathol 1997; 151: 1751.
veolar lavage of fluid of histamine challenged asthmatic 1256. Maxwell JR, Campbell JD, Kim KH, Velli HT. CD40 activation
patients. J Allergy Clin Immunol 1998; 101: 786–792. boosts T cell immunity in vivo by enhancing T cell clonal
1237. Mastorakos G, Chrousos GP and Weber JS. Recombinant inter- expansion and delaying peripheral T cell deletion. J Immunol
leukin-6 activates the hypothalamic-pituitary-adrenal axis in 1999; 162: 2024–2034.
humans. J Clin Endocrinol Metab 1993; 77: 1690–1694. 1257. May LT, Helfgott DC, Sehgal PB. Anti-β-interferon antibodies
1238. Mateo RB, Reichner JS, and Albina JE. Interleukin-6 activity in inhibit the increased expression of HLA-B7 mRNA in tumor
wounds. Am J Physiol 1994; 226 (Suppl): R1840. necrosis factor-treated human fibroblasts: structural studies of
1239. Mathy NL, Bannert N, Norley SG, Kurth R. CD4 is not required the β2 interferon involved. Proc Natl Acad Sci USA 1986; 83:
for functional activity of IL-16. J Immunology 2000; 164: 8957–8961.
4429–4432. 1258. Mayer P, Giessler K, Ward M, Metcalf D. Recombinant human
1240. Mathy NL, Scheuer W, Lanzendoerfer M, et al. IL-16 stimulates leukemia inhibitory factor induces acute phase proteins and
the expression and production of proinflammatory cytokines by raises blood platelet counts in nonhuman primates. Blood 1993;
human monocytes. Immunology 2000; 100: 63–69. 81: 3226.
1241. Matikainen S, Siren J, Tissari J, et al. Tumor necrosis factor α 1259. Mayordoma JI, Zorina T, Storkus WJ, et al. Bone marrow
enhances influenza A virus-induced expression of antiviral derived dendritic cells pulsed with synthetic tumor peptides
cytokines by activating RIG-I gene expression. J Virol 2006; elicit protective and therapeutic antitumor immunity. Mature
80: 3515–3522. Medicine 1995; 1: 1297–1302.
1242. Matsuda M, Motoji T, Oshimi K, Mizoguchi H. Effects of inter- 1260. Mazodier K, Marin V, Novick D. Severe imbalance of IL-18/
leukin 7 on proliferation of hematopoietic malignant cells. Exp IL-18BP in patients with secondary hemophagic syndrome.
Hematol 1990; 18: 965–967. Blood 2005; 106: 3483–3489.
1243. Matsuda M, Salazar F, Peterson M, et al. Interleukin 10 pretreat- 1261. McBride WH, Thacker JD, Comora S, et al. Genetic modifica-
ment protects target cells from tumor and allo-specific cytotoxic tion of a murine fibrosarcoma to produce Interleukin 7 stimu-
T cells and downregulates HLA class 1 expression. J Exp Med lates host cell infiltration and tumor immunity. Cancer Res
1994; 180: 2371. 1992; 52: 3931–3937.
1244. Matsuda R, Takahashi T, Nakamura S, et al. Expression of the 1262. McCaffery PG, Perrino BA, Soderling TR, Rao A. NF-ATp, a
c-kit protein in human solid tumors and in corresponding fetal T-lymphocyte DNA-binding protein that is a target for calcineurin
and adult normal tissues. Am J Pathol 1993; 142: 339–349. and immunosuppressive drugs. J Biol Chem 1993; 269: 3747.
248 Cytokines

1263. McCaffrey PG, Luo C, Kerppola TK, et al. Isolation of the 1282. Medzhitov R, Preston-Hurlburt P, Janeway Jr. CA. A human
cyclosporin sensitive transcription factor NF-ATp. Science homologue of the Drosophila Toll protein signals activation of
1993; 262: 750. adaptive immunity. Nature 1996; 388: 394.
1264. McCain RW, Dessypris EN, Christman JW. Granulocyte/mac- 1283. Mehrota PT, Grant AJG and Siegel JP. Synergistic effects of
rophage colony stimulating factor stimulates human polymor- IL-7 and IL-12 on Human T cell activation. J Immunol 1995;
phonuclear leukocytes to produce interleukin 8 in vitro. J Respir 154: 5093–5102.
Cell Mol Biol 1993; 8: 28. 1284. Mehrota PT, Wu D, Crim JA, et al. Effects of IL-12 on the gen-
1265. McClanahan T, Culpepper J, Campbell D, et al. Biochemical eration of cytotoxic activity in human CD8+ T lymphocytes.
and genetic characterization of multiple splice variants of the J Immunol 1993; 151: 2444–2452.
Flt3 ligand. Blood 1996; 88: 3371–3382. 1285. Mehta DS, Wurster AL, Whitters MJ, et al. IL-21 induces the
1266. McDermott DF, Terhu EG, et al. A two part phase I trial of high apoptosis of resting and activated primary B cells. J Immunol
dose interleukin 2 in combination with soluble (Chinese 2003; 170: 4111–4118.
Hamster Ovary) interleukin 1 receptor. Clin Cancer Res 1998; 1286. Meierhoff G, Dehmel U, Gruss H-J, et al. Expression of Flt3
4: 1203–1213. receptor and Flt3-ligand in human leukemia-lymphoma cell
1267. McGeachy MJ, Bak-Jensen KS, Chen Y, et al. TGF-β and IL-6 lines. Leukemia 1995; 9: 1368–1372.
drive the production of IL-17 and IL-10 by T cells and restrain 1287. Melani C, Mattia GF, Silvani A, et al. Interleukin-6 expression
TH−17 cell-mediated pathology. Nat Immunol 2007; 8: 1390–1397. in human neutrophil and eosinophil peripheral blood granulo-
1268. McInnes IB, Al-Mugales J, Field M, et al. The role of interleu- cytes. Blood 1993; 81: 2744–2749.
kin 15 in T cell migration and activation in rheumatoid arthritis. 1288. Melchjorsen J, Siren J, Julkunen I, et al. Induction of cytokine
Nat Med 1996; 2: 175. expression by herpes simplex virus in human monocyte-derived
1269. McKenna HJ, de Vries P, Brasel K, Lyman SD, Williams DE. macrophages and dendritic cells is dependent on virus replica-
Effect of flt 3 ligand on the ex vivo expansion of human CD34+ tion and is counteracted by ICP27 targeting NF-κB and IRF-3.
hematopoietic progenitor cells. Blood 1995; 86: 3413. J Gen Virol 2006; 87: 1099–1108.
1270. McKenna HJ, Morrisey PJ. Flt3 ligand plus IL-7 supports the 1289. Melero I, Johnson JV, Shuford WW, et al. NK 1.1 cells express 4–1
expansion of murine thymic B bell progenitors that can mature BB (CDw137) and are required for tumor immunity elicited by anti-
intrathymically. J Immunol 1998; 160: 4801–4809. 4–1 BB monoclonal antibodies. Cell Immunol 1998; 190: 167.
1271. McKenna HJ, Smith OF, Brasel K, et al. Effects of Flt3 ligand 1290. Melero I, Shuford LL, Newby SA, et al. Monoclonal antibodies
on acute myeloid and lymphocytic leukemic blast cells from against 4-1BB T cell activation molecule eradicate established
children. Exp Hematol 1996; 24: 378–385. tumors. Nat Med 1997; 3: 682.
1272. McKenna HJ, Stocking KL, Miller RE, et al. Mice lacking FLT3 1291. Mellin GW, Katsenstein M. The saga of thalidomide. N Eng
ligand have deficient hematopoiesis affecting hematopoietic J Med 1962; 264: 1184–1193.
progenitor cells, dendritic cells and natural killer cells. Blood 1292. Mennechet FJ, Uze G. Interferon-λ-treated dendritic cells spe-
2000; 95: 3489–3497. cifically induce proliferation of FOXP3-expressing dendritic
1273. McKenna SD, Chen F, Lai L, Goldschneider I. Identification cells. Blood 2006; 107: 4417–4423.
of an IL-7 associated pre-pro-B cell growth-stimulating 1293. Merberg DM, Wolf SF, Clark SC. Sequence similarity between
factor (PPBSF). I. Production of the non-IL-7 component NKSF and the IL-6/G-CSF family. Immunol Today 1992; 14: 77.
by bone marrow stromal cells from IL-7 gene deleted mice. 1294. Merino R, Grillot AM, Simonian PL, et al. Modulation of anti-
J Immunology 1998; 160: 2272–2279. IgM-induced B cell apoptosis by Bcl-XL and CD40 in WEHI-
1274. McKenzie ANJ, Culpepper JA, De Waal Malefyt R, et al. 231 cells. J Immunol 1995; 155: 3830.
Interleukin 13, a novel T cell derived cytokine that regulates 1295. Metcalf D, Nicola NA, Gearing DP. The effects of injected leu-
human monocyte and B cell function. Proc Nat Acad Sci USA kemia inhibitory factor on hematopoietic and other tissues in
1993; 90: 3735–3739. mice. Blood 1990; 77: 2150.
1275. McKenzie ANJ, Li X, Largaespada DA, et al. Structural com- 1296. Metcalf D. Control of granulocytes and macrophages: molecu-
parison and chromosomal localization of the human and mouse lar, cellular and clinical aspects. Science 1991; 254: 529.
IL-13 genes. J Immunol 1993; 150: 5436–5444. 1297. Metcalf D. Leukemia inhibitory factor (LIF). Int J Cloning
1276. McKenzie GJ, Emson CL, Bell SE, et al. Impaired development 1991; 9: 95.
of Th2 cells in IL-13-deficient mice. Immunity 1998; 8: 423–432. 1298. Meunch MO, Roncarolo MG, Menon S, et al. Flk-2/Flt-3 ligand
1277. McKenzie RC, Paglia D, Kondo S, Sauder DN. A novel endog- regulates the growth of early myeloid progenitors isolated from
enous mediator of cutaneous inflammation: leukemia inhibitory human fetal liver. Blood 1995; 85: 963–972.
factor. Acto Dermato-Venereol 1996; 76: 111. 1299. Mian BM, Dinney CP, Bermejo CE, et al. Fully human anti-
1278. McKinnon M, Page K, Uings IJ, et al. An interleukin 5 mutant interleukin-8 antibody inhibits tumor growth in orthotopic
distinguishes between two functional responses in human bladder cancer xenografts via down-regulation of matrix metal-
eosinophils. J Exp Med 1997; 186: 121–129. loproteases and nuclear factor-κB. Clin Cancer Res 2003; 9:
1279. McLane MP, Haczku A, van der Rijn M, et al. Interleukin-9 pro- 3167–3175.
motes allergen-induced eosinophilic inflammation and airway 1300. Micallef MJ, Ohtsuki T, Kohno K, et al. Interferon-γ-inducing
hyperresponsiveness in transgenic mice. Am J Respir Cell Mol factor enhances T helper 1 cytokine production by stimulated
Biol 1998; 19: 713. human T cells: synergism with interleukin-12 for interferon-γ
1280. Medico E, Gentile A, Lo Celso C, et al. Osteopontin is an auto- production. Eur J Immunol 1996; 26: 1647–1651.
crine mediator of hepatocyte growth factor-induced invasive 1301. Micallef MJ, Tanimoto T, Kohno K, Ikeda M, Kurimoto M.
growth. Cancer Res 2001; 61: 5861–5868. Interleukin-18 induces the sequential activation of natural killer
1281. Medvedev AE, Espevik T, Ganges G, Sundan A. Distinct roles of cells and cytotoxic T lymphocytes to protect syngeneic mice
the two tumor necrosis factor (TNF) receptors in modulating TNF from transplantation with Meth A sarcoma. Cancer Res 1997b;
and lymphotoxin α effects. J Biol Chem 1996; 271: 9778–9784. 57: 4557–4563.
Walter M. Lewko and Robert K. Oldham 249

1302. Micallef MJ, Yoshida K, Kawai S, et al. In vivo antitumor 1321. Miura S, Ohtani K, Numata M, et al. Molecular cloning and
effects of murine interferon-γ-inducing factor IL-18 in mice characterization of a novel glycoprotein gp34, that is specifi-
bearing syngeneic Meth A sarcoma malignant ascites. Cancer cally induced by the human T cell leukemia virus I transactiva-
Immunol Immunother 1997a; 43: 361–367. tor p40tax. Mol Cell Biol 1991; 11: 1313.
1303. Micham JL, Parnet P, Bonnert TP, et al. T1/ST2 signaling estab- 1322. Miyahira Y, Akiba H, Katae M, et al. A potent adjuvant effect of
lishes it as a member of the expanding interleukin-1 receptor ligand to receptor Activator of NF-κB gene for inducing anti-
family. J Biol Chem 1996; 271: 5777. gen-specific CD8+ T cell response by DNA and viral vector
1304. Michie HR, Manogue KR, Spriggs DR, et al. Detection of cir- vaccination. J Immunol 2003; 171: 6344–6348.
culating tumor necrosis factor after endotoxin administration. N 1323. Miyajima A, Mui AL, Ogorouchi T, Sakamaki K. Receptors for
Engl J Med 1988; 318: 1481. granulocyte-macrophage colony stimulating factor, interleu-
1305. Mier JW, Gallo RC. Isolation and purification of human T-cell kin-3 and interleukin-5. Blood 1993; 82: 1960.
growth factor. Proc Natl Acad Sci (USA) 1980; 77: 6134. 1324. Miyazawa K, Mori A, Yamamoto K, Okudaira H. Constitutive
1306. Migliorati G, Pagilacci C, Moraca R, et al. Interleukins modu- transcription of the human interleukin-6 gene by rheumatoid
late glucocorticoid induced thymocyte apoptosis. Int J Clin Lab synoviocytes: spontaneous activation of NF-κB and CBF1. Am
Res 1992; 21: 300–303. J Pathol 1998; 152: 793.
1307. Miki S, Iwano Y, Miki Y, et al. Interleukin 6 functions as an in 1325. Miyomoto M, Prause O, Sjostrand M. Endogenous IL-17
vitro autocrine growth factor in renal cell carcinoma. FEBS Lett as a mediator of neutrophil recruitment caused by endo-
1989; 250: 607–610. toxin exposure in mouse airways. J Immunol 2003; 170:
1308. Miles SA, Martinez-Mara O, et al. Oncostatin M is a potent 4665–4672.
mitogen for AIDS-Kaposi’s sarcoma derived cells. Science 1326. Mizel SB, Dawyer JM, Krane SM, et al. Stimulation of rheuma-
1992; 255: 1432. toid synovial cell collagenase and prostaglandin production by
1309. Miller AM, Lundberg K, Ozenci V, et al. CD4+ CD25 high T partially purified lymphocyte-activating factor (interleukin-1).
cells are enriched in the tumor and peripheral blood of prostate Proc Natl Acad Sci (USA) 1981; 78: 2474.
cancer patients. J Immunol 2006; 177: 7398–7405. 1327. Mizel SB. The interleukins. FASEB J 1989; 3: 2379–2388.
1310. Miller AM, Xu D, Asquith DL, et al. IL-33 reduces the develop- 1328. Mizuno A, Amizuka N, Irie K, et al. Severe osteoporosis in mice
ment of atherosclerosis. J Exp Med 2008; 205: 339–346. lacking osteoclastogenesis inhibitory factor/osteoprotegerin.
1311. Miller LL, Korn EL, Stevens DS, et al. Abrogation of the hema- Biochem Biophys Res Comm 1998; 247: 610.
tological and biological activities of the interleukin-1/granulo- 1329. Mochizuki DY, Eisenman JR, Conlon PJ, et al. Interleukin 1
cyte macrophage colony stimulating factor fusion protein regulates hematopoietic activity, a role previously ascribed to
PIXY321 by neutralizing antibodies in cancer patients receiving hemopoietin 1. Proc Natl Acad Sci (USA) 1987; 84: 5267–5271.
high dose carboplatin. Blood 1999; 93: 3250–3258. 1330. Modrowski D, Godet D, Marie PJ. Involvement of interleukin-1
1312. Miller RE, Jones J, Le T, et al. 4-1BB-specific monoclonal anti- and tumor necrosis factor α as endogenous growth factors in
body promotes the generation of tumor-specific immune human osteoblastic cells. Cytokine 1995; 7: 720–726.
responses by direct activation of CD8 T cells in a CD40- 1331. Modur V, Li Y, Zimmerman GA, Prescott SM, McIntyre TM.
dependent manner. J Immunol 2002; 160: 1792–1800. Retrograde inflammatory signaling from neutrophils to endothe-
1313. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 lial cells by soluble interleukin-6 receptor α. J Clin Invest 1997;
Macrophages and the Th1/Th2 paradigm. J Immunol 2000; 164: 100: 2752–2756.
6166–6173. 1332. Moeller J, Hultner L, Schmitt E, Dormer P. Partial purification
1314. Miner KT and Croft M. Generation, persistence and modulation of mast cell growth-enhancing activity and its separation from
of Th0 effector cells: a role of autocrine IL-4 and IFN-γ. J IL-3 and IL-4. J Immunol 1989; 142: 3447.
Immunol 1998; 160: 5280–5287. 1333. Mogi S, Sakurai J, Kohsaka T, et al. Tumor rejection by gene
1315. Minty A, Chalon P, Derocq J-M, et al. Interleukin-13: a novel transfer of 4-1BB ligand into a CD80+ murine squamous cell
human cytokine regulating inflammatory and immune response. carcinoma and the requirements of costimulatory molecules on
Nature 1993; 362: 248–250. tumor and host cells. Immunology 2000; 101: 541–547.
1316. Mire-Sluis A, Page LA, Wadhwa M, Thorpe R. Evidence for a 1334. Mohamadzadeh M, Takashima A, Dougherty I, et al. Ultraviolet
signaling role for the α chains of granulocyte-macrophage col- B radiation up regulates the expression of IL-15 in human skin.
ony stimulating factor (GM-CSF), interleukin-3 (IL-3), and J Immunol 1995; 155: 4492–4496.
IL-5 receptors: divergent signaling pathways between GM-CSF/ 1335. Moissec P, Yu CL, Ziff M. Lymphocyte chemotactic activity
IL-3 and IL-5. Blood 1995; 86: 2679. of interleukin 1. J Immunol 1984; 133: 2007.
1317. Mitsuyama K, Andus T, Caesar I, Roth M, Scholmerich J. 1336. Molin D, Fisher M, Xiang Z, et al. Mast cells express functional
Soluble interleukin-6 receptors in inflammatory bowel disease: CD30 ligand and are the predominant CD30L-positive cells in
relation to circulating interleukin-6. Gut 1995; 36: 45. Hodgkin’s disease. Br J Haematol 2001; 114: 616–623.
1318. Mittleman A, Puccio C, Gafney E, et al. A phase I pharmacokinetic 1337. Moller A, Lippert U, Lessmann D, et al. Human mast cells
study of recombinant human tumor necrosis factor administered by a produce IL-8. J Immunol 1993; 151: 3261.
5-day continuous infusion. Invest New Drugs 1992; 10: 183–190. 1338. Moller P, Moller H, Sun Y, Dorbic T, et al. Increased non-major
1319. Mittler RS, Bailey TS, Klussman K, et al. Anti-4-1BB monoclo- histocompatibility complex-restricted lytic activity in mela-
nal antibodies abrogate T cell-dependent humoral immune noma patients vaccinated with cytokine gene-transfected autol-
responses in vivo through the induction of helper T cell anergy. ogous tumor cells. Cancer Gene Ther 2000; 7: 976–984.
J Exp Med 1999; 190: 1535–1540. 1339. Moller P, Sun Y, Dorbic T, et al. Vaccination with IL-7 gene-
1320. Miu AL, Wakao H, O’Farrell AM, Harada N, Miyajima A. modified autologous melanoma cells can enhance the anti-
Interleukin-3, granulocyte-macrophage colony stimulating factor melanoma lytic activity in peripheral blood of patients with
and interleukin-5 transduce signals through two STAT5 good clinical performance status: a clinical phase I study. Br J
homologs. EMBO J 1995; 14: 1166–1175. Cancer 1998; 77: 1907–1916.
250 Cytokines

1340. Molmenti EP, Ziambaras T, Perlmutter DH. Evidence for an 1359. Morisaki T, Yuzuki D, Lin R, et al. Interleukin-4 receptor
acute phase response in human intestinal epithelial cells. J Biol expression and growth inhibition of gastric carcinoma cells by
Chem 1993; 268: 12116–14124. interleukin-4. Cancer Res 1992; 52: 6059–6065.
1341. Mongkolsapaya J, Cowper AE, Screaton GR, et al. Lymphocyte 1360. Morisset S, Patry C, Lora M, deBrum-Fernandes AJ. Regulation
inhibitor of TRAIL (TNF-Related Apoptosis-Inducing Ligand): of cyclooxygenase-2 expression in bovine chondrocytes in cul-
a new receptor protecting lymphocytes from the death ligand ture by interleukin-1, tumor necrosis factor-α, glucocorticoids
TRAIL. J Immunol 1998; 160: 3–6. and 17β-estradiol. J Rheumatol 1998; 25: 1146.
1342. Monroe JG, Haldar S, Prystowsky MB, Lammie P. Lymphokine 1361. Moroz A, Eppolito C, Li Q, et al. Interleukin-21 enhances
regulation of inflammatory process: IL-4 stimulates fibroblast and sustains CD8+ T cell responses to achieve durable tumor
proliferation. Clin Immunol Immunopathol 1988; 49: 292. immunity: comparative evaluation of IL-2, IL-15 and IL-21.
1343. Montaner L, Doyle AG, Collin M, et al. Interleukin-13 inhibits J Immunol 2004; 173: 900–909.
human immunodeficiency virus type 1 production in primary 1362. Morris A, Vetto JT, Ramstad T, et al. Induction of anti-mam-
blood-derived human macrophages in vitro. J Exp Med 1993; mary cancer immunity by engaging the OX-40 receptor in vivo.
178: 743. Breast Cancer Res Treatment 2001; 67: 71–80.
1344. Montgomery RI, Warner MS, Lum BJ, Spear PG. Herpes sim- 1363. Morris JC, Janik JE, White JD, et al. Preclinical and phase I
plex virus-1 entry into cells mediated by a novel member of the clinical trial of blockade of IL-15 using Mikbeta1 monoclonal
tumor necrosis factor superfamily. Cell 1996; 87: 427–436. antibody in T cell large granular lymphocyte leukemia. Proc
1345. Moon C, Yoo J-Y, Matarazzo V, et al. Leukemia inhibitory Natl Acad Sci USA 2006; 103: 401–406.
factor inhibits neuronal terminal differentiation through STAT3 1364. Morrisey PJ, McKenna HJ, Widmer MB, et al. Steel factor
activation. Proc Natl Acad Sci USA 2003; 99: 9015–9020. (c-kit ligand) stimulates the in vitro growth of immature CD3−/
1346. Moore BB, Arenberg DA, Addison CL, Keane MP, Strieter RM. CD4−/CD8−thymocytes: synergy with IL-7. Cell Immunol
Tumor angiogenesis is regulated by CXC chemokines. J Lab 1994; 22: 1037.
Clin Med 1998; 132: 97–103. 1365. Morrissey PJ, Goodwin RG, Nordan RP, et al. Recombinant
1347. Moore KW, O’Garra A, de Waal Malefyt R, Viera P, Mosmann interleukin 7, pre-B cell growth factor has costimulatory activ-
TR. Interleukin-10. Ann Rev Immunol 1993; 11: 165. ity on purified mature T cells. J Exp Med 1989; 169: 707–716.
1348. Moore KW, Viera P, Fiorentino DF, et al. Homology of cytokine 1366. Morse MA, Nair S, Fernandez-Casal M, et al. Preoperative
synthesis inhibitor factor (IL-10) to the Epstein Barr virus gene mobilization of circulating dendritic cells by Flt3 ligand admin-
BCRFI. Science 1990; 248: 1230–1234. istration to patients with metastatic colon cancer. J Clin Oncol
1349. Moore MAS, Warren DJ. Synergy of interleukin-1 and granulocyte 2000; 18: 3879–3882.
colony-stimulating factor: in vivo stimulation of stem-cell recovery 1367. Mortarini R, Borri A, Tragni G, et al. Peripheral burst of tumor-
and hematopoietic regeneration following 5-fluorouracil treatment specific cytotoxic T lymphocytes and infiltration of metastatic
of mice. Proc Natl Acad Sci (USA) 1987; 84: 7134–7138. lesions by memory CD8+ T cells in melanoma patients receiv-
1350. Moore TA, Zlotnik A. Differential effects of Flt2/flt3 ligand and ing interleukin-12. Cancer Res 2000; 60: 3559–3568.
stem cell factor on murine thymic progenitor cells. J Immunol 1368. Mortarini R, Scarito A, Nonaka D, et al. Constitutive expression
1997; 158: 4187. and costimulatory function of LIGHT/TNFSF14 on human
1351. Morel F, Delwail V, Brizard A, et al. Effects of sCD23 on prolif- melanoma cells and melanoma-derived microvesicles. Cancer
eration of leukemic cells from a patient with chronic myeloge- Res 2005; 65: 3428–3436.
nous leukemia during blast crisis. Amer J Hematology 1993; 44: 1369. Mortier E, Bernard J, Plet A, Jacques Y. Natural, proteolytic
60–62. release of a soluble form of human IL-15 receptor α-chain that
1352. Morel Y, Schiano de Colella J-M, et al. Reciprocal expression of behaves as a specific, high affinity IL-15 antagonist. J Immunol
the TNF family receptor herpes virus entry mediator and its 2004; 173: 1681–1688.
ligand LIGHT on activated T cells: LIGHT down regulates its 1370. Moseley TA, Haudenschild DR, Rose L, Reddi AH.
own receptor. J Immunol 2000; 165: 4397–4404. Interleukin-17 family and IL-17 receptors. Cytokine Growth
1353. Morgan H, Tumber A, Hill PA. Breast cancer cells induce osteo- Factor Rev 2003; 14: 155–174.
clast formation by stimulating host IL-11 production and down- 1371. Moser B, Schumacher C, von Tscharner V, et al. Neutrophil
regulating granulocyte/macrophage colony stimulating factor. activating peptide 2 and melanoma growth stimulatory activity
Int Journal Cancer 2004; 109: 653–660. interact with neutrophil activating peptide 1/interleukin 8 recep-
1354. Mori A, Suko M, Kaminuma O, Inoue S, et al. IL-15 promotes tors on human neutrophils. J Biol Chem 1991; 266: 10667.
cytokine production of human T helper cells. J Immunol 1996; 1372. Mosley B, Beckman MP, March CJ, et al. The murine interleu-
156: 2400–2405. kin-4 receptor: molecular cloning and characterization of
1355. Mori L, Iselin S, De Libero G, Lesslauer W. Attenuation of secreted and membrane bound forms. Cell 1989; 59: 335–348.
collagen-induced arthritis in 55-kDa TNF receptor type I 1373. Mosley B, De Imus C, Friend D, et al. Dual oncostatin M (OSM)
(TNFR1)-IgG1-treated and TNFR1-deficient mice. J Immunol receptors: cloning and characterization of an alternative signal-
1996; 157: 3178. ing subunit conferring OSM specific receptor activation. J Biol
1356. Moriera AL, Sampaio EP, Zmuidzinas A, et al. Thalidomide Chem 1996; 271: 32635–32643.
exerts its inhibitory action on tumor necrosis factor α by enhanc- 1374. Mosmann T, Bond M, Coffman R, et al. T cell and mast cell
ing mRNA degradation. J Exp Med 1993; 177: 1675–1680. lines respond to B cell stimulatory factor 1. Proc Nat Acad Sci
1357. Morinaga Y, Suzuki H, Takatsuki F, et al. Contribution of IL-6 USA 1986; 83: 1857.
to the antiproliferative effect of IL-1 and tumor necrosis factor 1375. Mosmann T, Cherwinski H, Bond M, et al. Two types of murine
on tumor cell lines. J Immunol 1989; 143: 3538. helper T cell clones. 1. Definition to profiles of lymphokine
1358. Morisaki T, Uchiyama A, Yuzuki D, et al. Interleukin 4 regu- activities and secreted proteins. J Immunol 1986; 136: 2348.
lates G1 cell cycle progression in gastric carcinoma cells. 1376. Mosmann TR. Cytokine secretion patterns and cross regulation
Cancer Res 1994; 54: 1113–1118. of T cell subsets. Immunol Res 1991; 10: 183–188.
Walter M. Lewko and Robert K. Oldham 251

1377. Moss ML, Jin S-LC, Milla ME, et al. Cloning of a disintegrin 1396. Murata K, Ishii N, Takano H, et al. Impairment of antigen-
metalloproteinase that processed precursor tumor necrosis presenting cell function in mice lacking expression of OX40
factor-α. Nature 1997; 385: 733. ligand. J Exp Med 2000; 191: 365.
1378. Motro B, Itin A, Sachs L, et al. Pattern of interleukin 6 gene 1397. Murata S, Ladle BH, Kim PS, et al. OX40 costimulation syner-
expression in vivo suggests a role for this cytokine in angio- gizes with GM-CSF whole cell vaccination to overcome estab-
genesis. Proc Natl Acad Sci USA 1990; 87: 3092–3096. lished CD8+ T cell tolerance to an endogenous tumor antigen.
1379. Motzer RJ, Rakhit A, Thomson JA, et al. Randomized multi- J Immunol 2006; 176: 974–983.
center phase II trial of subcutaneous recombinant human inter- 1398. Murata Y, Takaki S, Migita M, et al. Molecular cloning and
leukin-12 versus interferon-α 2a for patients with advanced renal expression of the human interleukin-5 gene. J Exp Med 1992;
cell carcinoma. J Interferon Cytokine Res 2001; 21: 257–263. 175: 341.
1380. Movat HZ, Burrowes CE, Cybulsky MI, Dinarello CA. Acute 1399. Murphy CA, Langrish CL, Chen Y, et al. Divergent roles of
inflammation and a Shwartzman-like reaction induced by inter- IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med
leukin-1 and tumor necrosis factor. Synergistic action of the 2003; 198: 1951–1957.
cytokines in the induction of inflammation and microvascular 1400. Murphy P, Tiffany H. Cloning of complimentary DNA encoding
injury. Am J Pathol 1987; 129: 463. a functional human IL-8 receptor. Science 1991; 253: 1280.
1381. Mrozek E, Anderson P, Caliguri MA. Role of interleukin 15 in 1401. Murphy PA, Chesney PA, Wood WB. Further purification of
the development of human CD56+ natural killer cells from rabbit leukocyte pyrogen. J Lab Clin Med 1974; 83: 310.
CD34+ hematopoietic progenitor cells. Blood 1996; 87: 2632. 1402. Murphy WJ, Back TC, Conlon KC, et al. Antitumor effects of
1382. Muchamuel T, Menon S, Piscane P, Howard MC, Cockayne DA. interleukin 7 and adoptive immunotherapy on human colon car-
IL-13 protects mice from lipopolysaccharide-induced lethal cinoma xenografts. J Clinical Invest 1993; 92: 1918–1924.
endotoxemia: correlation with down-modulation of TNF-α, 1403. Murrell GA, Jang CD, Williams RJ. Nitric oxide activates
IFN-γ and IL-12 production. J Immunol 1997; 158: 2898–2903. metalloprotease in articular cartilage. Biochem Biophys Res
1383. Muegge K, Vial MP, Durum SK. Interleukin 7: a cofactor for Commun 1995; 206: 15.
V(D)J rearrangement of the T cell receptor β gene. Science 1404. Musashi M, Clark SC, Sudo T, Urdal DL, Ogawa M. Synergistic
1993; 261: 93–95. interactions between interleukin-11 and interleukin-4 in support
1384. Mukaida N, Harada A, Yasumoto K, et al. Properties of proin- of proliferation of primitive hematopoietic progenitors in mice.
flammatory cell type specific leukocyte chemotactic cytokines Blood 991; 78: 1448–1451.
interleukin 8 (IL-8) and monocyte chemotactic and activating 1405. Musashi M, Yang Y-C, Paul SR, et al. Direct and synergistic
factor (MCAF). Microbiol Immunol 1992; 36: 773–789. effects of interleukin 11 on murine hematopoiesis in culture.
1385. Mule JJ, Custer MC, Travis WD, Rosenberg SA. Cellular mech- Proc Nat Acad Sci USA 1991; 88: 765–769.
anisms of the antitumor activity of recombinant IL-6 in mice. 1406. Musgrave BL, Phu T, Butler JJ, Makrigiannis AP, Hoskin DW.
J Immunol 1992; 148: 2622. Murine TRAIL (TNF-related apoptosis-inducing ligand) expres-
1386. Mule JJ, Shu S, Rosenberg SA. The antitumor efficacy of lym- sion induced by T cell activation is blocked by rampamycin,
phokine-activated killer cells and recombinant interleukin 2 in cyclosporin A, and inhibitors of phosphatidylinositol 3-kinase,
vivo. J Immunol 1985; 135: 646. protein kinase C, and protein tyrosine kinases: evidence for
1387. Mule JJ, Shu S, Schwarz SL, et al. Adoptive immunotherapy of TRAIL induction via the T cell receptor signaling pathway. Exp
established pulmonary metastases with LAK cells and recombi- Cell Res 1999; 252: 96.
nant interleukin-2. Science 1984; 225: 1487. 1407. Muta H, Boise LH, Fang L, Podack ER. CD30 signals integrate
1388. Mule JJ, Smith CA, Rosenberg SA. Interleukin-4 (B cell stimu- expression of cytotoxic effector molecules, lymphocyte traf-
latory factor) can mediate the induction of lymphokine activated ficking signals, and signals for proliferation and apoptosis.
killer cell activity directed against fresh tumor cells. J Exp Med J Immunol 2000; 165: 5105–5111.
1987; 166: 792–797. 1408. Muzio M, Re F, Sironi M, et al. Interleukin-13 induces the pro-
1389. Mullberg J, Durie FH, Otten-Evans C, et al. A metalloproteinase duction of interleukin-1 receptor antagonist (IL-1ra) and the
inhibitor blocks shedding of the IL-6 receptor and the p60 TNF expression of the mRNA for the intracellular (keratinocyte)
receptor. J Immunol 1995; 155: 5198–5205. form of the IL1-ra in human myelomonocytic cells. Blood 1994;
1390. Mullberg J, Geib T, Jostock T, et al. IL-6 receptor independent 83: 1738.
stimulation of human gp130 by viral IL-6. J Immunol 2000; 1409. Nagasawa T, Kikutani H, Kishimoto T. Molecular cloning and
164: 4672–4677. structure of a pre-B cell growth-stimulating factor. Proc Natl
1391. Mullberg J, Schooltink H, Stoyan T, et al. The soluble interleu- Acad Sci USA 1994; 91: 2305.
kin-6 receptor is generated by shedding. Eur J Immunol 1993; 1410. Nagata S and Suda T. Fas and Fas ligand: lpr and gld mutations.
23: 473. Immunol Today 1995; 16: 39.
1392. Mummery RS, Rider CC. Characterization of the heparin binding 1411. Nagata S, Ise T, Onda M, et al. Cell membrane specific epitopes
properties of IL-6. J Immunol 2000; 165: 5671–5679. on CD30: Potentially superior targets for immunotherapy. Proc
1393. Munder M, Mallo M, Eichmann K, Modolell M. Murine mac- Natl Acad Sci USA 2005; 102: 7946–7951.
rophages secrete interferon-γ upon combined stimulation with 1412. Nagayama H, Sato K, Kawasaki H, et al. IL-12 responsiveness
interleukin-12 and IL-18: a novel pathway of autocrine and expression of IL-12 receptor in human peripheral blood
macrophage activation. J Exp Med 1998; 187: 2103. monocyte-derived dendritic cells. J Immunol 2000; 165: 59–66.
1394. Muraguchi A, Hirano T, Tang B, et al. The essential role of B 1413. Naka T, Narazaki M, Hirata M, et al. Structure and function of a
cell stimulatory factor 2 (BSF-2/IL-6) for the terminal differen- new STAT-induced STAT inhibitor. Nature 1997; 387: 924.
tiation of B cells. J Exp Med 1988; 167: 332. 1414. Nakae S, Saijo S, Horai R, et al. IL-17 production from acti-
1395. Muranski P, Boni A, Antony PA, et al. Tumor specific Th17- vated T cells is required for the spontaneous development of
polarized cells eradicate large established melanoma. Blood destructive arthritis in mice deficient in IL-1 receptor antago-
2008: 112: 362–373. nist. Proc Natl Acad Sci USA 2003; 100: 5986–5990.
252 Cytokines

1415. Nakajima A, Kodama T, Morimoto S, et al. Antitumor effect of mononuclear cells in multiple sclerosis. J Neuroimmunol 1996,
CD40 ligand elicitation on focal and systemic antitumor 64: 63.
responses by IL-12 and B7. J Immunol 1998; 1661: 1901. 1435. Nawroth P, Handley D, Matsueda G, et al. Tumor necrosis fac-
1416. Nakajima H, Gleich GJ, Kita H. Constitutive production of IL-4 tor/cachechtin-induced intravascular fibrin formation in meth A
and IL-10 and stimulated production of IL-8 by normal periph- fibrosarcomas. J Exp Med 1988; 168: 637–647.
eral blood eosinophils. J Immunol 1996; 156: 4859–4866. 1436. Ndhlovu LC, Ishii N, Murata K, Sato T, Sugamura K. Critical
1417. Nakamura K, Okamura H, Wada M, et al. Endotoxin-induced involvement of the OX40 ligand signals in the T cell priming
serum factor that stimulates γ interferon production. Infect events during experimental autoimmune encephalomyelitis.
Immunol 1989; 57: 590. J Immunol 2001; 167: 2991–2999.
1418. Nakamura S, Kashimoto S, Kajikawa F, Nakata K. Combination 1437. Neipel F, Albrecht JC, Ensser A, et al. Human herpesvirus 8
effect on recombinant interleukin 1α with antitumor drugs on encodes a homolog of interleukin-6. J Virol 1997; 71: 839.
syngeneic tumors in mice. Cancer Res 1991; 51: 215–221. 1438. Neis MM, Peters B, Dreuw A, et al. Enhanced expression levels
1419. Nakanishi K, Malek TR, Smith KA, et al. Both interleukin 2 and of IL-31 correlate with IL-4 and IL-13 in atopic and allergic
a second T cell derived factor in EL-4 supernatant have activity contact dermatitis. J Allergy Clin Immunol 2006; 118: 930–937.
as differentiation factors in IgM synthesis. J Exp Med 1984; 1439. Nelms K, Huang H, Ryan J, Keegan A, Paul WE. Interleukin-4
160: 1605. receptor signaling mechanisms and their biological significance.
1420. Nakano H, Kishida T, Asada H, et al. Interleukin-21 triggers Adv Exp Med Biol 1998; 452: 37–43.
both cellular and humoral immune responses leading to thera- 1440. Nelson DR, Lauwers GY, Lau JY, Davis GL. Interleukin 10
peutic antitumor effects against head and neck squamous cell treatment reduces fibrosis in patients with chronic hepatitis C: a
carcinoma. J Gene Med 2006; 8: 90–99. pilot trial of interferon non-responders. Gastroenterology 2000;
1421. Naldini A, Leali D, Pucci A, et al. IL-1β mediates the proangio- 118: 655–660.
genic activity of osteopontin-activated human monocytes. 1441. Neta R, Douches SD, Oppenheim JJ. Interleukin-1 is a radio-
J Immunol 2006; 177: 4267–4270. protector. J Immunol 1986; 136: 2483–2488.
1422. Namen AE, Schierer AE, March CJ, et al. B cell precursor-growth 1442. Neta R, Oppenheim JJ, Douces SD. Interdependence of the
promoting activity and characterization of a growth factor active radioprotective effects of human recombinant interleukin 1α,
on lymphocyte precursors. J Exp Med 1988; 167: 988–1002. tumor necrosis factor α, granulocyte-monocyte colony-stimu-
1423. Namen AE, Lupton S, Hjerrild K, et al. Stimulation of B cell lating factor and murine recombinant granulocyte-macrophage
progenitors by cloned murine IL-7. Nature 1988; 333: 571. colony stimulating factor. J Immunol 1988; 140: 108.
1424. Namikawa R, Muench MO, DeVries, JE, Roncarolo M-G. The 1443. Netea MG, Azam T, Ferwerda G, et al. IL-32 synergizes with
flt2/flt3 ligand synergizes with interleukin-7 in promoting nucleotide oligomerization domain (NOD) 1 and NOD2 ligands
stromal-cell-independent expansion and differentiation of for IL-1β and IL-6 production through a caspase 1-dependent
human fetal pro-B cells in vitro. Blood 1996; 87: 1881. mechanism. Proc Natl Acad Sci USA 2005; 102: 16309–16314.
1425. Nastala CL, Edington HD, McKinney TG, et al. Recombinant 1444. Netea MG, Lewis EC, Azam T, et al. Interleukin-32 induces the
IL-12 administration induced tumor regression in association differentiation of monocytes into macrophage-like cells. Proc
with IFN-production. J Immunol 1994; 153: 1697–1706. Natl Acad Sci USA 2008; 102: 16309–16314.
1426. Natali PG, Nicotra MR, Sures I, et al. Breast cancer is associ- 1445. Neuman B, Luz A, Pfeffer K, Holzamnn B. Defective Peyer’s
ated with loss of c-kit oncogene product. Int J Cancer 1992; 52: patch organogenesis in mice lacking the 55-kD receptor for
713–717. tumor necrosis factor. J Exp Med 1996; 184: 259.
1427. Natali PG, Nicotra MR, Sures I, et al. Expression of c kit receptor 1446. Neumann D, Kollewe C, Martin MU, Boraschi D. The mem-
in normal and transformed human nonlymphoid tissues. Cancer brane form of the type II IL-1 receptor accounts for inhibitory
Res 1992; 52: 6139–6143. function. J Immunol 2000; 165: 3350–3357.
1428. Natali PG, Nicotra MR, Winkler AB, et al. Progression of human 1447. Nicholls SE, Heyworth CM, Dexter TM, et al. IL-4 promotes
cutaneous melanoma is associated with loss of expression of macrophage development by rapidly stimulating lineage restric-
c-kit protooncogene receptor. Int J Cancer 1992; 52: 197–201. tion of bipotent granulocyte macrophage colony forming cells.
1429. Nathan CF. Neutrophil activation on biological surfaces. Massive J Immunol 1995; 155: 845–853.
secretion of hydrogen peroxide in response to products of mac- 1448. Nicholls SE, Winter S, Mottram R, Miyan JA, Whetton AD.
rophages and lymphocytes. J Clin Invest 1987; 80: 1550. Flt3 ligand can promote survival and macrophage development
1430. Nau GJ, Guilfoile P, Chupp GL, et al. A chemoattractant without proliferation in myeloid progenitor cells. Exp Hematol
cytokine associated with granulomas in tuberculosis and silico- 1999; 27: 663–672.
sis. Proc Natl Acad Sci USA 1997; 6414–6419. 1449. Nichols J, Evans EP, Smith AG. Establishment of germ-cell-line
1431. Naume B, Espevik T, Sundan A. Gene expression and secretion competent embryonic stem cells using differentiation-inhibiting
of cytokines and cytokine receptors from highly purified CD56+ activity. Development 1990; 110: 1341–1348.
natural killer cells stimulated with interleukin 2 interleukin 7 1450. Nickolaides NC, Holroyd KJ, Ewart SL, et al. Interleukin 9: a can-
and interleukin 12. Eur J Immunol 1993b; 23: 1831–1838. didate gene for asthma. Proc Natl Acad Sci USA 1997; 94: 13175.
1432. Naume B, Gately M, Espevik T. A comparative study of IL-12 1451. Niedbala W, Wei XO, Cai B, et al. IL-35 is a novel cytokine
(cytotoxic lymphocyte maturation factor), IL-2, and IL-7 with therapeutic effects against collagen-induced arthritis
induced effects on immunomagnetically purified CD56+ NK through the expansion of regulatory T cells and suppression of
cells. J Immunol 1992; 148: 2429–2436. Th17 cells. Eur J Immunol 2007; 37: 3021–3029.
1433. Naume B, Gately MK, Desai BB, et al. Synergistic effects of 1452. Niehans GA, Brunner T, Frizelle SP, et al. Human lung carcino-
interleukin 4 and interleukin 12 on NK cell proliferation. mas express Fas ligand. Cancer Res 1997; 57: 1007.
Cytokine 1993; 5: 38–46. 1453. Nieland JD, Graus YF, Dortmans YE, et al. CD40 and CD70
1434. Navikas V, Matusevicius D, Soderstom M, et al. Increased inter- co-stimulate a potent in vivo antitumor response. J Immunother
leukin-6 mRNA expression in blood and cerebrospinal fluid 1998; 21: 225–236.
Walter M. Lewko and Robert K. Oldham 253

1454. Nikolaus S, Raedler A, Kuhbacker T, Sfikas N, Folsch UR, 1473. Obiri N, Hillman GG, Haas GP, et al. Expression of high affinity
Schreiber S. Mechanisms in failure of infliximab for Crohn’s interleukin-4 receptors on human renal cell carcinoma cells and
disease. Lancet 2000; 356: 1475–1479. inhibition of tumor growth in vitro by interleukin-4. J Clin
1455. Nilsen EM, Johansen FE, Jahnsen FL, et al. Cytokine profiles of Investigation 1993; 91: 88–93.
cultured microvascular endothelial cells from human intestine. 1474. Obiri NI and Puri RK. Characterization of interleukin-4 recep-
Gut 1998; 42: 635–642. tors expressed in human renal cell carcinoma cells. Oncol Res
1456. Nilsson SK, Johnston HM, Whitty GA, et al. Osteopontin, a key 1995; 6: 419.
component of the hematopoietic stem cell niche and regulator of 1475. Obiri NI, Hussain SR, Debinski W, Puri RK. Interleukin 13
primitive hematopoietic progenitor cells. Blood 2005; 106: inhibits growth of human renal carcinoma cells independently
1232–1239. of the p140 interleukin 4 receptor chain. Clin Cancer Res 1996;
1457. Nishimura H, Hirimatsu K, Kobayashi N, et al. IL-15 is a novel 2: 1743–1749.
growth factor for murine γδ T cells induced by Salmonella 1476. Ogasawara H, Takeda-Hirokawa N, Sekigawa I, et al. Inhibitory
infection. J Immunol 1996; 156: 663. effects of interleukin-16 on interleukin-2 production by CD4+ T
1458. Nishimura T, Uchiyama Y, Yagi H, Hashimoto Y. Administration cells. Immunology 1999; 96: 215–219.
of slowly released recombinant interleukin 2. J Immunol 1477. Ogasawara K, Hida S, Azimi N, et al. Requirement for IRF-1 in
Methods 1986; 91: 21. the microenvironment supporting the development of natural
1459. Noelle R, Krammer P, Ohara J, et al. Increased expression of Ia killer cells. Nature 1998; 391: 700.
antigens on resting B cells. A new role for B cell growth factor 1478. Ogata A and Anderson KC. Therapeutic strategies in the inhibi-
1. Proc Nat Acad Sci USA 1984; 81: 6149. tion of interleukin-6 mediated multiple myeloma-mediated cell
1460. Noguchi E, Shibasaki M, Arinami T, et al. Evidence for a linkage growth. Leuk Res 1996; 20: 303.
between asthma/atopy in childhood and chromosome 5q31-q33 in a 1479. Ogawa N, Itoh M, Gately Y. Abnormal production of B cell
Japanese population. Am J Respir Crit Care Med 1997; 156: 1390. growth factor in patients with systemic lupus erythematosis.
1461. Noguchi M, Nakamura Y, Russel SM, et al. Interleukin receptor Clin Exp Immunol 1992; 89: 26–38.
γ chain, a functional component of the interleukin 7 receptor. 1480. Oh J-W, Van Wagner N, Rose-John S, Benveniste EN. Role of
Science 1993; 262: 1877–1880. IL-6 and the soluble IL-6 receptor in inhibition of VCAM-1
1462. Nogueira AC, Neubert R, Helge H, Neubert D. Thalidomide and gene expression. J Immunol 1998; 161: 4992.
the immune system. 3. Simultaneous up- and down-regulation of 1481. Ohkobo C, Bigos D, Jain R. Interleukin-2 inducted leukocyte
different receptors on human white blood cells. Life Sci 1994; adhesion to the normal and tumor microvascular endothelium in
55: 77–92. vivo and its inhibition by dextran sulfate: implications for vas-
1463. Nonaka M, Nonaka R, Wooley K, et al. Distinct immunohis- cular leak syndrome. Cancer Res 1991; 512: 1561.
tochemical localization of IL-4 in human inflamed airway tissues. 1482. Ohlsson K, Bjork P, Bergenfeldt M, Hageman R, Thompson
IL-4 is localized to eosinophils in vivo and is released by periph- RC. Interleukin-1 receptor antagonist reduces mortality from
eral blood eosinophils. J Immunol 1995; 155: 3234–3244. endotoxin shock. Nature 1990; 348: 550.
1464. Nophar Y, Kemper O, Brakebusch C, et al. Soluble forms of 1483. Ohm JE, Shurin MR, Esche C, et al. Effect of vascular endothe-
tumor necrosis factor receptors (TNF-R’s). The cDNA for the lial growth factor on dendritic generation in vivo. J Immunol
type I TNF-R cloned using amino acid sequence data of its sol- 1999; 163: 3260–3268.
uble form, encodes both the cell surface and a soluble form of 1484. Ohno R, Yamaguchi Y, Tohge T, et al. A dose escalation and
the receptor. EMBO J 1990; 9: 3269–3278. pharmacology study of subcutaneously administered recombi-
1465. Novick D, Kim S-H, Fantuzzi G, et al. Interleukin 18 binding nant human interleukin-12 and its biological effects in Japanese
protein: a novel modulator of the Th1 cytokine response. patients with advanced malignancies. Clin Cancer Res 2000; 6:
Immunity 1999; 10: 127–136. 2661–2669.
1466. Numasaki M, Tagawa M, Iwata F, et al. IL-28 elicits antitumor 1485. Okada H, Pollack IF, Lieberman F, et al. Gene therapy of malig-
responses against murine fibrosarcoma. J Immunol 2007, 178: nant gliomas: a pilot study of vaccination with irradiated autolo-
5086–5098. gous glioma and dendritic cells admixed with IL-4 transduced
1467. Numasaki M, Watanabe M, Suzuki T, et al. IL-17 enhances the fibroblasts to elicit an immune response. Hum Gene Ther 2001;
net angiogenic activity and the in vivo growth of human non- 12: 575–595.
small cell lung cancer in SCID mice through promoting CXCR- 1486. Okahara H, Yagita H, Miyake K, Okumura K. Involvement of
2-dependent angiogenesis. J Immunol 2005; 175: 6177–6189. very late activation antigen 4 (VLA-4) and Vascular Cell
1468. O’Connell J, Bennett MW, O’Sullivan GC, Collins JK, Shannon Adhesion Molecule 1 (VCAM-1) in tumor necrosis factor α
F. Fas counterattack: a molecular mechanism of tumor privi- enhancement of experimental metastasis. Cancer Res 1994; 54:
lege. Mol Med 1997; 3: 294. 3233–3236.
1469. O’Connell J, O’Sullivan GC, Collins JK, Shannon F. Fas coun- 1487. Okamoto I, Kohno K, Tanimoto T, Ikegami H, Kurimoto M.
terattack: Fas mediated T cell killing by colon cancer cells Development of CD8+ effector T cells is differentially regu-
expressing Fas ligand. J Exp Med 1996; 184: 1075. lated by IL-18 and IL-12. J Immunol 1999; 162: 3202–3211.
1470. O’Garra A, Chang R, Hatings R, et al. Ly IB (B-1) cells are the main 1488. Okamura H, Tsutsui H, Kashiwamura S-I, Yashimoto T-,
source of B-cell derived IL-10. Eur J Immunol 1992; 22: 711. Nakanishi K. IL18: A novel cytokine that augments both innate
1471. O’Malley WE, Archinstein B, Shear MJ. Action of bacterial and acquired immunity. Adv Immunol 1998; 70: 281.
polysaccharide on tumors: II. Damage of sarcoma 37 by serum 1489. Okamura H, Tsutsui H, Komatsu T, et al. Cloning a new cytokine
of mice treated with Serratia marcescens polysaccharide and that induces IFN-γ production by T-cells. Nature 1995; 378: 88.
induced tolerance. Nutr Rev 1986; 46: 389–391. 1490. Okano F, Merad M, Furumoto K, et al. In vivo manipulation of
1472. O’Neal KD, Yu-Lee L. The proline rich motif (PRM): a novel dendritic cells overcomes tolerance to unmodified tumor-asso-
feature of the cytokine/hematopoietin receptor superfamily. ciated self-antigens and induces potent antitumor immunity.
Lymphokine Cytokine Res 1993; 12: 309–312. J Immunol 2005; 174: 2645–2652.
254 Cytokines

1491. Okayashiki K, Miyauchi J, Ohyashiki JH, et al. Interleukin 7 1508. Ortaldo JR, Mason AT, Gerard JP, et al. Effects of natural and
enhances colony growth and induces CD 20 antigen of a Ph+ recombinant IL-2 on regulation of IFNγ production and natural
acute lymphoblastic leukemia cell line. Leukemia 1993; 7: killer activity: lack of involvement of Tac antigen for these
1034–1040. immunoregulatory effects. J Immunol 1984; 133: 779.
1492. Okazaki H, Ito M, Sudo T, et al. IL-7 promotes thymocytes 1509. Osaki T, Peron J-M, Cai Q, Okamura H, et al. IFN-γ-inducing fac-
bearing α/β or γ/δ T cell receptors in vitro: Synergism with IL-2. tor/IL-18 Administration mediates IFN-γ-and IL-12-independent
J Immunol 1989; 143: 2917–2922. antitumor effects. J Immunol 1998; 160: 1742–1749.
1493. Oldham RK, Lewko W, Good R, et al. Growth of tumor derived 1510. Oshima S, Saeki Y, Mina T, et al. Interleukin 6 plays a key role
activated T-cells for the treatment of cancer. Cancer Biotherapy in the development of antigen-induced arthritis. Proc Natl Acad
1994; 9(3): 211–224. Sci USA 1998; 95: 8222.
1494. Oldham RK, Lewko WM, Good RW, Sharp E. Cancer biother- 1511. Oshima Y, Tanaka Y, Tozawa H, Takahashi Y, Maliszewski C,
apy with interferon, interleukin 2 and tumor derived activated Delepesse G. Expression and function of Ox 40 ligand on
cells (TDAC). In Vivo 1994; 8: 653–664. human dendritic cells. J Immunol 1997; 159: 3838–3848.
1495. Oldham RK, Thurman GB. Effects of cytokines on recovery 1512. Oshimi K, Oshimi Y, Akutsu M, et al. Cytotoxicity of interleu-
from radiation exposure. In: Rotman M, Rosenthal CJ, eds. kin 2-activated lymphocytes for leukemia and lymphoma cells.
Medical radiology – Diagnostic imaging and radiation oncol- Blood 1986; 68: 938.
ogy. Concomitant continuous infusion chemotherapy and radia- 1513. Ostensen ME, Thiele DL, and Lipsky PE. Tumor necrosis factor
tion. Springer-Verlag, New York, 1990. enhances cytolytic activity of human natural killer cells. J
1496. Olieman AF, Lienard D, Eggermont AM, et al. Hyperthermic Immunol 1987; 138: 4185.
isolated limb perfusion with tumor necrosis factor α, interferon 1514. Osterlund P, Veckman V, Siren J, et al. Gene expression and
γ, and melphalan for locally advanced nonmelanoma skin antiviral activity of α/β interferons and interleukin 29 in virus
tumors of the extremities: a multicenter study. Arch Surg 1999; infected human myeloid dendritic cells. J Virol 2005; 79:
134: 303–307. 9608–9617.
1497. Oniki S, Nagai H, Horikawa T, et al. Interleukin-23 and inter- 1515. Ouaaz F, Li M, Begg AA. A critical role for the RelA subunit of
leukin-27 exert quite different antitumor and vaccine effects on nuclear factor κB in regulation of multiple immune-response
poorly immunogenic melanoma. Cancer Res 2006; 66: genes and Fas-induced cell death. J Exp Med 1999; 189: 999.
6395–6404. 1516. Overwiik WW, de Visser KE, Tirion FH, et al. Immunologic and
1498. Onozaki K, Matsushima K, Aggarwal BB, Oppenheim JJ. antitumor effects of IL-23 as a cancer vaccine adjuvant. J
Human interleukin 1 is a cytocidal factor for several tumor cell Immunol 2006; 176: 5213–5222.
lines. J Immunol 1985; 135: 3962–3968. 1517. Owen-Schaub LB, Gutterman JU, Grimm EA. Synergy of tumor
1499. Opal SM, Fisher CJ Jr, Dhainaut JF, et al. Confirmatory inter- necrosis factor and interleukin 2 in the activation of human
leukin-1 receptor antagonist trial in severe sepsis: a phase III, cytotoxic lymphocytes: Effect of tumor necrosis factor and
randomized, double-blind, placebo controlled, multicenter trial. Interleukin 2 in the generation of human lymphokine activated
The interleukin-1 receptor antagonist sepsis investigator group. killer cell cytotoxicity. Cancer Res 1988; 48: 788–792.
Crit Care Med 1997; 25: 1115–1124. 1518. Owyang AM, Zaph C, Wilson EH, et al. Interleukin 25 regulates
1500. Oppenheim JJ, Kovacs EJ, Matsushima K, Durum SK. There is type 2 cytokine-dependent immunity and limits inflammation in
more than one interleukin-1. Immunol Today 1986; 7: 45–56. the gastrointestinal tract. J Exp Me 2006; 203: 843–849.
1501. Oppenheim JJ, Neta R, Tiberghien P, et al. Interleukin-1 1519. Owyang AM, Zaph C, Wilson EH, et al. Interleukin-25 regu-
enhances survival of lethally irradiated mice treated with allo- lates type 2 cytokine –dependent immunity and limits chronic
geneic bone marrow cells. Blood 1989; 74: 2257. inflammation in the gastrointestinal tract. J Exp Med 2006; 203:
1502. Oppmann B, Lesley R, Blom B, et al. Novel p19 protein 843–849.
engages IL-12p40 to form a cytokine, IL-23, with biological 1520. Ozaki K, Kikly K, Michalovich D, Young PR, Leonard WJ.
activities similar as well as distinct from IL-12. Immunity Cloning of a type 1 cytokine receptor most related to the IL-2
2000; 13: 715. receptor β chain. Proc Nat Acad Sci USA 2000; 97: 11439.
1503. Orazi A, Cooper RJ, Tong J, et al. Effects of recombinant human 1521. Ozaki K, Spolski R, Ettinger R, et al. Regulation of B cell dif-
inteleukin-11 (Neumega rhIL-11 growth factor) on megakaryo- ferentiation and plasma cell generation by IL-21, a novel inducer
cytopoiesis in human bone marrow. Exp Hematol 1966; 24: of Blimp-1 and Bcl-6. Immunol 2004; 173: 5361–5371.
1289–1297. 1522. Ozaki K, Spolski R, Feng CG, et al. A critical role for IL-21 in
1504. Orencole SF, Dinarello CA. Characterization of a subclone regulating immunoglobulin production. Science 2002; 298:
(D10S) of D10.G4.1 helper T-cell line, which proliferates to 1630.
attomolar concentrations of interleukin-1 in absence of mito- 1523. Page TH, Willcocks JL, Taylor-Fishwick DA, Foxwell BMJ.
gens. Cytokine 1989; 1: 14–22. Characterization of a novel high affinity human IL-7 receptor.
1505. Orengo AM, Cantoni C, Neglia F, Biassoni R, Ferrini S. Expression on T cells and association with IL-7 driven prolif-
Reciprocal expression of CD70 and of its receptor, CD27, in eration. J Immunol 1993; 151: 4753–4763.
human long term-activated T and natural killer (NK) cells: 1524. Paige CJ, Figarella EF, Cuttito M, et al. Natural cytotoxic cells
inverse regulation by cytokines and role in induction of cytotox- against solid tumors in mice: II. Some characteristics of the
icity. Clin Exp Immunol 1997; 107: 608–613. effector cells. J Immunol 1978; 121: 1827.
1506. Orlando S, Sironi M, Bianchi G, et al. Role of metalloprotease 1525. Pajkrt D, Camoglio L, Tiel-van Buul MCM, et al. Attenuation of
in the release of IL-1 type II decoy receptor. J Biol Chem 1997: pro-inflammatory response by recombinant human IL-10 in
272: 31764. human endotoxemia: effect of timing of recombinant human
1507. Ortaldo JR, Mason A, Overton R. Lymphokine-activated killer IL-10 administration. J Immunol 1997; 158: 3971–3977.
cells. Analysis of the progenitors and effectors. J Exp Med 1526. Pan G, French D, Mao W, et al. Forced expression of murine
1986; 164: 1193. IL-17 E induces growth retardation, jaundice, a Th2-biased
Walter M. Lewko and Robert K. Oldham 255

response, and multiorgan inflammation in mice. J Immunol cells provide protection against acute myelogenous leukemia in
2001; 167: 6559–6567. nontransplanted or syngeneic bone marrow-transplanted mice.
1527. Pan G, Ni J, Yu G, Wei YF, Dixit VM. TRUNDD, a new mem- Blood 2001; 97: 1474–1482.
ber of the TRAIL receptor family that antagonizes TRAIL sig- 1547. Peace DJ, Cheever MA. Toxicity and therapeutic efficacy of
naling. FEBS Lett 1998; 424: 41. high dose interleukin-2. J Exp Med 1989; 169: 161.
1528. Pan G, O’Roarke K, Chinnaian AM, et al. The receptor for the 1548. Pechon JJ, Morrisey PJ, Grabstein KH, et al. Early lymphocyte
cytotoxic ligand TRAIL. Science 1997; 276: 111. expansion is severly impaired in interleukin-7 receptor-deficient
1529. Pan PY, Zhang Y, Weber K, et al. OX40 ligation enhances pri- mice. J Exp Med 1994; 180: 1955–1960.
mary and memory cytotoxic T lymphocyte responses in an 1549. Peck R, Brockhaus M, Frey JR. Cell surface tumor necrosis fac-
immunotherapy for hepatic colon metastases. Mol Ther 2002; 6: tor (TNF) accounts for monocyte- and lymphocyte-mediated kill-
528–536. ing of TNF-resistant target cells. Cell Immunol 1989; 122: 1.
1530. Pan Y, Lloyd C, Zhou H, et al. Neurotactin, a membrane-an- 1550. Peluso I, Fantini MC, Fina D, et al. IL-21 counteracts the regu-
chored chemokine up regulated in brain inflammation. Nature latory T cell-mediated suppression of human CD4+ T lympho-
1997; 387: 611. cytes. J Immunol 2007; 178: 732–739.
1531. Pandrau D, Frances V, Martinez Valdez H, et al. Characterization 1551. Pene J, Gauchat JF, Lecart S, et al. IL-21 is a switch factor for
of a t(1;19) pre-B acute lymphoblastic leukemia (ALL) cell line, the production of IgG1 and IgG3 by human B cells. J Immunol
which proliferates in response to IL-7. Leukemia 1993; 7: 2004; 172: 5142–5147.
635–642. 1552. Penn J, Rousset F, Briere F, et al. IgE production by normal
1532. Parada NA, Center DM, Kornfeld H, et al. Synergistic activa- human lymphocytes is induced by interleukin-4 and suppressed
tion of CD4+ T cells by IL-16 and IL-2. J Immunol 1998; 160: by interferon-γ and α and prostaglandin E2. Proc Natl Acad Sci
2115–2120. USA 1988; 85: 880.
1533. Parham C, Chirica M, Timans J, et al. A receptor for the het- 1553. Pennica D, Nedwin GE, Hayflick JS, et al. Human tumor necro-
erodimeric IL-23 is composed of IL-12Rβ1 and a novel cytokine sis factor: precursor structure, expression and homology to lym-
receptor subunit, IL23 R. J Immunol 2002; 168: 5699–56708. photoxin. Nature 1984; 312: 724.
1534. Parihar R, Nadella P, Jensen R, et al. Phase I trial of Herceptin 1554. Peritt D, Robertson S, Gri G, Showe L, Aste-Amezaga M,
and Interleukin-12 in patients with HER2-overexpressing Trinchieri G. Differentiation of human NK cells into NK1 and
malignancies. Proc ASCO 2001; 20: 1031. NK2 subsets. J Immunol 1998; 161: 5821–5824.
1535. Park CC, Morel JCM, Amin MA, et al. Evidence of IL-18 as a 1555. Peron J-M, Esche C, Subbotin VM, et al. FLT3 ligand adminis-
novel angiogenesis mediator. J Immunol 2001; 167: tration inhibits liver metastases: role of NK cells. J Immunol
1644–1653. 1998; 161: 6164–6170.
1536. Park H, Li Z, Yang XO, et al. A distinct lineage of CD4 T cells 1556. Perper SJ, Browning B, Burkly LC, et al. TWEAK is a novel
regulates tissue inflammation by producing interleukin 17.Nat arthritogenic mediator. J Immunol 2006; 177: 2610–2620.
Immunol 2005; 6: 1133–1141. 1557. Perregaux D, Gabel CA. Interleukin-1β maturation and release in
1537. Park LS, Friend D, Sassenfeld HM, Urdal DL. Characterization response to ATP and nigericin. J Biol Chem 1994; 269: 15195.
of human B cell stimulatory factor 1 receptor. J Exp Med 1987; 1558. Perrigoue JG, Li J, Zaph C, et al. IL-31–IL-31R interactions
166: 476–488. negatively regulate type 2 inflammation in the lung. J Exp Med
1538. Parnet P, Garka KE, Bonnert TP, Dower SK, Sims JE. IL-1 Rrp 2007; 204: 481–487.
is a novel receptor like molecule similar to the type I interleukin 1559. Pertyl U, Luster AD, Varki NM, et al. IFN-γ-inducible protein is
1 receptor and its homologues T1/ST2 and IL-R AcP. J Biol essential for the generation of a protective tumor-specific CD8
Chem 1996; 271: 3967. T cell response induced by single-chain IL-12 gene therapy. J
1539. Parrish-Novak J, Dillon SR, Nelson A, et al. Interleukin-21 and Immunol 2001; 166: 6944–6951.
its receptor are involved in NK expansion and regulation of 1560. Perussia B, Ramoni C, Anegon I, et al. Preferential proliferation
lymphocyte function. Nature 2000; 408: 57. of natural killer cells among peripheral blood mononuclear cells
1540. Parrish-Novak J, Xu W, Brender T, et al. Interleukins 19, 20, cocultured with B lymphoblastoid lines. Nat Immun Cell
and 24 signal through two distinct receptor complexes. Growth Regul 1987; 6: 171.
Differences in receptor–ligand interactions mediate unique bio- 1561. Pesce J, Kaviratne M, Ramalingam TR, et al. The IL-21 recep-
logical functions. J Biol Chem 2002; 277: 47517–47523. tor augments Th2 effector function and alternative macrophage
1541. Parronchi P, Maggi E, Romagnani S. Redirecting Th2 responses activation. J Clin Invest 2006; 116: 2044–2055.
in allergy. Curr Top Microbiol Immunol 1999; 238: 27–56. 1562. Peters M, Muller A, Rose-John S. Interleukin-6 and soluble
1542. Paterson DJ, Jeffries WA, Green JR, et al. Antigens of activated interleukin-6 receptor: direct stimulation of gp130 and
rat T thymocytes including a molecule of 50,000 Mr detected on hematopoiesis. Blood 1998; 92: 3495.
CD4 positive T blasts. Mol Immunol 1987; 24: 1281. 1563. Peters M, Schirmacher P, Goldschmidtt J, et al. Extramedullary
1543. Paul SR, Bennett F, Calvetti JA, et al. Molecular cloning of a expansion of hematopoietic progenitor cells in interleukin (IL)-
cDNA encoding interleukin 11, a novel stromal cell derived 6-sIL-6R double transgenic mice. J Exp Med 1997; 185: 755.
lymphopoietic and hematopoietic cytokine. Proc Nat Acad Sci 1564. Petzer AL, Hogge DE, Landsdorp PM, Reid DS, Eaves CJ. Self-
USA 1990; 87: 7512. renewal of primitive human hematopoietic cells (long-term-
1544. Paul WE, Ohara J. B-cell stimulatory factor-1/interleukin 4. culture initiating cells) in vitro and their expansion in defined
Annu Rev Immunol 1987; 5: 429. medium. Proc Natl Acad Sci USA 1996; 93: 1470–1474.
1545. Paulukat J, Bosmann M, Nold M, et al. Expression and release 1565. Peveri P, Walz A, Dewald B, Baggiolini M. A neutrophil activat-
of IL-18 binding protein in response to interferon-γ.J Immunol ing factor produced by human mononuclear phagocytes. J Exp
2001; 167: 7038–7043. Med 1988; 167: 1547.
1546. Pawlowska AB, Hashino S, McKenna H, Taylor PA, Blazar BR. 1566. Pflanz S, Kurth I, Grotzinger J, Heinrich PC, Muller-Newen G.
In vitro tumor pulsed or in vivo Flt3 ligand-generated dendritic Two different epitopes of the signal transducer gp130 sequen-
256 Cytokines

tially cooperate in IL-6-induced receptor activation. J Immunol 1586. Poltorak A, He X, Smirnova I, et al. Defective LPS signaling in
2000, 165: 7042–7049. CeH/HeJ and C57BL/10ScCr mice: mutations in the Tlr gene.
1567. Phillips JH, Lanier LL. Dissection of the lymphokine-activated Science 1998; 282: 2085.
killer phenomenon. Relative contribution of peripheral blood 1587. Porteu F, Nathan C. Shedding of tumor necrosis factor receptors
natural killer cells and T lymphocytes to cytolysis. J Exp Med by activated human neutrophils. J Exp Med 1990; 172: 599.
1986; 164: 814. 1588. Portielje JE, Kruit WH, Schuler M, et al. Phase I study of
1568. Piacibello W, Sanavio F, Garetto L, et al. Extensive amplifica- subcutaneously administered recombinant human interleukin
tion and self-renewal of human primitive hematopoietic stem 12 in patients with advanced renal cell cancer. Clin Cancer Res
cells form cord blood. Blood 1997; 89: 2644–2653. 1999; 5: 3983–3989.
1569. Picarella D, Kratz A, Li C-B, Ruddle NH, Flavel RA. Insulinitis 1589. Powell DJ Jr, Dudley ME, Robbins PF, Rosenberg SA.
in transgenic mice expressing TNF-β (lymphotoxin) in the pan- Transition of late stage effector T cells to CD27+ CD28+ tumor-
creas. Proc Natl Acad Sci USA 1992; 89: 10036. reactive effector T cells in humans after adoptive cell transfer
1570. Pinto A, Aldinucci D, Gloghini A, et al. Human eosinophils therapy. Blood 2005; 105: 241–250.
express functional CD30 ligand and stimulate proliferation of a 1590. Powell DJ, Filipe-Silva A, Merino MJ, et al. Administration of
Hodgkin’s disease cell line. Blood 1996; 88: 3299–3305. a CD25-directed immunotoxin, LMB-2, to patients with met-
1571. Pippig SD, Pena-Rossi C, Long J, et al. Robust B cell immunity astatic melanoma induces a selective partial reduction in regula-
but impaired T cell proliferation in the absence of CD134 tory T cells in vivo. J Immunol 2007; 179: 4919–4928.
(Ox40). J Immunol 1999; 163: 6520. 1591. Prasad KV, Ao Z, Yoon Y, et al. CD27, a member of the tumor
1572. Pippin BA, Rosenstein M, Jacob WF, et al. Local IL-4 delivery necrosis factor receptor family, induces apoptosis and binds
enhances immune reactivity to murine tumors: Gene therapy in Siva, a proapoptotic protein. Proc Natl Acad Sci USA 1997; 94:
combination with IL-2. Cancer Gene Therapy 1994; 1: 35–42. 6346–6351.
1573. Pirhonen J, Matikainen S, Julkunen I. Regulation of virus- 1592. Prieyl JA, LeBien TW. Interleukin 7 independent development of
induced IL-12 and IL-23 expression in human macrophages. human B cells. Proc Natl Acad Sci USA 1006; 93: 10348–10353.
J Immunol 2002; 169: 5673–5678. 1593. Pruitt JH, Welborn MB, Edwards PD, et al. Increased soluble inter-
1574. Pisa P, Halapi E, Pisa EK, et al. Selective expression of interleu- leukin-1 type II receptor concentrations in postoperative patients
kin 10, interferon γ, and granulocyte macrophage colony stimu- and in patients with sepsis syndrome. Blood 1996: 87: 3282.
lating factor in ovarian cancer biopsies. Proc Nat Acad Sci 1594. Pulaski BA, Yeh KY, Shastri N, et al. Interleukin-3 enhances
1992; 89: 7708. cytotoxic T lymphocyte development and class I major histo-
1575. Piskin G, Sylva-Steenland RM, Bos JD, et al. In vitro expres- compatibility complex re-presentation of exogenous antigen by
sion of IL-23 by keratinocytes in healthy skin and psoriasis tumor-infiltrating antigen-presenting cells. Proc Natl Acad Sci
lesions: enhanced expression in psoriatic skin. J Immunol 2006; USA 1996; 93: 3669.
176: 1908–1915. 1595. Pulendran B, Banchereau J, Burkeholder S, et al. Flt-3 ligand
1576. Pitti RM, Marsters SA, Ruppert S, et al. Induction of apoptosis and granulocyte-macrophage colony stimulating factor mobi-
by Apo-2 ligand, a new member of the tumor necrosis factor lize distinct human dendritic cell subsets in vivo. J Immunol
cytokine family. J Biol Chem 1996; 271: 12687. 2000; 165: 566–572.
1577. Pizaro TT, Michie MH, Bentz M, et al. IL-18, a novel immuno- 1596. Pulendran B, Smith JL, Caspary G, et al. Distinct dendritic cell
regulatory cytokine, is upregulated in Crohn’s disease: expres- subsets differentially regulate the class of immune response in
sion and localization in intestinal mucosal cells. J Immunol vivo. Proc Natl Acad Sci USA 1999; 96: 1036.
1999; 162: 6829. 1597. Punnonen J, Aversa G, Cocks BG, et al. Interleukin 13 induces
1578. Plate JMD, Knospe WH, Harris JE, Gregory SA. Normal and interleukin 4 independent IgG4 and IgE synthesis and CD 23
aberrant expression of cytokines in neoplastic cells from expression by B cells. Proc Nat Acad Sci USA 1993; 90:
lymphocytic leukemias. Human Immunol 1993; 36: 249–258. 3730–3735.
1579. Platzer C, Meisel C, Vogt K, Platzer M, Volk HD. Up-regulation 1598. Punnonen J, de Vries JE. IL-13 induces proliferation, Ig isotype
of IL-10 by tumor necrosis factor-α and cAMP elevating drugs. switching, and Ig synthesis by immature fetal B cells. J Immunol
Int Immunol 1995; 7: 517. 1994; 152: 1094.
1580. Plummer R, Attard G, Pacey S, et al. Phase I and pharmacoki- 1599. Puolakkainen P, Twardzik D, Ranchalis J, et al. Increase of
netic study of lexatumumab in patients with advancer cancers. plasma transforming growth factor β (TGFβ) during immuno-
Clin Cancer Res 2007; 13: 6187–6194. therapy with IL-2. Cancer Invest 1995; 13: 583–589.
1581. Pober JS, Lapeirre LA, Stolpen AH. Activation of cultured 1600. Puren AJ, Fantuzzi G, Dinarello CA. Gene expression, synthe-
human endothelial cells by recombinant lymphotoxin: com- sis and secretion of IL-1β and IL-18 are differentially regulated
parison with tumor necrosis factor and interleukin-1 species. in human blood mononuclear cells and mouse spleen cells. Proc
J Immunol 1987; 138: 319. Natl Acad Sci 1999; 96: 2556.
1582. Poindexter NJ, Walch ET, Chada S, Grimm EA. Cytokine 1601. Puren AJ, Fantuzzi G, Gu Y, Su M S-S, Dinarello CA.
induction of interleukin-24 in human peripheral blood mononu- Interleukin-18 (INF-γ-inducing factor) induces IL-1β and IL-8
clear cells. J Leukocyte Biol 2005; 78: 745–752. via TNFα production from non-CD14+ human blood mononu-
1583. Polek TC, Talpaz M, Darnay BG, et al. Tweak mediates signal clear cells. J Clin Invest 1998; 101: 711.
transduction and differentiation of RAW264.7 cells in the 1602. Puri RK, Ogata M, Leland GM, et al. Expression of high affinity
absence of Fn/TweakR. J Biol Chem 2003; 278: 32317–32323. Interleukin 4 receptors on murine sarcoma cells and receptor-
1584. Pollok KE, Kim YJ, Hurtado J, et al. 4-1BB T cell antigen binds mediated cytotoxicity of tumor cells to chimeric protein between
to mature B cells and macrophages, and costimulates anti- Interleukin 4 and pseudomonas exotoxin. Cancer Res 1991; 51:
mu-primed B cells. Eur J Immunol 1994; 24: 367–374. 3011–3017.
1585. Pollok KE, Kim YJ, Zhou Z, et al. Inducible T cell antigen 1603. Qi J, Kreutzer DL. Fibrin activation of vascular endothelial
4-1BB: analysis of expression and function. J Immunol 1993; cells. Induction of IL-8 secretion. J Immunol 1995; 155:
150: 771–781. 867–876.
Walter M. Lewko and Robert K. Oldham 257

1604. Qi JC, Wang J, Mandadi S, et al. Human and mouse mast cells 1622. Reali C, Curto M, Sogos V, et al. Expression of CD137 and its
use the tetraspanin CD9 as an alternate interleukin-16 receptor. ligand in neurons, astrocytes, and microglia: modulation by
Blood 2007; 107: 135–142. FGF-2. J Neurosci Res 2003; 74: 67–73.
1605. Queen MM, Ryan RE, Holzer RG, et al. Breast cancer cells stim- 1623. Redlich CA, Gao X, Rockwell S, Kelly M, Elias JA. IL-11
ulate neutrophils to produce oncostatin M: Potential implications enhances survival and decreases TNF production after radia-
for tumor progression. Cancer Res 2005; 65: 8896–8904. tion-induced thoracic injury. J Immunol 1996; 157: 1705.
1606. Quinn TD, Miller FN, Wilson MA, et al. Interleukin-2-induced 1624. Redlinger RE Jr., Maillard RB, Lotze MT, et al. Synergistic
lymphocyte infiltration of multiple organs is differentially sup- Interleukin-18 and low dose interleukin-2 promote regression of
pressed by soluble tumor necrosis factor receptor. J Surg Res established murine neuroblastoma in vivo. J Pediatric Surg
1990; 56: 117. 2003; 38: 301–307.
1607. Quinn, Dejoy S, Jeyasellan R, Torley L, et al. Attenuation of 1625. Redman BG, Abubakr Y, Chou T, Esper P, Flaherty LE. Phase II
interleukin-2 induced pulmonary vascular leak syndrome by recombinant interleukin-1β in patients with metastatic renal cell
low doses of oral methotrexate. Cancer Res 1995; 55: 4929. carcinoma. J Immunother Emphasis Tumor Immunol 1994; 16:
1608. Rabin EM, Ohara J, and Paul WE. B cell stimulatory factor 1 211–215.
activates resting B cells. Proc Nat Acad Sci USA 1985; 82: 1626. Refaeli Y, Van Parijs L, London CA, Tschopp J, Abbas A.
2935–2939. Biochemical mechanisms of IL-2 regulated Fas mediated T cell
1609. Rabinovitch A, Suarez-Pinson O, Sorenson O, Rajotte RV, apoptosis. Immunity 1998; 9: 615.
Power RF. TNF-α down-regulates type I cytokines and prolongs 1627. Reich K, Garbe C, Blaschke V, et al. Response of psoriasis to
survival of syngeneic islet grafts in nonobese diabetic mice. J interleukin-10 is associated with suppression of cutaneous type
Immunol 1997; 159: 6298. I inflammation, downregulation of the epidermal interleukin-8/
1610. Racadot E, Audhuy B, Duvernoy H, et al. Clinical and immuno- CXCR2 pathway and normalization of keratinocyte maturation.
logical follow-up of patients with AIDS-associated Kaposi’s J Invest Dermatol 2001; 116: 319–329.
sarcoma treated with an anti-IL-6 monoclonal antibody. 1628. Reitamo SA, Remitz A, Uitto J. Interleukin-10 modulates type I
Cytokines Mol Ther 1995; 1: 133–1338. collagen and matrix metalloprotease gene expression in cul-
1611. Radaeva S, Sun R, Pan HN, et al. Interleukin 22 (IL-22) plays a tured human skin fibroblasts. J Clin Invest 1994; 94: 2489.
protective role in T cell-mediated murine hepatitis: IL-22 is a 1629. Reitschel E, Brade H, Holst O, et al. Bacterial endotoxin: chem-
survival factor for hepatocytes via STAT3 activation. Hepatology ical constitution, biological recognition, host response, and
2004; 39: 1332–1342. immunological detoxification. Curr Topics Microbial Immunol
1612. Rafi AQ, Zeytun A, Bradley MJ, et al. Evidence for involvement 1996; 216: 40.
of Fas ligand and perforin in the induction of vascular leak syn- 1630. Reittie JE, Hoffbrand AV. Interleukin-4 (IL-4) inhibits prolifera-
drome. J Immunol 1998; 161: 3077–3086. tion and spontaneous cytokine release by chronic leukemia
1613. Rahangdale S, Morgan R, Heijens C, et al. Chemokine receptor cells. Leukemia Res 1994; 18: 55–60.
CXCR3 desensitization by IL-16/CD4 signaling is dependent 1631. Ren F, Zhan X, Martens G, et al. Pro-IL-16 regulation in
on CCR5 and intact membrane cholesterol. J Immunol 2006; activated murine CD4+ lymphocytes. J Immunol 2005; 174:
176: 2337–2345. 2738–2745.
1614. Ralston SH, Graham R, Russel G, Gowen M. Estrogen inhibits 1632. Renauld J-C, van der Lugt, Vink A, et al. Thymic lymphomas in
release of tumor necrosis factor from peripheral blood mononu- interleukin 9 transgenic mice. Oncogene 1994; 9: 1327.
clear cells in postmenopausal women. J Bone Miner Res 1990; 1633. Renauld J-C, Vink A, Louahed A, Van Snick J. Interleukin-9 is
5: 983. a major anti-apoptotic factor for thymic lymphomas. Blood
1615. Raman VS, Akondy RS, Rath S, et al. Ligation of CD27 on B 1995; 85: 1300.
cells in vivo during primary immunization enhances commitment 1634. Renauld JC, Druez C, Kermouni A, et al. Expression cloning of
to memory B cell responses. J Immunol 2003; 171: 5876–5881. murine and human Interleukin 9 receptor DNAs. Proc Nat Acad
1616. Raman VS, Bal V, Rath S, George A. Ligation of CD27 on Sci USA 1992; 89: 5690–5694.
murine B cells responding to T-dependent and T-independent 1635. Renkl AC, Wussler J, Ahrens T, et al. Osteopontin functionally
stimuli inhibits the generation of plasma cells. J Immunol 2000; activates dendritic cells and induces their differentiation toward
165: 6809–6815. a Th1-polarization phenotype. Blood 2005; 106: 946–955.
1617. Ramesh R, Stewart AL, Boqui A. Melanoma differentiation 1636. Rennert PD, Browning JL, Mebius R, Mackey F, Hochman P.
associated antigen7/interleukin (IL)-24 is a novel ligand that Surface lymphotoxin α/β complex is required for the devel-
regulates angiogenesis via the IL-22 receptor. Cancer Res 2003; opment of peripheral lymphoid organs. J Exp Med 1996;
63: 5105–5113. 184: 1999.
1618. Rand T, Cruikshank WW, Center DM, Weller PF. CD4-mediated 1637. Rennick D, Yang G, Muler-Sieburg C, et al. Interleukin 4 (B cell
stimulation of eosinophils: lymphocyte chemoattractant factor stimulatory factor 1) can enhance or antagonize the factor
and other CD4-binding ligands elicit eosinophil migration. J dependent growth of hematopoietic progenitor cells. Proc Nat
Exp Med 1991; 173: 1521. Acad Sci USA 1987; 84: 6889.
1619. Ranheim EK, Cantwell MJ, Kipps TJ. Expression of CD27 and 1638. Renz H, Domenico J, Gelfand EW. IL-4 dependent upregulation
its ligand, CD70, on chronic lymphocytic leukemia B cells. of IL-4 receptor expression in murine T and B cells. J Immunol
Blood 1995; 85: 3556–3565. 1991; 146: 3049.
1620. Ray A. A SAF binding site in the promoter region of human 1639. Revhaug A, Michie HR, Manson JM, et al. Inhibition of
γ-fibrinogen gene functions as an IL-6 response element. cyclooxygenase attenuates the metabolic response to endotoxin
J Immunol 2000; 165: 3411–3417. in humans. Arch Surg 1988; 123: 162.
1621. Re F, Sironi M, Muzio M, et al. Inhibition of interleukin-1 1640. Reznikov LL, Kim SH, et al. IL-18 binding protein
responsiveness by type II receptor gene transfer: a surface increases spontaneous and IL-1 induced prostaglandin
“receptor” with anti-interleukin-1 function. J Exp Med 1996; production via inhibition of IFN-γ. Proc Natl Acad Sci
183: 1841. USA 2000; 97: 2174.
258 Cytokines

1641. Rich BE, Kupper TS. Cytokines: IL-20 – a new effector in skin 1660. Romer J, Hasselager E, Norby PL, et al. Epidermal overexpres-
inflammation. Curr Biol 2001; 10: R531–534. sion of interleukin-19 and IL-20 mRNA in psoriatic skin dissap-
1642. Richards CD, Brown TJ, Shoyab M, Baumann H, Gaudie J. pears after short-term treatment with cyclosporine or calcipotriol.
Recombinant oncostatin-M stimulates the production of acute J Invest Dermatol 2003; 121: 1306–1311.
phase proteins in hepatocytes in vitro. J Immunol 1992; 148: 1661. Rook AH, Zaki MH, Wysocka M, et al. The role for interleu-
1731. kin-12 therapy of cutaneous T cell lymphoma. Ann NYAS 2001;
1643. Richards CD, Langdon C, Botelho F, Brown TJ, Agro A. 941: 177–184.
Oncostatin M inhibits IL-1-induced expression of IL-8 and 1662. Rooney IA, Butrovich KD, Glass AA, et al. The lymphotoxin-β
granulocyte-macrophage colony-stimulating factor by synovial is necessary and sufficient for LIGHT-mediated apoptosis of
and lung fibroblasts. J Immunol 1996; 156: 343–349. tumor cells. J Biol Chem 2000; 275: 14307.
1644. Richards CD, Shoyab M, Brown TJ, Gauldie J. Selective regu- 1663. Rosa LE, Satokar AA, Roth KM, et al. Interleukin-27R (WSX-
lation of metalloproteinase inhibitor (TIMP-1) by oncostatin M 1/T-cell cytokine receptor) gene deficient mice display enhanced
in fibroblasts in culture. J Immunol 1993; 150: 5596. resistance to leishmania donovani infection but develop severe
1645. Ridge JP, Rosa FD, Matsinger P. A conditioned dendritic cell liver immunopathology. Am J Pathol 2006; 168: 158–169.
can be a temporal bridge between a CD4+ T-helper and a T-killer 1664. Rose TM, Bruce AG. Oncostatin M is a member of the cytokine
cell. Nature 1998; 393: 474. family that includes leukemia-inhibitory factor, granulocyte
1646. Ringheim GE, Burgher KL, Heroux JA. Interleukin-6 mRNA colony-stimulating factor, and interleukin-6. Proc Natl Acad Sci
expression by cortical neurons in culture: evidence for neuronal USA 1991; 88: 8641.
sources of interleukin-6 production in the brain. J Neuroimmunol 1665. Rose-John S, Heinrich PC. Soluble receptors for cytokines and
1995; 63: 113–123. growth factors: generation and biological function. Biochem
1647. Rissoan MC, Soumelis V, Kadowaki N, et al. Reciprocal control J 1994; 300: 281.
of T helper cell and dendritic cell differentiation. Science1999; 1666. Rosenberg SA, Grimm EA, McGrogan M, et al. Biological
283: 1183. activity of recombinant human interleukin-2 produced in
1648. Roake JJ. Rao AS, Morris PJ, et al. Dendritic cell loss from non- Escherichia coli. Science 1984; 223: 1412.
lymphoid tissues after systemic administration of lipopolysac- 1667. Rosenberg SA, Lotze MT, Muul LM, et al. A progress report on
charide, tumor necrosis factor and interleukin 1. J Exp Med the treatment of 157 patients with advanced cancer using lym-
1995; 181: 2237. phokine-activated killer cells and interleukin-2 or high-dose
1649. Robb RJ. Human T-cell growth factor: purification and interac- interleukin-2 alone. N Engl J Med 1987; 316: 889.
tion with a cellular receptor. Lymphokine Res 1982; 1: 37. 1668. Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the
1650. Robertson MJ, Cameron C, Atkins MB, et al. Immunological systemic administration of autologous lymphokine-activated
effects of interleukin-12 administration by bolus intravenous killer cells and recombinant interleukin-2 to patients with meta-
injection to patients with cancer. Clin Cancer Res 1999; 5: static cancer. N Engl J Med 1985; 313: 1485.
9–16. 1669. Rosenberg SA, Lotze MT, Yang JC, et al. Prospective random-
1651. Robertson MJ, Mier JW, Logan T, et al. Clinical and biological ized trial of high-dose interleukin-2 alone or in conjunction
effects of recombinant human interleukin-18 administered by with lymphokine-activated killer cells for the treatment of
intravenous infusion to patients with advanced cancer. Clin patients with advanced cancer. J Natl Cancer Inst 1993; 85:
Cancer Res 2006; 15: 4265–4273. 622–632.
1652. Robinson D, Shibuya A, Mui A, et al. IGIF does not drive Th1 1670. Rosenberg SA, Mule JJ, Speiss PJ, et al. Regression of estab-
development but synergizes with IL-12 for interferon-γ produc- lished pulmonary metastases and subcutaneous tumor mediated
tion and activates IRAK and NFκB. Immunity 1997; 7: 571. by the systemic administration of high-dose recombinant inter-
1653. Rochester CL, Ackerman SJ, Zheng T, Elial JA. Eosinophil– leukin 2. J Exp Med 1985; 161: 1169.
fibroblast interactions: granule major basic protein interacts 1671. Rosenberg SA, Packard BS, Aebersold PM, et al. Use of tumor-
with IL-1 and Transforming-growth factor-β in the stimulation infiltrating lymphocytes and interleukin-2 in the immunother-
of lung fibroblast interleukin-6-type cytokine production. apy of patients with metastatic melanoma. N Engl J Med 1988;
J Immunol 1996; 156: 4449. 319: 1676.
1654. Rock FL, Hardiman G, Timans JC, Kastelein RA, Bazan JF. A 1672. Rosenberg SA, Spiess P, LaFreniere R. A new approach to the
family of human receptors structurally related to Drosophila adoptive immunotherapy of cancer with tumor-infiltrating lym-
Toll. Proc Natl Acad Sci USA 1998; 95: 588. phocytes. Science 1986; 233: 1318.
1655. Roda JM, Parihar R, Lehman A. Interleukin-21 enhances NK 1673. Rosenberg SA, Sportes C, Ahmadzadeh M, et al. IL-7 admin-
cell activation in response to antibody-coated targets. J Immunol istration to humans leads to the expansion of CD8+ and
2006; 177: 120–129. CD4+ cells but a relative decrease of CD4+ T-regulatory
1656. Rollins BJ. Chemokines. Blood 997; 90: 909. cells. J Immunother 2006; 29: 313–319.
1657. Rollo E, Laskin DE, Denhardt DT. Osteopontin inhibits nitric 1674. Rosenberg SA, Yang JC, Topalian SL, et al. The treatment of
oxide production and cytotoxicity by activated RAW264.7 mac- 283 consecutive patients with metastatic melanoma or renal cell
rophages. J Leukocyte Biol 1996; 60: 397. cancer using high-dose bolus interleukin-2. JAMA 1994; 271:
1658. Rolph MS, Kaufman SHE. CD40 signaling converts a mini- 907–913.
mally immunogenic antigen into a potent vaccine against the 1675. Rosenberg SA, Yang JC, White DE, Steinberg SM. Durability
intracellular pathogen Listeria monocytogenes. J Immunol of complete responses in patients with metastatic cancer treated
2001; 166: 5115–5121. with high dose interleukin-2: identification of antigens mediat-
1659. Romagnani S. Human TH1 and TH2 subset: Regulation of dif- ing the response. Ann Surg 1998; 228: 307–319.
ferentiation and role in protection and immunopathology. Int 1676. Rosenstein M, Ettinghausen SE, Rosenberg SA. Extravasation
Arch Allergy Immunopathology Int Arch Allergy Immunol of intravascular fluid mediated by the systemic administration
1992; 98: 279–285. of recombinant interleukin 2. J Immunol 1986; 137: 1735.
Walter M. Lewko and Robert K. Oldham 259

1677. Rosenstein M, Yron I, Kaufmann Y, Rosenberg SA. Lymphocyte- 1696. Rudland PS, Platt-Higgins A, El-Tanani M, et al. Prognostic
activated killer cells: lysis of fresh syngeneic natural killer-re- significance of the metastasis-associated protein osteopontin in
sistant murine tumor cells by lymphocytes cultured in interleukin human breast cancer. Cancer Res 2002; 62: 3417–3427.
2. Cancer Res 1984; 44: 1946. 1697. Rumsaeng V, Cruikshank WW, Foster B, et al. Human mast
1678. Rosnet O, Buhring H-J, Marchetto S, et al. Human FLT3/FLK2 cells produce the CD4+ lymphocyte chemoattractant factor,
receptor tyrosine kinase is expressed at the surface of normal and IL-16. J Immunol 1997; 159: 2904–2910.
malignant hematopoietic cells. Leukemia 1996; 10: 238–248. 1698. Russell JH, Rush B, Weaver C, Wand R. Mature T cells of auto-
1679. Rosnet O, Marchetto S, de Lapeyriere O, Birnbaum D. FLT3, a immune lpr/lpr mice have a defect in antigen stimulated suicide.
gene encoding a novel tyrosine kinase receptor of the PDGFR/ Proc Natl Acad Sci USA 1993; 90: 4409.
CDF1R family. Oncogene 1991; 3: 1641–1650. 1699. Russell SM, Johnson JA, Noguchi M, et al. Interaction of IL-2R
1680. Rosnet O, Mattel MG, Marchetto S, Birnbaum D. Isolation and and c chains with Jak1 and Jak3: Implications for XSCID and
chromosomal location of a novel FMS-like tyrosine kinase XCID. Science 1994; 266: 1042–1044.
gene. Genomics 1991; 9: 380–385. 1700. Ryan TC, Cruikshank WW, Kornfeld H, Collins TL, Center
1681. Rossi V, Breviario F, Ghezzi P, Dejena E, Mantovani A. DM. The CD4 associated tyrosine kinase p56 lck is required for
Prostacyclin synthesis induced in vascular cells by interleu- lymphocyte chemoattractant factor induced T-lymphocyte
kin-1. Science 1985; 229: 174. migration. J Biol Chem 1995; 270: 17081–17086.
1682. Roth I and Fisher SJ. IL-10 is an autocrine inhibitor of human 1701. Rygard K, Nakamura T, Spang-Thompsen M. Expression of the
placental cytotrophoblast MMP-9 production and invasion. Dev protooncogene cmet and c kit and their ligands, hepatocyte
Biol 1999; 205: 194. growth factor/scatter factor and stem cell factor in SCLC cell
1683. Roth MD, Gitlitz BJ, Kiertscher SM, et al. Granulocyte mac- lines and xenografts. British J Cancer 1993; 67: 37–46.
rophage colony-stimulating factor and interleukin 4 enhance the 1702. Saas P, Boucraut J, Walker PR, et al. Tweak stimulation of astro-
number and antigen presenting activity of circulating CD14+ and cytes and the proinflammatory consequences. Glia 2000; 32:
CD38+ cells in cancer patients. Cancer Res 2000; 60: 1934–1941. 102–107.
1684. Rothe H, Hausman A, Casteels K, et al. IL-18 inhibits diabetes 1703. Saas P, Walker PR, Hahne M, et al. Fas ligand expression by
development in non-obese diabetic mice by counter regulation of astrocytoma in vivo: maintaining immune privilege in the brain?
Th1-dependent destructive insulitis. J Immunol 1999; 163: 1230. J Clin Invest 1997; 99: 1173.
1685. Rothe J, Lesslauer W, Lotscher H, et al. Mice lacking the tumor 1704. Sabelko K, Blumenthal D, Ford R, Ambrus J. IL-14 producing
necrosis factor receptor 1 are resistant to TNF-mediated toxicity and responsive cells are found in germinal centers. FASEB J
but highly susceptible to infection by Listeria monocytogenes. 1994; 8: A999.
Nature 1993; 364: 798. 1705. Sacca R, Cuff C, Lesslauer W, Ruddle NH. Differential activities
1686. Rothenberg EV, Diamond RA, Pepper KA, Yang JA. IL-2 gene of secreted lymphotoxin-α1β2 in lymphotoxin-induced inflam-
inducibility in T cells before T cell receptor expression. Changes mation: critical role of TNF receptor 1 signaling. J Immunol
in signaling pathways and gene expression requirements during 1998; 160: 485–491.
intrathymic maturation. J Immunol 1990; 144: 1614. 1706. Sacca R, Turley S, Soong L, Mellman I, Ruddle NH. Transgenic
1687. Rousset, F, Garcia E, Peronne C, et al. Interleukin-10 is a potent expression of lymphotoxin restores lympnodes to lymphotoxin-
growth and differentiation factor for activated human B lym- α-deficient mice. J Immunol 1997; 159: 4252–4260.
phocytes. Proc Nat Acad Sci USA 1992; 89: 1890. 1707. Sad S, Krishnan L, Bleackley RC, et al. Cytotoxicity and weak
1688. Rouvier E, Luciani M-F, Mattei M-G, Denizot F, Goldstein P. CD40 ligand expression of CD8+ type 2 cytotoxic T cells
CTLA-8, cloned from an activated T cell, bearing AU rich restricts their B helper activity. Eur J Immunol 1997; 27: 914.
mRNA instability sequences and homologous to a herpesvirus 1708. Sad S, Marcotte R, Mosmann TR. Cytokine-induced differentia-
samiri gene. J Immunol 1993; 150: 5445. tion of precursor mouse CD8+ T cells into cytotoxic CD8+ T cells
1689. Rovere P, Vallinoto C, Bondanza A, et al. Bystander apoptosis secreting Th1 or Th2 cytokines. Immunity 1995; 2: 271–279.
triggers dendritic cell maturation and antigen presentation func- 1709. Sad S, Mosmann TR. Single IL-2 secreting precursor CD4 T
tion. J Immunol 1998; 161: 4467. cell can develop into either Th1 or Th2 cytokine secreting
1690. Roy EJ, Gawlick U, Orr BA, Rund LA, Webb AG, Kranz DM. phenotype. J Immunol 1994; 153: 3514.
IL-12 treatment of endogenously arising murine brain tumors. J 1710. Sadlack B, Lohler J, Schorle H, et al. Generalized autoimmune
Immunol 2000; 165: 7293–7299. disease in interleukin-2-deficient mice is triggered by an
1691. Roy M, Waldschmidt T, Arrufo A, Ledbetter A, Noelle RJ. The uncontrolled activation and proliferation of CD4+ T cells. Eur
regulation and the expression of gp39, the CD40 ligand, on nor- J Immunol 1995; 25: 3053.
mal and cloned CD4 T cells. J Immunol 1993; 151: 2497. 1711. Sadlack B, Merz H, Schorle H, et al. Ulcerative colitis-like
1692. Ruchatz H, Leung BP, Wei XQ, McInnes IB, Liew HY. Soluble disease in mice with a disrupted interleukin-2 gene. Cell
IL-15 receptor α chain administration prevents murine colla- 1993; 75: 253.
gen-induced arthritis: a role for IL-15 in antigen induced immu- 1712. Saeki T, Mhashilkar A, Swanson X, et al. Inhibition of human
nopathology. J Immunol 1998; 160: 5654. lung cancer growth following adenovirus-mediated mda-7 gene
1693. Ruchert R, Asadullah K, Seifert M, et al. Inhibition of keratino- expression in vivo. Oncogene 2002; 21: 4558–4566.
cyte apoptosis by IL-15: a new parameter in the pathogenesis of 1713. Saito F, Ohno Y, Morisawa K, et al. Role of IL-27-producing
psoriasis? J Immunol 2000; 165: 2240–2250. dendritic cells in Th1-immunity polarization in Lewis rats.
1694. Ruddle NH, Bergman CM, McGrath KM, et al. An antibody to Biochem Biophys Res Commun 2005; 338: 1773–1778.
lymphotoxin and tumor necrosis factor prevents transfer of exper- 1714. Sakaguchi S. The origin of FOXP3-expressing CD4+ regulatory T
imental allergic encephalomyelitis. J Exp Med 1990; 172: 1193. cells: thymus or periphery. J Clin Invest 2003; 112: 1310–1312.
1695. Ruddle NH, Waksman BH. Cytotoxic effect of lymphocyte– 1715. Sakai T, Kusugami K, Nishimura H, et al. Interleukin 15
antigen interaction in delayed hypersensitivity. Science 1967; activity in the rectal mucosa of inflammatory bowel disease.
157: 1060–1062. Gastroenterology 1998; 114: 1237.
260 Cytokines

1716. Sakata T, Iwagami S, Tsurata Y, et al. Constitutive expression of 1735. Scala G, Kuang YD, Hall RE, et al. Accessory cell function of
interleukin 7 mRNA and production of IL-7 by a cloned murine human B cells. I. Production of both interleukin-1 like activity
thymic stromal cell line. J Leukocyte Biol 1990; 144: 3015. and interleukin-1 inhibitory factor by an EBV-transformed B
1717. Salih HR, Kososksi SG, Haluska VF, Trail PA, Kiener PA, et al. cell line. J Exp Med 1984; 159: 1637.
Constitutive expression of functional 4-1BB (CD137) ligand on 1736. Schadendorf D, Bohm M, Moller P, Grunewald T, Czarnetzki
carcinoma cells. J Immunol 2000; 165: 2903–2910. BM. Interleukin 7 induces differential lymphokine-activated
1718. Samoilova EB, Horton JL, Hilliard B, Liu TST, Chen Y. killer cell activity against human melanoma cell, keratinocytes
IL-6-deficient mice are resistant to experimental autoimmune and endothelial cells. J Invest Dermatol 1994; 102: 838–842.
encephalomyelitis: roles of IL-6 in the activation and differen- 1737. Schadendorf D, Moller A, Algermissen B, et al. IL-8 produced
tiation of autoreactive T cells. J Immunol 1998; 161: 6480. by human malignant melanoma cells in vitro is an essential
1719. Sampaio EP, Kaplan G, Miranda A, et al. The influence of thali- autocrine growth factor. J Immunol 1993; 15: 2667–2675.
domide on the clinical and immunologic manifestation of 1738. Schaefer G, Venkataraman C, Schindler U. FISP (IL-4-Induced
erythema nodosum leprosum. J Infect Dis 1993; 168: 408–414. secreted protein), a novel cytokine-like molecule secreted by
1720. Sanborg CI, Imfeld KL, Zaldivar F Jr, et al. IL-4 expression Th2 cells. J Immunol 2001; 166: 5859–5863.
in human T cells is selectively inhibited by IL-1α and IL-1β. 1739. Scheu S, Alferink J, Potzel T, et al. Targeted disruption of
J Immunol 1995; 155: 5206–5212. LIGHT causes defects in costimulatory T cell activation and
1721. Sandborn WJ, Hanauer SB, Katz S, et al. Etanercept for active reveals cooperation with lymphotoxin β in mesenteric
Crohn’s disease: a randomized double blind, placebo-controlled lymphnode genesis. J Exp Med 2002; 95: 161324.
trial. Gastroenterology 2001; 121: 1241–1246. 1740. Schieferdecker HL, Schlaf G, Koleva M, Gotze O, Jungermann
1722. Sanderson CJ, Campbell HD, Young IG. Molecular and cellular K. Induction of functional anaphylatoxin C5a receptors on hepa-
biology of eosinophil differentiation factor (interleukin 5) and tocytes by in vivo treatment of rats with IL-6. J Immunol 2000;
its effects on human and mouse B cells. Immunol Rev 1988; 164: 5453–5458.
109: 29–50. 1741. Schindler H, Lutz MB, Rollinghoff M, Bogdan C. The produc-
1723. Sanderson CJ. Interleukin-5, eosinophils, and disease. Blood tion of interferon-γ by IL-12/IL-18-activated macrophages
1992; 79: 3101. requires STAT4 signaling and is inhibited by IL-4. J Immunol
1724. Sareneva T, Julkunen I, Matikainen S. IFN-α and IL-12 2001: 166: 3075.
Induce IL-18 receptor gene expression in human NK and T 1742. Schindler R, Mancilla J, Endres S, et al. Collelations and inter-
cells. J Immunol 2000; 165: 1933–1938. actions in the production of interleukin-6 (IL-6), IL-1, and
1725. Sareneva T, Matikainen S, Kurimoto M, Julkunen I. Influenza A tumor necrosis factor (TNF) in human blood mononuclear
virus-induced IFN-α/β and IL-18 Synergistically enhance IFN-γ cells: IL-6 suppresses IL-1 and TNF. Blood 1990; 75: 40.
gene expression in human T cells. J Immunol 1998; 160: 6032. 1743. Schluns KS, Cook JE, Le PL. TGF-β differentially modulates
1726. Sarin A, Conal-Cibotti M, Henkart PA. Cytotoxic effect of epidermal growth factor-mediated increases in leukemia-inhib-
TNF and lymphotoxin in T lymphoblasts. J Immunol 1995; itory factor, IL-6, IL-1α, and IL-1β in human thymic epithelial
155: 3716. cells. J Immunol 1997; 158: 2704–2712.
1727. Sarris AH, Kliche KO, Pethambaram O, et al. Interleukin-10 1744. Schmidt JA, Mizel SB, Cohen D, et al. Interleukin-1: a potential
levels are often elevated in serum of adults with Hodgkin’s dis- regulator of fibroblast proliferation. J Immunol 1982; 128: 2177.
ease and are associated with inferior failure-free survival. Ann 1745. Schmitt E, Beuscher HU, Huls C, et al. IL-1 serves as a second-
Oncol 1999; 10: 433–440. ary signal for IL-9 expression. J Immunol 1991; 147: 3848.
1728. Sato A, Ohtsuki M, Hata M, et al. Antitumor activity of 1746. Schmitt E, Germann T, Goedert P, et al. IL-9 production of
IFN-lambda in murine tumor models. J Immunol 2006; 176: naive CD4+ T cells depends on IL-2, is synergistically enhanced
7686–7694. by a combination of TGF-β and IL-4 and is inhibited by IFN-γ.
1729. Sato K, Akaki T, Tomioka H. Differential potentiation of anti- J Immunol 1994; 153: 3989.
mycobacterial activity and reactive nitrogen intermediate- 1747. Schmitt E, van Brandwijk R, Fischer H-G, Rude E. Establishment
producing ability of murine peritoneal macrophages activated of different T cell sublines using either interleukin 2 of interleu-
by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). kin 4 as growth factors. Eur J Immunol 1990; 20: 1709.
Clin Exp Immunol 1998; 112: 63. 1748. Schmitt E, van Brandwijk R, Van Snick J, Siebold B, Rude E.
1730. Sato TA, Widmer MB, Finkelman FD, et al. Recombinant solu- TCGFIII/P40 is produced by naive CD4+ T cells but is not a
ble murine IL-4 receptor can inhibit of enhance IgE responses in general T cell growth factor. Eur J Immunol 1989; 19: 2167.
vivo. J Immunol 1991; 150: 2717. 1749. Schmitter D, Lauber B, Fagg B, Stahel RA. Hematopoietic
1731. Sauane M, Gopalkrishnan RV, Lebedeva I, et al. MDA-7/IL-24 growth factors secreted by seven human pleural mesothelioma
induces apoptosis if diverse cancer cell lines through JAK/STAT cell lines: interleukin 6 production as a common feature. Int
independent pathways. J Cell Physiol 2003; 196: 334–345. J Cancer 1992; 51: 296–301.
1732. Sauder DN, Mounessa NL, Katz SI, et al. Chemotactic cytokines: 1750. Schmitz J, Owyang A, Oldham E, et al. IL-33 an interleukin-1
the role of leukocyte pyrogen and epidermal cell thymocyte-acti- like cytokine that signals via IL-1 receptor-related proteiST2
vating factor in neutrophil chemotaxis. J Immunol 1984; 132: 828. and induces T helper type 2-associated cytokines. Immunity
1733. Saura M, Martinez-Dalmau R, Minty A, Perez-Sala D, Lamas 2005; 23: 479–490.
S. Interleukin-13 inhibits inducible nitric oxide synthase expres- 1751. Schneider P, Bodmer JL, Thome M, Hoffmann K, Holler N,
sion in human mesangial cells. Biochem J 1996; 313: 641. Tschopp J. Characterization of two receptors for TRAIL. FEBS
1734. Scala E, D’Offizi G, Rosso R, et al. C-C chemokines, IL-16 and Lett 1997; 416: 329.
soluble antiviral factor activity are increased in cloned T cells 1752. Schobitz B, de Kloet ER, Sutano W, Holsber F. Cellular local-
from subjects with long term nonprogressive HIV infection. ization of interleukin-6 mRNA and interleukin 6 receptor
J Immunol 1997; 158: 4485–4492. mRNA in rat brain. Eur J Neurosci 1993; 5: 1426–1435.
Walter M. Lewko and Robert K. Oldham 261

1753. Schoenberger SP, Toes REM, van der Voort EIH, Offringa R, caspase-3-dependent mechanism. J Immunol 2000; 164:
Melief CJM. T cell help for cytotoxic T lymphocytes is medi- 3806–3814.
ated by CD40–CD40L interactions. Nature 1998; 393: 480. 1771. Scott JM, Wier ML, Wilson SM, et al. Osteopontin inhibits
1754. Schorle H, Holtschke T, Hunig T, Schimpl A, Horak I. inducible nitrogen oxide synthetase activity in rat vascular tis-
Development and function of T cells in mice rendered interleu- sue. Am J Physiol 1998; 275: H2258.
kin-2 deficient by gene targeting. Nature 1991; 352: 621. 1772. Screaton RG, Mongkolsapaya J, Xu XN, et al. TRICK2, a new
1755. Schroder J, Mrowietz U, Morita E, Christophers E. Purification alternatively spliced receptor that transduces the cytoplasmic
and partial biochemical characterization of a human monocyte- signal from TRAIL. Curr Biol 1997; 7: 693.
derived neutrophil activating peptide that lacks interleukin 1 1773. Sebire G, Delfraissy J-F, Demotes-Mainard A, et al.
activity. J Immunol 1987; 139: 3474. Interleukin-13 and interleukin-4 act as interleukin-6 inducers in
1756. Schroder JM, Christophers E. Secretion of novel and homoge- human microglial cells. Cytokine 1996; 8: 636.
neous neutrophil activating peptides by LPS stimulated human 1774. Sebok K, Woodside D, al-Aoukaty A. IL-8 induces the locomo-
endothelial cells. J Immunol 1989; 142: 244. tion on human IL-2 activated natural killer cells. Involvement of
1757. Schuerer-Maly CC, Eckmann L, Kagoff MF, et al. Colonic epi- a guanine nucleotide binding protein. J Immunol 1993; 150:
thelial cell lines as a source of interleukin-8: stimulation by 1524–1534.
inflammatory cytokines and bacterial lipopolysaccharide. 1775. Seder RA and Paul WE. Acquisition of lymphokine producing
Immunology 1994; 81: 85–91. phenotype by CD4+ T cells. Annu Rev Immunol 1994; 12: 635.
1758. Schuler M, Bruntsch U, Spath-Schwalbe E, Schrezenmeier H, 1776. Sedger LM, Shows DM, Blanton RA, et al. IFN-γ mediates a
et al. Lack of efficacy of recombinant human interleukin-6 in novel antiviral activity through dynamic modulation of TRAIL
patients with advanced renal cell cancer: results of a phase II and TRAIL receptor expression. J Immunol 1999; 163: 920.
study. Eur J Cancer 1998; 34: 754–756. 1777. Sehmi R, Cromwell O, Wardlaw AJ, et al. Interleukin 8 is a
1759. Schwab U, Stein H, Gerdes J, Lemke H, Kirchner HH, Schaat chemoattractant for eosinophils purified from subjects with
M. Production of a monoclonal antibody specific for Hodgkin’s blood eosinophilia but not from normal healthy subjects. Clin
disease and a subset of normal lymphoid cells. Nature 1982; Exp Allergy 1993; 23: 1027.
299: 65. 1778. Seino K, Kayagaki N, Okumura K, Yagita H. Antitumor effect
1760. Schwalb DM, Han H-M, Marino M, et al. Identification of a of locally produced CD95 ligand. Nat Med 1997; 3: 165.
new receptor subtype for tumor necrosis factor α. J Biol Chem 1779. Seki E, Tsutsui H, Nakano H, et al. Lipopolysaccharide-induced
1993; 268: 9949–9954. IL-18 secretion from murine Kupffer cells independently of
1761. Schwarz H, Valbracht J, Tuckwell J, Kempis J, Lotz M. ILA, the myeloid differentiation factor 88 that is critically involved in
human 4-1BB homologue, is inducible in lymphoid and their induction of production of IL-12 and IL-1β.J Immunol 2001;
cell lines. Blood 1995; 85: 1043. 166: 2651–2657.
1762. Schwarz GN, MacVittie TJ, Vignuelle RM, Patchen ML, 1780. Sekido N, Mukaida N, Harada A, et al. Prevention of lung rep-
Douches SD, Oppenheim JJ, Neta R. Enhanced hematopoietic erfusion injury in rabbits by a monoclonal antibody against
recovery in irradiated mice pretreated with interleukin-1 (IL-1). interleukin-8. Nature 1993; 365: 654–657.
Immunopharmacol Immunotoxicol 1987; 9: 371. 1781. Sekido Y, Obata Y, Ueda R, et al. Preferential expression of c-kit
1763. Schwarz H, Arden K, Lotz M. CD137, a member of the tumor protooncogene transcripts in small cell lung cancer. Cancer Res
necrosis factor receptor family is located on chromosome 1p36, 1991; 51: 2416–2419.
in a cluster of related genes, and co-localizes with several malig- 1782. Selin LK, Santolucito PA, Pinto AK, Szomolanyi-Tsuda E,
nancies. Biochem Biophys Res Commun 1997; 235: 699. Welsh RM. Innate immunity to viruses: Control of vaccinia virus
1764. Schwarz H, Blanco F, Valbracht J, Kempis J, Lotz M. ILA, a infection by γδ T cells. J Immunol 2001; 166: 6784–6794.
member of the human NGF/TNF receptor family regulates T 1783. Senaldi G, Stolina M, Guo J, et al. Regulatory effects of novel
lymphocyte proliferation and survival. Blood 1996; 87: 2839. neurotrophin-1/B cell-stimulating factor-3 (Cardiotrophin-like
1765. Schwarz H, Tuckwell J, Lotz M. A receptor induced by lymphocyte cytokine) on B cell function. J Immunol 2002; 168: 5690–5698.
activation (ILA): a new member of the human nerve growth factor/ 1784. Senaldi G, Varnum BC, Sarmiento U, et al. Novel neurotrophin-
tumor necrosis factor receptor family. Gene 1993; 134: 295. 1/B cell-stimulating factor-3: a cytokine of the IL-6 family. Proc
1766. Schwarzenberger P, Huang W, Oliver P, et al. IL-17 mobilizes Natl Acad Sci USA 1999; 96: 11458–11463.
peripheral blood stem cells with short- and long-term repopulat- 1785. Senger, DR, Peruzzi CA. Secreted phosphoprotein markers for
ing ability in mice. J Immunol 2001; 167: 2081–2086. neoplastic transformation of human epithelial and fibroblast
1767. Schwarzenberger P, Huang W, Ye P, et al. Requirement for cells. Cancer Res 1985; 45: 5818–5823.
endogenous stem cell factor and granulocyte-colony-stimulat- 1786. Serretta V, Piazza B, Pavone C, Piazza S, Pavone-Macaluso M.
ing factor for IL-17 mediated granulopoiesis. J Immunol 2000; Is there a role for recombinant tumor necrosis factor α in the
164: 4783–4789. intravesical treatment of superficial bladder tumors? A phase II
1768. Schwarzenberger P, La Russa V, Miller A, et al. IL-17 stimulates study. Int J Urol 1995; 2: 100–103.
granulopoiesis in mice: Use of an alternate, novel gene therapy 1787. Setareh M, Schwarz H, Lotz M. A mRNA variant encoding a
derived method for in vivo evaluation of cytokines. J Immunol soluble form of 4-1BB, a member of the murine NGF/TNF
1998; 161: 6383–6389. receptor family. Gene 1995; 164: 311–315.
1769. Schweizer RC, Welmers BAC, Raaijmakers JAM, et al. RANTES 1788. Sguadari C, Angiolillo AL, Cherney BW, et al. Interferon-
and Interleukin 8 induced responses in normal human eosino- inducible protein-10 identified as a mediator of tumor necrosis
phils: effects of priming with IL-5. Blood 1994; 83: 3697. in vivo. Proc Natl Acad Sci USA 1996; 93: 13791–13796.
1770. Sciaky D, Brazer W, Center DM, Cruikshank WW, Smith TJ. 1789. Shade RJ, Younes A. In vitro and in vivo biologic effects of
Cultured human fibroblasts express constitutive IL-16 mRNA: interleukin-3 in follicular low-grade lymphoma. Leuk
Cytokine induction of active IL-16 protein synthesis through a Lymphoma 1997; 26: 17–25.
262 Cytokines

1790. Shaefer G, Venkataraman C, Schindler U. FISP (IL-4 induced 1809. Shibuya K, Robinson D, Zonin F, et al. IL-1α and TNF-α are
secreted protein), a novel cytokine-like molecule secreted by required for IL-12-induced development of Th1 cells producing
Th2 cells. J Immunol 2001; 166: 5859–5863. high levels of IFN-γ in BALB/c but not C57BL/6 mice. J
1791. Shah AJ, Smogorzewska EM, Hannum C, Crooks GM. Flt-3 Immunol 1998; 160: 1708–1716.
ligand induces proliferation of quiescent human bone marrow 1810. Shiloni E, Eisenthal A, Sachs D, Rosenberg SA. Antibody-
CD34+ CD38− cells and maintains progenitor cells in vivo. dependent cellular cytotoxicity mediated by murine lympho-
Blood 1996; 87: 3563. cytes activated in recombinant interleukin 2. J Immunol 1987;
1792. Shalaby MR, Aggarwall BB, Rinderknetch E, et al. Activation 138: 1992.
of human polymorphonuclear neutrophil function by interferon-γ 1811. Shimizu M, Shimamura M, Owaki T, et al. Antiangiogenic and
and tumor necrosis factor. J Immunol 1985; 135: 2069. antitumor activities of IL-27. J Immunol 2006; 176: 7317–7324.
1793. Shalhoub V, Faust J, Boyle WJ, et al. Osteoprotegerin and 1812. Shintani F, Nakati T, Kanba S, et al. Involvement of unterleu-
osteoprotegerin ligand effects on osteoclast formation from kin-1 in immobilization stress-induced increase in plasma adre-
human peripheral blood mononuclear cell precursors. J Cell nocorticotropic hormone and in the release of hypothalmic
Biochem 1999; 72: 251. monoamines in the rat. J Neurosci 1995; 15: 1961–1970.
1794. Shan B, Yu L, Shimazoto O, et al. Expression of interleukin-21 1813. Shipman CM and Croucher PI. Osteoprotegerin is a soluble
and IL-23 in human esophageal tumors produced antitumor decoy receptor for tumor necrosis factor apoptosis-inducing
effects in nude mice. Anticancer Res 2004; 24: 79–82. ligand/apo2 ligand and can function as a paracrine survival fac-
1795. Shan BE, Hao JS, Li OX, et al. Antitumor activity and immune tor for human myeloma cells. Cancer Res 2003; 63: 912–916.
enhancement of murine interleukin-23 expressed in murine 1814. Shiraki K, Tsuji N, Shioda T, Isselbacher KJ, Takahashi H.
colon carcinoma cells. Cell Mol Immunol 2006; 3: 47–52. Expression of Fas ligand in liver metastases of colon adenocar-
1796. Shanker G, Sorci-Thomas M, Adams ME. Estrogen modulates cinoma. Proc Natl Acad Sci USA 1997; 94: 6420.
the expression of tumor necrosis factor mRNA in phorbol ester- 1815. Shoham S, DavenneD, Cady AB, Dinarello CA, and Krueger
stimulated human monocytic THP-1 cells. Lymphokine JM. Recombinant tumor necrosis factor and IL-1 enhance slow-
Cytokine Res 1994; 13: 377. wave sleep. Am J Physiol 1987; 253: 142.
1797. Sharma V, Sparks JL Vail JD. Human B-cell lines constitutively 1816. Shrikant P, Weber E, Jilling T, Beneviste EN. ICAM gene
express and secrete interleukin-16. Immunology 2000; 99: expression by glial cells: differential mechanisms of inhibition
266–271. by IL-6 and IL-10. J Immunol 1995; 155: 1489.
1798. Shaw J-P, Utz PJ, Durand DB, et al. Identification of a puta- 1817. Shu U, Kiniwa M, Wu CY, et al. Activated T cells induce inter-
tive regulator of early T cell activation genes. Science 1988; leukin-12 production by monocytes via CD-40–CD-40L inter-
241: 202. action. J Immunol 1995; 25: 1125–1128.
1799. Shaw J, Caplan B, Paetkau V, et al. Cellular origins of co-stim- 1818. Shuford WW, Klussman K, Trichler DD, et al. 4-1BB costimu-
ulator (IL-2) and its activity in cytotoxic T-lymphocyte latory signals preferentially induce CD8+ T cell proliferation
responses. J Immunol 1980; 124: 2231. and lead to the amplification in vivo of cytotoxic T cell response.
1800. Shaw J, Monticone V, Paetkau V. Partial purification and molec- J Exp Med 1997; 186: 47.
ular characterization of a lymphokine (co-stimulator) required 1819. Shurin MR, Pandharipande PP, Zorina TD, et al. FLT3-ligand
for the mitogenic response of mouse thymocytes in vitro. J induces the generation of functionally active dendritic cells in
Immunol 1978; 120: 1967. mice. Cell Immunol 1997; 179: 174.
1801. Shaw SG, Maung AA, Steptoe RJ, et al. Expansion of functional 1820. Sica D, Rayman P, Edinger M, et al. Interleukin 7 enhances the
NK cells in multiple tissue compartments of mice treated with proliferation and effector function of tumor infiltrating lympho-
Flt3-ligand: implications for anticancer and antiviral activity. J cytes from renal cell carcinoma. International J Cancer 1993;
Immunol 1998; 161: 2817–2814. 53: 941–947.
1802. Sheikh F, Baurin VV, Lewis-Antes A, et al. IL-26 signals 1821. Sideras P, Palacios R. Bone marrow pre-T and pre-B lympho-
through a novel receptor complex composed of Il-20 receptor 1 cyte clones express functional receptors for interleukin (IL)-3
and IL-10 receptor 2. J Immunol 2004; 172: 2006–2010. and interleukin 4/BSF-1 and non-functional receptors for IL-2.
1803. Shen L, Zhang C, Wang T, et al. Development of autoimmunity Eur J Immunol 1987; 17: 217.
in IL-14α-transgenic mice. J Immunol 2006; 177: 5676–5686. 1822. Siebruth D, Jabs EW, Warrington JA, et al. Assignment of
1804. Shen MM, Skoda RC, Cardiff RD, et al. Expression of LIF in NKSF/IL-12, a unique cytokine composed of two unrelated
transgenic mice results in altered thymic epithelium and appar- subunits to chromosomes 3 and 5. Genomics 1992; 14: 59–62.
ent intraconversion of thymic and lymphnode morphologies. 1823. Siegel JP, Sharon M, Smith PL, Leonard WJ. The IL-2 receptor
EMBO J 1994; 13: 1375. β chain (p70): role in mediating signals for LAK, NK, and pro-
1805. Sheridan JP, Marsters SA, Pitti RM, et al. Control of TRAIL- liferative activities. Science 1987; 238: 75.
induced apoptosis by a family of signaling and decoy receptors. 1824. Sillaber C, Sperr WR, Agis H, et al. Inhibition of stem cell fac-
Science 1997; 277: 818. tor dependent formation of human mast cells by interleukin-3
1806. Shevach EM. Certified professionals: CD4+ CD25+ suppressor and interleukin-4. Int Arch Allergy Immunol 1994; 105: 264.
T cells. J Exp Med 2001; 193: F41–46. 1825. Silvani A, Ferrari G, Paonessa G, et al. Down regulation of
1807. Shi Y, Ullrich SJ, Zhang J, et al. A novel cytokine receptor-li- interleukin 6 receptor α chain in interleukin 6 transduced mela-
gand pair. Identification, molecular characterization, and in vivo noma cells causes selective resistance to interleukin 6 but not to
immunomodulatory activity. J Biol Chem 2000; 275: oncostatin M. Cancer Res 1995; 55: 2200–2205.
19167–19176. 1826. Silver DF, Hempling RE, Piver MS, Repasky EA. Flt-3 ligand
1808. Shi Y, Wang W, Youri PA, et al. Computational EST database inhibits growth of human ovarian tumors engrafted in severe
analysis identifies a novel member of the neuropoietic cytokine combined immunodeficient mice. Gynecol Oncol 2000, 77:
family. Biochem Biophys Res Commun 1999; 262: 132. 377–382.
Walter M. Lewko and Robert K. Oldham 263

1827. Simic MM, Stosic-Grujicic S. The dual role of interleukin 1 1846. Smyth MJ, Jonstone RW, Cretney E, et al. Multiple deficiencies
in lectin-induced proliferation of T cells. Folia Biol 1985; underlie NK cell inactivity in lymphotoxin-α gene targeted
31: 410. mice. J Immunol 1999; 163: 1350.
1828. Simonet WS, Lacey DL, Dunstan CR, et al. Osteoprotegerin, a 1847. Smyth MJ, Ortaldo JR. Comparison of the effect of IL-2 and
novel secreted protein involved in the regulation of bone den- IL-6 on the lytic activity of purified human peripheral blood
sity. Cell 1997; 89: 309. large granular lymphocytes. J Immunol 1991; 146: 1380–1384.
1829. Sims JE, March CJ, Cosman D, et al. cDNA expression cloning 1848. Smyth MJ, Taniguchi M, Street SEA. The antitumor activity of
of the IL-1 receptor, a member of the immunoglobulin super- IL-12: Mechanisms of innate immunity that are model and dose
family. Science 1988; 241: 585. dependent. J Immunol 2000; 165: 2665–2670.
1830. Singh B, Berry JA, Shorer A, et al. COX-2 induces IL-11 pro- 1849. Smyth MJ, Wallace ME, Nutt SL, et al. Sequential activation of
ductioin in breast cancer cells. J Surg Res 2006; 131: 276–275. NKT cells and NK cells provides effective innate immunother-
1831. Singh K, Sirokman G, Communal C, et al. Myocardial osteo- apy of cancer. J Exp Med 2005; 201: 1973–1985.
pontin expression coincides with the development of heart fail- 1850. Snapper CM, Finkelman FD, and Paul WE. Differential regula-
ure. Hypertension 1999; 33: 670. tion of IgG1 and IgE synthesis by interleukin 4. J Exp Med
1832. Singh N, Hong S, Scherer DC, et al. Activation of NK T cells 1988; 167: 183.
by CD1d and α-galactosylceramide directs conventional T 1851. Snapper CM, Paul WE. Interferon γ and B cell stimulatory fac-
cells to the acquisition of a Th2 phenotype. J Immunol 1999; tor I reciprocally regulate Ig isotope production. Science 1987;
163: 2373. 236: 944–947.
1833. Singh RK, Gutman M, Radinsky R, et al. Expression of inter- 1852. Snyder LA, Goletz TJ, Gunn GR, et al. A MUC1/IL-18 DNA
leukin 8 correlates with the metastatic potential of human mela- vaccine induces anti-tumor immunity and increased survival in
noma cells in nude mice. Cancer Res 1994; 54: 3232–3247. MUC1 transgenic mice. Vaccine 2006; 24: 3340–3352.
1834. Singhal H, Bautista DS, Tonkin KS, et al. Elevated plasma 1853. So E-Y, Park H-H, Lee S-E Lee CE. IFN-γ and IFN-α posttran-
osteopontin in metastatic breast cancer associated with increased scriptionally down-regulate the IL-4-induced IL-4 receptor
tumor burden and decreased survival. Clin Cancer Res 1997; 3: gene expression. J Immunol 2000; 165: 5472–5479.
605–611. 1854. Son YI, Dallal RM, Lotze MT. Combined treatment with interleu-
1835. Sivakumar PV, Foster DC, Clegg CH. Interleukin-21 is a kin-18 and low-dose interleukin-2 induced regression of a murine
T-helper cytokine that regulates humoral immunity and cell sarcoma and memory response. J Immunother 2003; 234–240.
mediated anti-tumor responses. Immunology 2004; 112: 1855. Sondel PM, Hank JA, Kohler PC, et al. Destruction of autolo-
177–182. gous human lymphocytes by interleukin 2-activated cytotoxic
1836. Skillings J, Wierzbicki R, Eisenhauer E, et al. A phase II study cells. J Immunol 1986; 137: 502.
of recombinant tumor necrosis factor in renal cell carcinoma: a 1856. Song K, Chen Y, Goke R, et al. Tumor necrosis factor-related
study of the National Cancer Institute of Canada Clinical Trials apoptosis-inducing ligand (TRAIL) is an inhibitor of autoim-
Group. J Immunother 1992; 11: 67–70. mune inflammation and cell cycle progression. J Exp Med 2000;
1837. Small D, Levenstein M, Kim E, et al. STK-1, the human 191: 1095.
homolog of Flk-2/Flt3, is selectively expressed in CD34+ 1857. Song MM, Shaui K. the suppressor of cytokine signaling
human bone marrow cells and is involved in the proliferation of (SOCS) 1 and SOCS3 but not SOCS2 proteins inhibit interfer-
early progenitor/stem cells. Proc Natl Acad Sci USA 1994 91: on-mediated antiviral and antiproliferative activities. J Biol
459–463. Chem 1998; 273: 35056.
1838. Smalley RV and Oldham RK. Biological response modifiers: 1858. Song X, Voronov E, Dvorkin T, et al. Differential effects of
preclinical evaluation and clinical activity. Crit Rev Oncol IL-1α and IL-1β on tumorigenicity patterns and invasiveness. J
Hematol 1984; 1: 259–294. Immunol 2003; 171: 6448–6456.
1839. Smith CA, Farrah T, Goodwin RG. The TNF receptor superfam- 1859. Sonkoly E, Muller A, Lauerma AI, et al. IL-31: a new link
ily of cellular and viral proteins: activation, costimulation and between T cells and pruritis in atopic skin inflammation. J
death. Cell 1994; 76: 959. Allergy Clin Immunol 2006; 117: 411–417.
1840. Smith CA, Gruss HJ, Davis G, et al. CD30 antigen, a marker for 1860. Sonoda Y, Kimura T, Sakabe H, et al. Human FLT3 ligand acts
Hodgkin’s lymphoma, is a receptor whose ligand defines an on myeloid as well as multipotential progenitors derived from
emerging superfamily of cytokines with homology to TNF. Cell purified CD34+ blood progenitors expressing different levels of
1993; 73: 1349. c-kit protein. Eur J Haematol 1997; 58: 257–264.
1841. Smith E, Zrabock, Stark MA, et al. IL-23 is required for neutro- 1861. Soong L, Xu J-C, Grewall IS, Kima P, et al. Disruption of
phil homeostasis in normal and neutrophilic mice. J Immunol CD40–CD40L interactions results in a susceptibility to
2007; 179: 8274–8279. Leishmania amazonensis infection. Immunity 1996; 4: 263.
1842. Smith JW 2nd, Longo DL, Alvord WG, et al. The effect of treat- 1862. Soroosh P, Ine S, Sugamura K, et al. OX40–OX40 ligand inter-
ment with interleukin-1α on platelet recovery after high-dose action through Tcell-Tcell contact contributes to CD4 T cell
carboplatin. N Engl J Med 1993; 328: 756–761. longevity. J Immunol 2006; 176: 5975–5987.
1843. Smith JW. Tolerability and side-effect profile of rhIL-11. 1863. Sotomayor EM, Borrello I, et al. Conversion of tumor-specific
Oncology 2000; 14: 41–47. CD4+ T-cell tolerance to T-cells priming through in vivo ligation
1844. Smith MR, Kung H, Durum SK, Colburn NH, Sun Y. TIMP-3 of CD40. Nat Med 1999; 5: 780.
induces cell death by stabilizing TNF-α receptors on the surface 1864. Southern E. Detection of specific sequences among DNA frag-
of human colon carcinoma cells. Cytokine 1997; 9: 770. ments separated by gel electrophoresis. J Mol Biol 1975; 98: 503.
1845. Smith XG, Bolton EM, Ruchatz H, et al. Selective blockade of 1865. Sower LE, Froelich CJ, Carney DH, et al. Thrombin induces
IL-15 by soluble IL-15 receptor-chain enhances cardiac allograft IL-6 production in Fibroblasts and Epithelial Cells. J Immunol
survival. J Immunol 2000; 165: 3444. 1995; 155: 895–901.
264 Cytokines

1866. Sparwasser T, Miethke T, Lipford G, et al. Macrophages sense 1884. Stefana B, Ray DW, Melmed S, et al. Leukemia inhibitory fac-
pathogens via DNA motifs: induction of tumor necrosis factor tor induces differentiation of pituitary corticotroph function: an
mediated shock. Eur J Immunol 1997; 27: 1671. immuno-neuroendocrine phenotype switch. Proc Natl Acad Sci
1867. Speer MY, McKee MD, Guldberg RE, et al. Inactivation of the USA 1996; 93: 12502–12506.
osteopontin gene enhances vascular calcification of matrix Gla 1885. Stefanovic-Racic M, Mollers MO, Miller LA, Evans CH. Nitric
protein-deficient mice: evidence for osteopontin as an inducible oxide and proteoglycan turnover in rabbit articular cartilage.
inhibitor of vascular calcification in vivo. J Exp Med 2002; 196: J Orthopaed Res 1997; 15: 442.
1047–1055. 1886. Steiger J, Nickerson PW, Steurer W, Moscovitch-Lopatin M,
1868. Spencer NFL and Daynes RA. IL-12 directly stimulates expres- Strom RB. IL-2 knockout recipient mice reject islet cell
sion of IL-10 by CD5+ B cells and IL-6 by both CD5+ and isografts. J Immunol 1995; 155: 489–498.
CD5− B cells: possible involvement in age-associated cytokine 1887. Steiner GE, Newman ME, Paikl D, et al. Expression and func-
dysregulation. Int Immunol 1997; 9: 745. tion of pro-inflammatory interleukin IL-17 and IL-17 receptor
1869. Spencer SD, Di Marco F, Hooley J, et al. The orphan receptor in normal, hyperplastic and malignant prostate. Prostate 2003;
CRF2–4 is an essential subunit of the interleukin-10 receptor. J 56: 171–182.
Exp Med 1998; 187: 571–578. 1888. Steinman RM. The dendritic cell system and its role in immuno-
1870. Spits H, Yssel H, Paliard X, et al. IL-4 inhibits IL-2 mediated genicity. Ann Rev Immunol 1991; 9: 271–296.
induction of human lymphokine activated killer cells but not the 1889. Steinshamn S, Brekke OL, Waage A. Soluble tumor necrosis
generation of antigen specific cytotoxic T lymphocytes in mixed factor receptors, tumor necrosis factor and interleukin-6 in
leukocyte cultures. J Immunol 1988; 141: 29–36. serum in granulocytopenic patients with fever. Brit J Haematol
1871. Splawski JB, Jelinek DF, Lipsky PE. Immunomodulatory role 1995; 89: 719.
of IL-4 on the secretion of Ig by human B cells. J Immunol 1890. Steptoe RJ, Fu F, Li W, Drakes ML, et al. Augmentation of den-
1989; 142: 1569. dritic cells in murine organ donors by Flt3 ligand alters the bal-
1872. Splawski JB, McAnally, Lipsky PE. IL-2 dependence of the ance between transplant tolerance and immunity. J Immunol
promotion of human B cell differentiation by IL-6 (BSF-2). J 1997; 159: 5483–5491.
Immunol 1990; 144: 562. 1891. Stern AS, Podlaski FJ, Hulmes JD, et al. Purification to homo-
1873. Spranger M, Lindholm D, Brandtlow C, et al. Regulation of geneity and partial characterization of cytotoxic lymphocyte
nerve growth factor (NGF) synthesis in the rat central nervous maturation factor from human B-lymphocyte cells. Proc Nat
system: comparison between the effects of interleukin-1 and Acad Sci USA 1990; 87: 6808–6812.
various growth factors in astrocyte cultures in vitro. Eur J 1892. Stewart CL, Kaspar P, Brunet LJ, et al. Blastocyst implantation
Neurosci 1990; 2: 69. depends on maternal expression of leukemia inhibitory factor.
1874. Squadari C, Farber JM, Angiolillo AL, et al. Mig, the monokine Nature 1992; 359: 76–79.
induced by interferon-gammma, promotes tumor necrosis in 1893. Stier S, Ko Y, Forkert R, et al. Osteopontin is a hematopoietic
vivo. Blood 1997; 89: 2635–2643. stem cell niche component that negatively regulates stem cell
1875. Srivatsa SS, Harrity PJ, Maercklein P, et al. Increased cellular pool size. J Exp Med 2005; 201: 1781–1791.
expression of matrix proteins, which regulate mineralization is 1894. Stoll S, Jonuleit H, Schmitt E, Muller G, et al. Production of
associated with calcification of human and porcine xenograft functional IL-18 by different subtypes of murine and human
bioprosthetic heart valves. J Clin Invest 1997; 99: 996–1009. dendritic cells (DC): DC-derived IL-18 enhances IL-12 depen-
1876. Stacey KJ, Sweet MJ, Hume DA. Macrophages ingest and are dent Th1 development. Eur J Immunol 1998; 28: 3231.
activated by bacterial DNA. J Immunol 1996; 157: 2116. 1895. Stoll SG, Mueller G, Kurimoto M, et al. Production of IL-18
1877. Stam TC, Swaak AJ, Kruit WH, Stoter G, Eggermont AM. (interferon-γ-inducing factor) messenger RNA and functional
Intrapleural administration of tumor necrosis factor α in patients protein by murine keratinocytes. J Immunol 1997; 159: 298.
with mesothelioma: cytokine patterns and acute-phase protein 1896. Stotter H, Custer MC, Bolton ES, et al. IL-7 induced lym-
response. Eur J Clin Invest 2000; 30: 336–343. phokine activated killer cell activity and is regulated by IL-4.
1878. Standiford T, Kunkel S, Basha M, et al. Interleukin 8 gene J Immunol 1991; 146: 150–155.
expression by a pulmonary epithelial cell line. J Clin Invest 1897. Strand S, Hoffmann WJ, Hug H, et al. Lymphocyte apoptosis
1990; 86: 1945. induced by CD95 (Apo-1/Fas) ligand-expressing tumor cells: a
1879. Starnes HF, Hartman G, Torti F, et al. Recombinant human mechanism for immune evasion? Nat Med 1996; 2: 1361.
interleukin-1β (IL-1β) has antitumor activity and acceptable 1898. Strasly M, Cavallo F, Geuna M, et al. IL-12 inhibition of
toxicity in metastatic malignant melanoma. Proc Am Soc Clin endothelial cell functions and angiogenesis depends on lym-
Oncol 1991; 10: 292. phocyte-endothelial cell cross talk. J Immunol 2001; 166:
1880. Starnes T, Broxmeyer HE, Robertson MJ, Hromas R. IL-17D, a 3890–3899.
novel member of the IL-17 family, stimulates cytokine produc- 1899. Strassen M, Arnold M, Hultner L, et al. Murine bone marrow-
tion and inhibits hematopoiesis. J Immunol 2002; 169: 642–646. derived mast cells as potent producers of IL-9: Costimulatory
1881. Starnes T, Robertson MJ, Sledge G, et al. IL-17F, a novel function of IL-10 and kit ligand in the presence of IL-1. J
cytokine selectively expressed in activated T cells and mono- Immunol 2000; 164: 5549–5555.
cytes, regulates angiogenesis and endothelial cytokine produc- 1900. Strassman G, Jacob CO, Fong M, Bertolini DR. Mechanisms of
tion. J Immunol 2001 167: 4137–4140. paraneoplastic syndromes of colon-26: involvement of interleu-
1882. Starr R, Willson TA, Viney EM, et al. A family of cytokine-in- kin 6 in hypercalcemia. Cytokine 1993; 5: 463–468.
ducible inhibitors of signaling. Nature 1997; 387: 917. 1901. Strassman G, Fong M, Freter CE, et al. Suramin interferes with
1883. Stashenko P, Dewirst FE, Peros WJ, Kent RL. Synergistic inter- Interleukin-6 receptor binding in vitro and inhibits colon 26
action between IL-1, tumor necrosis factor, and lymphotoxin in mediated experimental cachexia in vivo. J Clin Invest 1993; 92:
bone resorption. J Immunol 1987; 138: 1464. 2152–2159.
Walter M. Lewko and Robert K. Oldham 265

1902. Strengell M, Lehtonen A, Matikainen S, Julkunen I. IL-21 1920. Sudo Y, Shimazaki C, Ashihara E, et al. Synergistic effect of
enhances SOCS gene expression and inhibits LPS-induced FLT-3 ligand on the granulocyte colony stimulating factor
cytokine production in human monocyte-derived dendritic cells. induced mobilization of hematopoietic stem cells and progeni-
J Leukoc Biol 2006; 79: 1279–1285. tor cells into blood in mice. Blood 1997; 89: 3186–3191.
1903. Strengell M, Matikainen S, Siren J, et al. IL-21 in synergy with 1921. Sugarman BJ, Aggarwal BB, Hass PE, et al. Recombinant human
IL-15 or IL-19 enhances interferon-γ production in human NK tumor necrosis factor α: effects on proliferation of normal and
and T cells. J Immunol 2003; 170: 5464–5469. transformed cells in vitro. Science 1985; 230: 943–945.
1904. Strieter R, Kunkel S, Showell H, et al. Endothelial cell gene 1922. Sugimoto K, Ogawa A, Mizoguchi E, et al. IL-22 ameliorates
expression of a neutrophil chemotactic factor by TNFα, LPS, intestinal inflammation in a mouse model of ulcerative colitis. J
and IL-1β. Science 1989; 243: 1467. Clin Invest 2008; 118: 534–544.
1905. Strieter RM, Kunkel SL, Elner VM, et al. Interleukin-8: a cor- 1923. Sugita K, Robertson MJ, Ritz J, Schlossman SF, Morimoto C.
neal factor that induces neovascularization. Am J Pathol 1992; Participation of the CD27 antigen in the regulation of IL-2 acti-
141: 1279–1284. vated human NK cells. J Immunol 1992; 149: 1199.
1906. Strieter RM, Poverini PJ, Kunkel SL, et al. The functional role 1924. Sugita K, Torimoto Y, Nojima Y, et al. The 1A4 molecule
of the ELR motif in CXC chemokine-mediated angiogenesis. J (CD27) is involved in T cell activation. J Immunol 1991;
Biol Chem 1995; 270: 27348. 147: 1477.
1907. Strohmeyer T, Peter S, Hartman M, et al. Expression of the hst-1 1925. Sugita M, Yue Y, Foskett JK. CFTR CL-channel and CFTR-
and c-kit protooncogene in human testicular germ cell tumors. associated ATP channel pores regulated by common gates.
Cancer Res 1991; 51: 181–1816. EMBO J 1998; 17: 898.
1908. Stuber E, Neurath M, Calderhead D, Fell HP, Strober W. Cross- 1926. Sugiura S, Lahav R, Han J, et al. Leukemia inhibitory factor is
linking of OX40 ligand, a member of the TNF/NGF cytokine required for normal inflammatory responses to injury in the
family, induces proliferation and differentiation in murine peripheral and central nervous systems in vivo and is chemotac-
splenic B cells. Immunity 1995; 2: 507. tic for macrophages in vitro. Eur J Neurosci 2000; 12: 457.
1909. Stuber E, Strober W. The T cell–B cell interaction via OX40– 1927. Sun Y, Blink SE, Liu W, et al. Inhibition of Th2-mediated
OX40L is necessary for the T cell-dependent humoral immune allergic airway inflammation disease by CD137 costimulation.
response. J Exp Med 1996; 183: 979. J Immunol 2006; 177: 814–821.
1910. Stuber E, Von Frier A, Marinescu D, Folsch UR. Involvement of 1928. Sun Y, Jurgovsky K, Moller P, et al. Vaccination with IL-12 gene-
OX40–OX40L in the intestinal manifestations of the murine acute modified autologous melanoma cells: preclinical results and a
graft versus host disease. Gastroenterology 1998; 115: 1205. first clinical phase I study. Gene Therapy 1998; 5: 481–490.
1911. Stumhofer JS, Laurence A, Wilson EH, et al. Interleukin 27 1929. Sun Z, Yeargeau DA, Tuypens T, et al. Identification and char-
negatively regulates the development of interleukin 17-produc- acterization of a functional promoter region in the human
ing helper T cells during chronic inflammation on the central eosinophil IL-5 receptor α subunit gene. J Biol Chem 1995;
nervous system. Nat Immunol 2006; 7: 936–045. 270: 1462.
1912. Stutman O, Paige CJ, Figarella EF. Natural cytotoxic cells 1930. Sutherland CL, Rabinowitch B, Chalupny NJ, et al. ULBPs,
against solid tumors in mice. I. Strain and age distribution and human ligands for the NKG2D receptor, stimulate tumor immu-
target cell susceptibility. J Immunol 1979; 121: 1819. nity with enhancement by IL-15. Blood 2006; 108: 1313–1319.
1913. Su Z, Emdad L, Sauane M, et al. Unique aspects of mda-7/IL-24 1931. Suto A, Nakajima H, Hirose K, et al. Interleukin 21 prevents
antitumor bystrander activity: establishing a role for secretion antigen-induced IgE production by inhibiting germ line Cε tran-
of MDA-7/IL-24 protein by normal cells. Oncogene 2005; 24: scription of IL-4 stimulated B cells. Blood 2002; 100: 4565.
7552–7566. 1932. Suto A, Wurster AL, Reiner SL, et al. IL-21 inhibits IFN-γ pro-
1914. Su ZZ, Lebedeva IV, Sarkar D, et al. Ionizing radiation enhances duction in developing Th1 cells through the repression of eome-
therapeutic efficiency of mda-7/IL-24: overcoming radiation- sodermin expression. J Immunol 2006; 177: 3721–3727.
and mda-7/IL-24-resistance in prostate cancer cells overex- 1933. Suzuki H, Kundig T, Furlonger C, Wakeman A, et al. Deregulated
pressing the antiapoptotic proteins bcl-xl or bcl-2. Oncogene T cell activation in mice lacking IL-2 receptor β. Science 1995;
2006 25: 2339–2348. 268: 1472.
1915. Su ZZ, Lebedeva IV, Sarkar D, et al. Melanoma differentiation 1934. Suzuki T, Tahara H, Narula S, et al. Viral interleukin 10 (IL-10),
associated gene-7, mda-7/IL-24, selectively induces growth sup- the human herpes virus 4 cellular IL-10 homologue, induces
pression, apoptosis and radiosensitization in malignant gliomas local anergy to allogeneic and syngeneic tumors. J Exp Med
in a p53-independent manner. Oncogene 2003; 22: 1164–1180. 1995; 182: 477.
1916. Su ZZ, Madiereddi MT, Lin JJ. Cancer growth suppressor gene 1935. Swain SL, McKenzie DT, Dutton RW, et al. The role of IL-2 and
mda-7 selectively induces apoptosis in human breast cancer IL-5: characterization of a distinct helper T cell subset that makes
cells and inhibits tumor growth in nude mice. Proc Natl Acad IL-4 and IL-5 (Th2) and requires priming before induction of
Sci USA 1998; 95: 14400–144405. lymphokine secretion. Immunol Rev 1988; 102: 77–106.
1917. Suda T, Murray R, Fischer M, et al. Tumor necrosis factor α and 1936. Swain SL, McKenzie DT, Weinberg AD, Hancock W.
P40 induce day 15 murine fetal thymocyte proliferation in com- Characterization of T helper 1 and 2 cell subsets in normal mice:
bination with IL-2. J Immunol 1990; 144: 1783–1787. helper T cells responsible for IL-4 and IL-5 production are pres-
1918. Sudo T, Nishikawa S, Ohno N, et al. Expression and function of ent as precursors that require priming before they develop into
interleukin 7 receptor on murine lymphocytes. Proc Natl Acad lymphokine secreting cells. J Immunol 1994; 141: 3445.
Sci USA 1993; 90: 9125. 1937. Swain SL. The activation and differentiation of T cells. The
1919. Sudo T, Suda J, Ogawa M, Ihle JN. Permissive role of interleukin-3 Immunologist 1995; 3: 209–211.
(IL-3) in proliferation and differentiation of multipotential 1938. Symons JA, Young PR, Duff GW. Soluble type II interleukin I
hemopoietic progenitors in culture. J Cell Physiol 1985; 124: 182. (IL-1) receptor binds and blocks processing of IL-1β precursor
266 Cytokines

and loses affinity for the IL-1 receptor antagonist. Proc Natl receptor superfamily genes and their constitutive and inducible
Acad Sci USA 1995; 92: 1714. expression in hematopoietic and non-hematopoietic cells. Gene
1939. Szabo SJ, Dighe AS, Gubler U, Murphy KM. Regulation of the 1997; 204: 35.
interleukin (IL)-12R β2 subunit expression in developing T 1960. Tan PL, Farmiloe S, Yeoman S, Watson JD. Expression of the
helper 1 (Th1) and Th2 cells. J Exp Med 1997; 185: 817–824. interleukin-6 gene in rheumatoid synovial fibroblasts. J
1940. Tabata Y, Chen W, Warrier MR, et al. Allergy-driven alternative Rheumatol 1990: 17; 1608.
splicing of IL-13 receptor α2 yields distinct membrane and 1961. Tan W, Huang W, Zhong O, et al. IL-17 receptor knockout mice
soluble forms. J Immunol 2006; 177: 7905–7912. have enhanced myelotoxicity and impaired hemopoietic recov-
1941. Tacchini-Cottier F, Vesin C, Redard M, Buurman Piquet PF. ery following γ irradiation. J Immunol 2006; 176: 6186–6193.
Role of TNFR1 and TNFR2 in TNF-induced platelet consump- 1962. Tanaka F, Hashimoto W, Okamura H, et al. Generation of potent
tion in mice. J Immunol 1998; 160: 6182–6186. and tumor-specific cytotoxic T lymphocytes by interleukin 18
1942. Taga K, Mostowski H, Tosato G. Human interleukin-10 can using dendritic cells and natural killer cells. Cancer Res 2000;
directly inhibit T cell growth. Blood 1993; 81: 2964. 60: 4838–4844.
1943. Taga T, Hibi M, Yamasaki K, et al. Interleukin 6 triggers the 1963. Taniguchi T, Matsui H, Fujita T, et al. Structure and expression
association of its receptor with a possible signal transducer of a cloned DNA for human interleukin-2. Nature 1983; 302:
gp130. Cell 1989; 58: 573–581. 305–310.
1944. Taga T, Kishimoto T. GP130 and the interleukin-6 family cytok- 1964. Tannenbaum CS, Tubbs R, Armstrong D, et al. The CXC
ines. Ann Rev Immunol 1997; 15: 797. chemokines IP-10 and Mig are necessary for IL-12 mediated
1945. Tagaya Y, Burton JD, Miyamoto Y, Waldmann TA. Identification regression of the mouse RENCA tumor. J Immunol 1998; 161:
of a novel receptor/signal transduction pathway for IL-15/T in 927–932.
mast cells. EMBO J 1996; 15: 4928–4939. 1965. Tartaglia LA, Rothe M, Hu Y-H, Goeddel DV. Tumor necrosis
1946. Tajima S, Tsuji K, Ebihara Y, et al. Analysis of interleukin-6 factor’s cytotoxicity is signaled by the p55TNF receptor. Cell
receptor and gp130 expressions and proliferative capability of 1993; 73: 213.
human CD34+ cells. J Exp Med 1996; 184: 1357–1364. 1966. Tartour E, Dorval T, Mosseri V, et al. Serum interleukin 6 and
1947. Takagi M, Nakahata T, Kubo T, et al. Stimulation of mouse con- C-reactive protein levels correlate with resistance to IL-2 ther-
nective tissue-type mast cells by hematopoietic stem cell factor, apy and poor survival in melanoma patients. Brit J Cancer 1994;
a ligand for the c-kit receptor. J Immunol 1992; 148: 3446. 69: 911–913.
1948. Takahashi C, Mittler RS, Vella AT. 4–1 BB is a bona fide CD8 T 1967. Tartour E, Fossiez F, Joyeux I, et al. Interleukin-17, a T cell
cell survival signal. J Immunol 1999; 162: 5037. derived cytokine, promotes tumorigenicity of human cervical
1949. Takahashi GW, Andrews DF, Lilly MB, et al. Effect of granulo- cancers in nude mice. Cancer Res 1999; 59: 3698.
cyte macrophage colony stimulating factor and interleukin 3 on 1968. Taskiran D, Stefanovic-Racic, Georgescu H, Evans CH. Nitric
interleukin 8 production by human neutrophils and monocytes. oxide mediates suppression of cartilage proteoglycan synthesis
Blood 1993; 81: 357. by IL-1. Biochem Biophys Res Commun 1994; 200: 142.
1950. Takahishi N, Udagawa N, Suda T. A new member of tumor 1969. Tatsumi T, Huang J, Gooding WE, et al. Intratumoral delivery of
necrosis factor ligand family, ODF/OPGL/TRANCE/RANKL, tumor cells engineered to secrete both interleukin (IL)-12 and
regulates osteoclast differentiation and function. Biochem IL-18 effectively treats local and distant disease in association with
Biophys Res Commun 1999; 256: 449. broad Tc1-type immunity. Cancer Res 2003; 63: 6378–6386.
1951. Takaki Y, Hayakawa Y, Nelson A, et al. IL-21 enhances tumor 1970. Tavernier J, Tuypens T, Plaetinck G, et al. Molecular basis of
rejection through an NKGD2-dependent mechanism. J Immunol the membrane-anchored and two soluble isoforms of the human
2005; 175: 2167–2173. interleukin-5 receptor α subunit. Proc Natl Acad Sci USA 1992;
1952. Takaoka A, Arai I, Sugimoto M, et al. Involvement of IL-31 on 89: 7041.
scratching behavior in NC/Nga mice with atopic-like dermati- 1971. Taylor CW, LeBlanc M, Fisher RI, et al. Phase II evaluation of
tis. Exp Dermatol 2006, 15 161–167. interleukin-4 in patients with non-Hodgkin’s lymphoma: a
1953. Takatsu T. Interleukin 5 and its receptor. Prog Growth Factor Southwest Oncology Group trial. Anticancer Drugs 2000; 11:
Res 1991; 3: 87. 695–700.
1954. Takeda K, Tsutsui H, Yoshimoto T, et al. Defective NK activ- 1972. Tchelingerian JL, Le Saux F, Jacque C. Identification and topog-
ity and Th1 response in IL-18-deficient mice. Immunity 1998; raphy of neuronal cell populations expressing TNFα and IL-1α
8: 383. in response to hippocampal lesion. J Neurosci Res 1996; 43:
1955. Takenawa J, Kaneko Y, Fukumoto M. Enhanced expression of 99–106.
interleukin-6 in primary human renal cell carcinomas. J Nat 1973. Te Velde AA, Huijbens RJF, Heije K, de Vries JE, Figor CG.
Cancer Inst 1991; 83: 1668–1672. Interleukin-4 inhibits the secretion of IL-1β, tumor necrosis fac-
1956. Takeuchi O, Hoshino K, Kawai T, et al. Differential roles of TLR2 tor α and IL-6 by human monocytes. Blood 1990; 76: 1392.
and TLR4 in recognition of Gram-negative and Gram-positive 1974. Te Velde AA, Klomp JPG, Yard BA, et al. Modulation of pheno-
bacterial cell wall components. Immunity 1999; 11: 443. typic and functional properties of human peripheral blood
1957. Tamada K, Shimozaki K, Chapoval AI, et al. LIGHT, a TNF- monocytes by IL-4. J Immunol 1988; 140: 1548–1554.
like molecule, costimulates T cell proliferation and it is required 1975. Teague RM, Sather BD, Sacks JA, et al. IL-15 rescues tolerant
for dendritic cell-mediated allogeneic T cell responses. J CD8+ T cells for use in adoptive immunotherapy of established
Immunol 2000; 164: 4105–4110. tumors. Nat Med 2006; 12: 335–341.
1958. Tan JT, Whitmire JK, Murali-Krishna K, et al. 4-1BB costimu- 1976. Teh H-S, Seebaran A, Teh S-J. TNF receptor 2-deficient
lation is required for protective anti-viral immunity after pep- CD8T cells are resistant to Fas/Fas ligand-induced cell death.
tide vaccination. J Immunol 2000; 164: 2320–2325. J Immunol 2000; 165: 4814–4821.
1959. Tan KB, Harrop J, Reddy M, et al. Characterization of a novel 1977. Temann UA, Geba GB, Rankin JA, Flavell RA. Expression of
TNF-like ligand and recently described TNF ligand and TNF interleukin 9 in the lungs of transgenic mice causes airway
Walter M. Lewko and Robert K. Oldham 267

inflammation, mast cell hyperplasia, and bronchial hyperre- involvement of the proteosome pathway. J Immunol 2000; 165:
sponsiveness. J Exp Med 1998; 188: 1307. 3959–3965.
1978. Tepler I, Elias L, Smith JW 2nd, Hussein M, et al. A randomized 1995. Tilden AB, Itoh K, Blach CM. Human lymphokine-activated
placebo-controlled trial of recombinant human interleukin-11 in killer (LAK) cells: identification of two types of effector cells. J
cancer patients with severe thrombocytopenia due to chemo- Immunol 1987; 138: 1068.
therapy. Blood 1996; 87: 3607–3614. 1996. Tilg H, Atkins MB, Dinarello CA Mier JW. Induction of circu-
1979. Tepper RI, Pattengale PK, Leder P. Murine interleukin 4 displays lating IL-10 by interleukin 1 and interleukin 2, but not interleu-
potent antitumor activity in vivo. Cell 1989; 57: 503–512. kin 6 immunotherapy. Cytokine 1995: 7: 734–739.
1980. Teunissen MB, Koomen CW, de Waal MR, Wierenga EA, Bos, 1997. Tilg H, Mier JW, Vogel W, et al. Induction of circulating IL-1
JD. Interleukin-17 and Interferon-γ synergize in the enhance- receptor antagonist by IFN treatment. J Immunol 1993; 150:
ment of proinflammatory cytokine production by human kerati- 4687–4692.
nocytes. J Invest Dermat 1998; 111: 645. 1998. Tilg H, Trehu E, Atkins MB, Dinarello CA, Mier JW.
1981. Theodore AC, Center DM, Nicoll J, et al. CD4 ligand IL-16 Interleukin-6 (IL-6) as an anti-inflammatory cytokine: induc-
inhibits the mixed lymphocyte reaction. J Immunol 1996; 157: tion of circulating IL-1 receptor antagonist and soluble tumor
1958–1964. necrosis factor receptor p55. Blood 1994; 83: 113.
1982. Thijs LC, Hack CE, Strack van Schijndel RJ, et al. Activation of 1999. Timeus F, Ricotti E, Crescenzio N, et al. FLT-3 and its ligand are
the complement system during immunotherapy with recombi- expressed in neural crest derived tumors and promote survival and
nant IL-2. J Immunol 1990; 144: 2419. its proliferation of their cell lines. Lab Invest 2001; 81: 1025–1037.
1983. Thoma-Uszynski S, Kiertscher SM, et al. Activation of toll-like 2000. Tittle TV, Weinberg AD, Steinkeler CN, Marziarz RT. Expression
receptor 2 on human dendritic cells triggers induction of IL-12, of the T cell activation antigen, OX-40, identifies alloreactive T
but not IL-10. J Immunol 2000; 165: 3804–3810. cells in acute graft-versus-host disease. Blood 1997; 89: 4652.
1984. Thomas B, Bird TA, Friend DJ, et al. Oncostatin M and leu- 2001. Tocher AW, Mita M, Meropol NJ, et al. Phase I pharmacokinetic
kemia inhibitory factor overlapping and different signals and biologic correlative study of mapatumumab, a fully human
through partially shared receptor complexes. J Biol Chem monpclonal antibody with agonist activity to tumor necrosis
1996; 271: 32635. factor-related apoptosis-inducing ligand receptor-1. J Clin
1985. Thomas WD, Hersey P. TNF-related apoptosis-inducing ligand Oncol 2007; 25: 1390–1395.
(TRAIL) induces apoptosis in Fas Ligand-resistant melanoma 2002. Toda A, Picarillo CA. Development and function of naturally
cells and mediates CD4 T cell killing of target cells. J Immunol occurring CD4+ CD25+ regulatory T cells. J Leukocyte Biol
1998; 161: 2195. 2006; 80: 458–470.
1986. Thomassen E, Bird TA, Renshaw RR, Kennedy KM, Sims JE. 2003. Tofaris GK, Patterson PH, Jessen KR, Mirsky R. Denervated
Binding of Interleukin-18 to the interleukin-1 receptor homolo- Schwann cells attract macrophages by secretion of leukemia
gous receptor IL-1Rrp 1 leads to activation of signaling path- inhibitory factor (LIF) and monocyte chemoattractant protein-1
ways similar to those used by interleukin-1. J Interferon in a process regulated by Interleukin-6 and LIF. J. Neurosci
Cytokine Res1998; 18: 1077. 2002; 22: 6696–6703.
1987. Thompsen MK, Larsen CG, Thompsen HK, et al. Recombinant 2004. Toi M, Bicknel R, Harris AL. Inhibition of colon and breast
human interleukin 8 is a potent activator of canine neutrophil carcinoma cell growth by interleukin-4. Cancer Res 1992; 52:
aggregation migration and leukotriene B4 biosynthesis. J Invest 275–279.
Dermatol 1991; 96: 260. 2005. Toi M, Harris AL, Bicknell R. Interleukin 4 is a potent mitogen
1988. Thompson BM, Mundy GR, Chambers TJ. Tumor necrosis fac- for capillary endothelium. Biochem Biophys Res Commun
tors α and β induce osteoblastic cells to stimulate osteoclastic 1991; 174: 1287–1293.
bone resorption. J Immunol 1987; 136: 775. 2006. Toksoz D, Williams D, Smith KA, et al. Expression of two dis-
1989. Thompson JA, Curti BD, Redman BG, et al. Interleukin-21 tinct DNAs of the steel gene (SCF) in deficient murine stromal
(IL-21): Tolerability and anti-tumor activity following two cell lines. Exp Hematol 1992; 20: 138–139.
5-day cycles in patients with stage IV melanoma or renal 2007. Toksoz D, Zsebo KM, Smith KA, et al. Support of human
cell carcinoma (RCC). Amer Soc Clin Oncol Proc 2006; hematopoiesis in the long term bone marrow cultures by murine
24: 2505. stromal cells selectively expressing the membrane bound and
1990. Thompson-Snipes LA, Dahr V, Bond MW, et al. Interleukin 10: secreted forms of the human analog of the steel gene product
a novel stimulatory factor for mast cells and their progenitors. stem cell factor. Proc Nat Acad Sci USA 1992; 89: 7350–7354.
J Exp Med 1991; 173: 507–510. 2008. Tomida MY, Yamamoto-Yamaguchi Y, Hozumi M. Purification
1991. Thorbecke GJ, Shah Leu CH, Kuruvilla AP, Hardison AM, of a factor inducing proliferation of mouse myeloid leukemia
Palladino MA. Involvement of endogenous tumor necrosis fac- M1 cells from conditioned medium of mouse fibroblast L929
tor α and transforming growth factor β during the induction of cells. J Biol Chem 1984; 259: 10978.
collagen type II arthritis in mice. Proc Natl Acad Sci USA 1992; 2009. Tomura M, Zhou X-Y, Maruo S, et al. A critical role for IL-18 in
89: 7375. the proliferation and activation of NK1.1+ CD3− cells. J
1992. Thornberry NA, Bull HG, Calaycay JR, et al. A novel heterodi- Immunol 1998; 160: 4738–4796.
meric cysteine protease is required for interleukin-1β process- 2010. Tong AW, Nemunaitis J, Su D. Intratumoral injection of INGN
ing in monocytes. Nature 1992; 356: 768. 241, a non-replicating adenovector expressing the melanoma-
1993. Tian J, Atkinson MA, Clare-Salzer M, et al. Nasal administra- differentiation associated gene-7 (mda-7/IL24): biologic outcome
tion of glutaminate decarboxylase (GAD65) peptides induces in advanced cancer patients. Mol Ther 2005; 11: 160–172.
Th2 responses and prevents murine insulin-dependent diabetes. 2011. Topalian SL, Solomon D, Avis FP, et al. Immunotherapy of
J Exp Med 1996; 183: 1561. patients with advanced cancer using tumor-infiltrating lympho-
1994. Tian Z, Shen X, Feng H, Gao B. IL-1β attenuates IFN-αβ- cytes and recombinant interleukin-2: a pilot study. J Clin Oncol
Induced antiviral activity and STAT1 activation in the liver: 1988; 6: 839.
268 Cytokines

2012. Topp MS, Koenigsmann M, Mire-Sluis A, et al. Recombinant 2030. Tsou CL, Haskell CA, Charo IF. Tumor necrosis factor-α-
human interleukin-4 inhibits growth of some human lung tumor converting enzyme mediates the inducible cleavage of fractalkine.
cell lines in vitro and in vivo. Blood 1993; 82: 2837–2844. J Biol Chem 2001; 276: 44622–44626.
2013. Topp MS, Papadimitriou CA, Eitelbach F, et al. Recombinant 2031. Tsuji K, Lyman SD, Sudo T, Clark SC, Ogawa M. Enhancement
human interleukin 4 has antiproliferative activity on human of murine hematopoiesis by synergistic interactions between
tumor cell lines derived from epithelial and nonepithelial steel factor (ligand for c-kit), interleukin-11, and other early act-
histologies. Cancer Res 1995; 55: 2173–2176. ing factors in culture. Blood 1992; 79: 2855.
2014. Torigoe K, Ushio S, Okura T, et al. Purification and character- 2032. Tsuji K, Nakahata T, Takagi M, et al. Effects of interleukin 3 and
ization of the human interleukin-18 receptor. J Biol Chem 1997; interleukin 4 on the development of connective tissue type mast
272: 25737. cells: interleukin 3 supports their survival and interleukin 4 trig-
2015. Tortorella C, Schulze-Koops H, Thomas R, et al. Expression gers and supports their proliferation synergistically with inter-
of CD45RB and CD27 identifies subsets of CD4+ memory T leukin 3. Blood 1990; 75: 421–427.
cells with different capacities to induce B cell differentiation. 2033. Tsuji K, Zsebo KM, Ogawa M. Murine mast cell colony forma-
J Immunol 1995; 155: 149. tion supported by IL-3, IL-4 and recombinant stem cell factor,
2016. Tosato G, Pike SE. Interferon β2/interleukin-6 is a co-stimulant ligand from c-kit. J Cell Physiol 1991; 148: 362.
for human T lymphocytes. J Immunol 1988; 141: 1556–1562. 2034. Tsunoda T, Tanimura H, Yamaue H, et al. The promotive effect
2017. Touw I, van Agthoven T, van Gurp R, et al. Interleukin 7 is a of interleukin-4 with interleukin-2 in the proliferation of tumor
growth factor of precursor B and T acute lymphoblastic leukemia. infiltrating lymphocytes from patients with malignant tumor.
Blood 1990; 75: 2097–2101. Biotherapy 1992; 4: 9–15.
2018. Tran NL, McDonough WS, Donohue PJ, et al. The human Fn14 2035. Tsutsui H, Nakanishi K, Matsui K, et al. IFN-γ-Inducing Factor
receptor gene is up regulated in migrating glioma cells in vitro Upregulates Fas ligand-mediated cytotoxic activity in murine
and overexpressed in advanced glial tumors. Am J Pathol 2003; natural killer cell clones. J Immunol 1996; 157: 3967–3973.
162: 1313–1321. 2036. Tuck AB, O’Malley FP, Singhal H, et al. Osteopontin expres-
2019. Tran NL, McDonough WS, Savitch BA, et al. The tumor necro- sion in a group of lymph node negative breast cancer patients.
sis factor-like weak inducer of apoptosis (TWEAK)-fibroblast Int J Cancer Predict Oncol 1998; 79: 502–508.
growth factor-inducible 14 (Fn14) signaling system regulates 2037. Turley JM, Fu T, Ruscetti FW, et al. Vitamin E succinate induces
glioma cell activation and BCL-XL/BCL-W expression. J Biol Fas-mediated apoptosis in estrogen receptor-negative human
Chem 2005; 280: 3483–3492. breast cancer cells. Cancer Res 1997; 57: 881–890.
2020. Trehu EG, Mier JW, Dubois JS, et al. Phase I trial of interleukin 2038. Turner JG, Rakhmilevich AL, Burdelya L, et al. Anti-CD40
2 in combination with the soluble tumor necrosis factor receptor antibody induces antitumor and antimetastatic effects: The role
p75 UgG chimera. Clin Cancer Res 1996; 2: 1341–1351. of NK cells. J Immunol 2001; 166: 89–94.
2021. Treisman J, Higuchi CM, Thompson JA, et al. Enhancement by 2039. Tuting T, Wilson CC, Martin DM, et al. Autologous human
interleukin 4 of interleukin 2 or antibody induced proliferation monocyte-derived dendritic cells genetically modified to
of lymphocytes from interleukin 2 treated cancer patients. express melanoma antigens elicit primary cytotoxic T cell
Cancer Res 1990; 50: 1160. responses in vitro: enhancement by cotransfection of genes
2022. Trepicchio WL, Ozawa M, Walters IB, et al. Interleukin-11 ther- encoding the Th1-biasing cytokines IL-12 and IFN-α. J
apy selectively downregulates type I cytokine proinflammatory Immunol 1998; 160: 1139–1147.
pathways in psoriasis lesions. J Clin Invest 1999; 104: 1527. 2040. Tuypens T, Plaetinct G, Baker E, et al. Organization and chro-
2023. Trepicchio WL, Wand L, Bozza M, Dorner AJ. IL-11 regulates mosomal localization of the human interleukin-5 receptor
macrophage effector function through inhibition of nuclear α-chain gene. Eur Cytokine Network 1992; 3: 451.
factor-κB. J Immunol 1997; 159: 5661–5670. 2041. Udagawa N, Takahashi N, Katagiri T, et al. Interleukin (IL)-6
2024. Trinchieri G, Matsumoto Kobayashi M, et al. Response of induction of osteoclast differentiation depends on IL-6 recep-
peripheral blood resting natural killer cells to interleukin-2. tors expressed on osteoblastic cells but not on osteoclast pro-
J Exp Med 1984; 160: 1147. genitors. J Exp Med 1995: 182: 1461.
2025. Trinchieri G. Interleukin-12: a cytokine at the interface of 2042. Ueda M, Imada K, Imura A, et al. Expression of functional
inflammation and immunity. Adv Immunol 1998; 70: 83. interleukin-21 receptor on adult T-cell leukemia cells. Br J
2026. Triozzi PL, Kin KA, Martin EW, et al. Phase I trial of escalating Haematol 2005; 128: 169–176.
doses of interleukin-1β in combination with a fixed dose of 2043. Ugai S, Shimozato O, Yu L, et al. Transduction of IL-21 and
interleukin-2. J Clin Oncol 1995; 13: 482–489. IL-23 genes in human pancreatic carcinoma cells produces nat-
2027. Tripp CS, Wolf SF, Unanue ER. Interleukin 12 and tumor necro- ural killer cell-dependent and cell-independent antitumor
sis factor α are costimulators of interferon γ production in effects. Cancer Gene Ther 2003; 10: 771–778.
severe combined immunodeficient mice with listeriosis and 2044. Urba WJ, Clark JW, Steis RG, et al. Intraperitoneal lymphokine-
interleukin 10 is a physiologic antagonist. Proc Natl Acad Sci activated killer cell/interleukin-2 therapy in patients with intra-
USA 1993; 90: 3725–3729. abdominal cancer: immunologic considerations. J Natl Cancer
2028. Truong MJ, Darcissac EC, Hermann E, et al. Interleukin-16 Inst 1989; 81: 602.
inhibits human immunodeficiency virus type entry and replica- 2045. Ushio S, Namba M, Okura T, et al. Cloning of the cDNA for
tion in macrophages and dendritic cells. J Virology 1999; 73: human IFN-γ-inducing factor in Escherichia coli, and studies on
7008–7013. the biologic activities of the protein. J Immunol 1996; 156: 4274.
2029. Tsai EY, Yie D, Thanos D, Goldfeld AE. Cell type specific 2046. Usui N, Mimnaugh EG, Sinha BK. A role for the interleukin 1
regulation of the human tumor necrosis factor α gene in B receptor in the synergistic antitumor effects of human interleu-
cells and T cells by NFATp and ATF-2/JUN. Mol Cell Biol kin 1α and etoposide against human melanoma cells. Cancer
1996; 16: 5232. Res 1991; 51: 769–774.
Walter M. Lewko and Robert K. Oldham 269

2047. Vaccarello L, Wang YL and Whiteside TL. Sustained outgrowth bodies in multiple myeloma: effects on soluble IL6 receptor and
of auto tumor reactive T lymphocytes from human ovarian soluble gp130. Leuk Lymphoma 1998; 31: 551–558.
carcinomas in the presence of tumor necrosis factor α and inter- 2066. Varghese J, Chattopopadhaya S, Sarin A. Inhibition of p38
leukin 2. Human Immunology 1990; 28: 216–227. Kinase reveals TNF-α-mediated, caspase-dependent, apoptotic
2048. Vagliani M, Rodolfo M, Cavallo F, et al. Interleukin 12 potenti- death pathway in a human myelomonocyte cell line. J Immunol
ates the curative effect of a vaccine based on interleukin 2-trans- 2001; 166: 6570–6577.
duced tumor cells. Cancer Res 1996; 56: 467. 2067. Vassali P. The pathophysiology of Tumor Necrosis Factor. Ann
2049. Valera LM, Ip MM. Tumor necrosis factor-α: multifunctional Rev Immunol 1992; 10: 411.
regulator of mammary gland development. Endocrinol 1996, 2068. Veiby OP, Jacobsen FW, Cui L, Lyman SD, Jacobsen SEW. The
137: 4915. flt-ligand promotes the survival of primitive hematopoietic pro-
2050. Van Antwerp DJ, Martin SJ, Kafri T, Green DR, Verma IM. genitor cells with myeloid as well as B lymphoid potential.
Suppression of TNF-α-induced apoptosis by NF-κB. Science J Immunol 1996; 157: 2953–2960.
1996; 274: 787–789. 2069. Veiby OP, Lyman SD, Jacobsen SEW. Combined signaling
2051. Van Arsdale TL, Van Arsdale SL, et al. Lymphotoxin-β receptor through interleukin-7 receptors and flt3 but not c-kit potently
signaling complex: role of tumor necrosis factor receptor asso- and selectively promotes B-cells commitment and differentiation
ciated factor 3 recruitment in cell death and activation of nuclear from uncommitted murine bone marrow progenitor cells. Blood
factor kappa B. Proc Natl Acad Sci USA 1997; 94: 2460–2465. 1996; 88: 1256.
2052. Van Damme J, Opdenakker G, et al. Identification of the human 2070. Veldhoen M, Hocking RJ, Atkins CJ, et al. TGFβ in the context
26 kD protein, interferon β2 (IFN-β2), as a B cell hybridoma/ of an inflammatory cytokine milieu supports de novo differen-
plasmacytoma growth factor induced by interleukin 1 and tumor tiation of IL-17-producing cells. Immunity 2006; 24: 179–189.
necrosis factor. J Exp Med 1987; 165: 914. 2071. Vellenga E, Esselink MT, Straaten J, et al. The supportive effects
2053. Van Damme J, Schaafsma MR, Fibbe WE, et al. Simultaneous of IL-7 on eosinophil progenitors from human bone marrow
production of interleukin 6, interferon β2 and colony stimulat- cells can be blocked by anti-IL-5. J Immunol 1992; 149: 2992.
ing activity by fibroblasts after viral and bacterial infection. Eur 2072. Verschraegen CF, Kudelka AP, Termrungruanglert W, et al.
J Immunol 1989; 19: 163. Effects of interleukin-1α on ovarian carcinoma in patients with
2054. Van De Griend RJ, Krimpen BA, Ranfeltap CPM and Bolhuis recurrent disease. Eur J Cancer 1996; 32A: 1609–1611.
RH. Rapidly expanded natural killer cell clones have strong 2073. Vetto JT, Lum S, Morris A, Sciotte M, et al. Presence of the T
antitumor cell activity and have surface phenotype of either T, cell activation marker OX-40 on tumor infiltrating lymphocytes
non-T, or null cells. J Immunol 1984; 132: 3185. and draining lymph node cells from patients with melanoma and
2055. Van den Berg WB, Joosten LA, Helsen M, Van de Loo FA. head and neck cancers. Am J Surg 1997; 174: 258.
Amelioration of established collagen-induced arthritis with 2074. Vidal-Vanaclocha F, Amezaga C, Asumendi A, Kaplanski G,
anti-IL-1 treatment. Clin Exp Immunol 1994; 95: 237. Dinarello CA. Interleukin 1 receptor blockade reduces the num-
2056. Van der Burg SH, Piersma SJ, de Jong A, et al. Association of ber and size of murine B16 melanoma hepatic metastases.
cervical cancer with the presence of CD4+ regulatory T cells Cancer Res 1994; 54: 2267.
specific for human papillomavirus antigens. Proc Natl Acad Sci 2075. Vidal-Vanaclocha F, Mendoza L, Telleria N. Clinical and exper-
USA 2007; 104: 12087–12092. imental approaches to the pathophysiology of interleukin-18 in
2057. Van Gameren MM, Willemse PH, Mulder NH, et al. Effects of cancer progression. Cancer Metastasis Rev 2006: 9:
recombinant human Interleukin-6 in cancer patients: a phase 1480–1486.
I-II trial. Blood 1994; 84: 1431–1441. 2076. Viera P, de Waal Malefyt R, Dang M-N, et al. Isolation and
2058. Van Kooten C, Gaillard C, et al. B cells regulate expression of expression of human cytokine synthesis inhibitory factor (CSIF/
CD40 ligand on activated T cells by lowering the mRNA level IL-10) cDNA clones: homology to Epstein Barr virus open
and through the release of soluble CD40. E J Immunol 1994; reading frame BCRFI. Proc Nat Acad Sci USA 1991; 88:
24: 787. 1172–1176.
2059. van Leeuen EM, Gamadia LE, Baars PA, et al. Proliferation 2077. Villers D, Dao T, Nguyen J-M, et al. Increased plasma levels of
requirements of cytomegalovirus-specific effector-type human human interleukin for DA1.a cells/leukemia inhibitory factor in
CD8+-T cells. J Immunol 2002; 169: 5838–5842. sepsis correlate with shock and poor prognosis. J Infect Dis
2060. Van Lier RAW, Borst J, et al. Tissue distribution and biochemi- 1995; 171: 232.
cal functional properties of Tp55 (CD27), a novel T cell differ- 2078. Vinay DS, Choi BK, Bae JS, et al. CD137-deficient mice have
entiation antigen. J Immunol 1987; 139: 1589. reduced NK/NKT cell numbers and function, are resistant to
2061. Van Oers HS, Pals ST, Evers LM, et al. Expression and release lipopolysaccharide-induced shock syndromes and have lower
of CD27 in human B-cell malignancies. Blood 1993; 82: 3430. IL-4 responses. J Immunol 2004; 173: 4218–4229.
2062. Van Parijs L, Buickians A, Ibragimov A, Alt F, Willerford D, 2079. Vink A, Renauld JC, Warnier G, Van Snick J. Interleukin-9
Abbas A. Functional responses and apoptosis of CD25 (IL-2 R stimulates in vitro growth of mouse thymic lymphomas. Eur
α)-deficient T cells expressing a transgenic antigen receptor. J J Immunol 1993; 23: 1134.
Immunol 1997; 158: 3738. 2080. Vissinga CS, Fatur-Saunders DJ and Takei F. Dual role of IL-7
2063. Van Snick J, Goethals A, Renauld J-C, et al. Cloning and char- in the growth and differentiation of immature thymocytes.
acterization of cDNA for a new mouse T cell growth factor Experimental Hematology 1992; 20: 998–1003.
(P40). J Exp Med 1989; 169: 363–368. 2081. Vistintin A, Mazzoni A, Spitzer JH, et al. Regulation of Toll-like
2064. Van Snick J. Interleukin 6: an overview. Ann Rev Immunol receptors in human monocytes and dendritic cells. J Immunol
1990; 8: 253–280. 2001; 166: 249–255.
2065. Van Zaanen HC, Lokhorst HM, Aarden LA, et al. Blocking 2082. Vitetta ES, Ohara J, Myers CD, et al. Serological biochemical
interleukin-6 activity with chimeric anti-IL-6 monoclonal anti- and functional identity of B cell stimulatory factor-1 and B cell
270 Cytokines

differentiation factor for IgG1. J Exp Med 1985; 162: 2101. Wang G, Tschoi M, Spolski R, et al. In vivo antitumor activity
1726–1731. of interleukin 21 mediated by natural killer cells. Cancer Res
2083. Volpert OV, Fong T, Koch AE, et al. Inhibition of angiogenesis 2003; 63: 9016–9022.
by interleukin-4. J Exp Med 1998; 88: 1039. 2102. Wang J, Homer RJ, Chen Q, Elias JA. Endogenous and exoge-
2084. Von Freeden-Jeffrey U, Viera P, Lucian LA, et al. Lymphopenia nous IL-6 inhibit aeroallerogen-induced Th2 inflammation. J
in interleukin (IL)-7 gene deleted mice identifies IL-7 and a Immunol 2000; 165: 4051–4056.
nonredundant cytokine. J Exp Med 1995; 181: 1519–1526. 2103. Wang J, Homer RJ, Hong L, et al. IL-11 selectively inhibits
2085. Von Freeden-Jeffry R, Solvason N, Howard M, Murray R. The aeroallergen-induced pulmonary eosinophilia and Th2 cytokine
earliest T lineage-committed cells depend on interleukin-7 for production. J Immunol 2000; 165: 2222–2231.
Bcl-2 expression and normal cell cycle progression. Immunity 2104. Wang M, Tan Z, Zhang R, et al. Interleukin 24 (MDA-7/MOB-5)
1997; 7: 147. signals through two heterodimeric receptors IL22R1/IL-20R2
2086. Von Leoprechting A, van der Bruggen P, Pahl HL, Aruffo A, and IL-20R1/IL-20R2. J Biol Chem 2002; 277: 7341–7347.
Simon JC. Stimulation of CD40 on immunogenic human malig- 2105. Wang P, Wu P, Cheewatrakoolpong B, et al. Selective inhibition
nant melanoma augments their cytotoxic T lymphocyte-medi- of IL-5 receptor α-chain gene transcription by IL-5, IL-3 and
ated lysis and induces apoptosis. Cancer Res 1999; 59: 1287. granulocyte-macrophage colony stimulating factor in human
2087. Vonderheide RH, Dutcher JP, Anderson JE, et al. Phase I study blood eosinophils. J Immunol 1998; 160: 4427–4432.
of recombinant human CD40 ligand in cancer patients. J Clin 2106. Wang R, Zhang L, Zhang X, et al. Differential regulation of the
Oncol 2001; 19: 3280–3287. expression of CD95 ligand, receptor activator of nuclear
2088. Wahl AF, Klussman K, Thompson JD, et al. The anti-CD30 factor-κB ligand (RANKL), TNF-related apoptosis-inducing
monoclonal antibody SGN-30 promotes growth arrest and DNA ligand (TRAIL), and TNF-α during T cell activation. J Immunol
fragmentation in vitro and affects antitumor activity in models 2001; 166: 1983–1990.
of Hodgkin’s lymphoma. Cancer Res 2002; 62: 3736–3742. 2107. Wang R, Zhang L, Zhang X, Yin D, Mufson RA, Shi Y. Protein
2089. Walczak H, Degli-Esposti MA, Johnson RS, et al. TRAIL-R2: a kinase C regulates FAS (CD95/Apo1) expression. J Immunol
novel apoptosis mediating receptor for TRAIL. EMBO J 1997; 1998; 161: 2201.
16: 5386. 2108. Wang X, Chen M, Wandinger KP, Williams G, Dhib-Jalbut S.
2090. Walczak H, Miller RE, Arail K, et al. Tumoricidal activity of IFN-β-1b inhibits IL-12 production in peripheral blood mono-
tumor necrosis factor-related apoptosis-inducing ligand in vivo. nuclear cells in an IL-10-dependent mechanism: Relevance to
Nat Med 1999; 5: 157. IFN-β-1b therapeutic effects in multiple sclerosis. J Immunol
2091. Waldmann T, Tagaya Y. The multifaceted regulation of interleu- 2000; 165: 548–557.
kin-15 expression and the role of this cytokine in NK cell 2109. Wang Y-H, Angkasekwinai P, Lu N, et al. IL-25 augments type
differentiation and host cell response to intracellular pathogens. 2 immune responses by enhancing the expansion and functions
Ann Rev Immunol 1999; 17: 19. of TSLP-DC-activated Th2 memory cells. J Exp Med 2007;
2092. Wallace PM, MacMaster JF, Rouleau KA, et al. Regulation of 204: 1837–1847.
inflammatory responses by oncostatin M. J Immunol 1999; 2110. Wang Y, Wang J, Sun Y, Wu Q, Fu Y-X. Complementary effects
162: 5547. of TNF and lymphotoxin on the formation of germinal center
2093. Wallach D, Holtmann H, Aderka D, et al. Mechanisms which and follicular dendritic cells. J Immunol 2001; 166: 330–337.
take part in the regulation of the response to tumor necrosis fac- 2111. Wang YL, Lusheng S, Kanbaul A, et al. Lymphocytes infiltrating
tor. Lymphokine Res 1989; 8: 359. human ovarian tumors: Synergy between tumor necrosis factor α
2094. Wallach D, Varfolomezv EE, Malinin NL, et al. Tumor necrosis and interleukin 2 in the generation of CD8+ effectors from tumor-
factor and Fas signaling mechanisms. Ann Rev Immunol 1999; infiltrating lymphocytes. Cancer Res 1989; 49: 5979–5985.
17: 331. 2112. Wang YO, Ugai S, Shimazato O, et al. Induction of systemic
2095. Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution of immunity by expression of interleukin-23 in murine colon car-
cellular immunity against cytomegalovirus in recipients of allo- cinoma cells. Int J Cancer 2003; 105: 820–824.
geneic bone marrow by transfer of T-cell clones from the donor. 2113. Wang Z, Karras JG, Howard RG, Rothstein TL. Induction of
N Eng J Med 1995; 333: 1038–1044. bcl-X by CD40 engagement rescues sIg-induced apoptosis in
2096. Walz A, Peveri P, Aschauer H, Baggiolini M. Purification and murine B cells. J Immunol 1995; 155: 3722–3725.
amino acid sequencing of NAF, a novel neutrophil activating 2114. Wanidworanum C and Strober W. Predominant role of tumor
factor produced by monocytes. Biochem Biophys Res Commun necrosis factor-α in human monocyte IL-10 synthesis. J
1987; 149: 755. Immunol 1993; 151: 6853.
2097. Wang A, Braun SE, Sonpavde G, Cornetta K. Antileukemic 2115. Wantanabe M, Ueno Y, Yajima T, et al. Interleukin-7 is pro-
activity of Flt3 ligand in murine leukemia. Cancer Res 2000; duced by human intestinal epithelial cells and regulates the pro-
60: 1895–1900. liferation of intestinal mucosal lymphocytes. J Clin Invest 1995;
2098. Wang C-Y, Cusack JC, Liu R, Baldwin AS. Control of induc- 95: 2945.
ible chemoresistance: enhanced anti-tumor therapy through 2116. Ware CF, Crowe PD, Grayson MH, Androlewicz MJ, Browning
increased apoptosis by inhibition of NF-κB. Nat Med 1999; JL. Expression of surface lymphotoxin and tumor necrosis fac-
5: 412. tor on activated T, B, and natural killer cells. J Immunol 1992;
2099. Wang C-Y, Mayo MW, Baldwin AS. TNF- and Cancer therapy- 149: 3881–3888.
induced apoptosis: potentiation by inhibition of NF-κB. Science 2117. Waring PM, Waring LJ, Billington T, Metcalf D. Leukemia
1996; 274: 784–787. inhibitory factor protects against lethal Escherichia coli septic
2100. Wang C-Y, Mayo MW, Korneluk RG, Goeddel DV, Baldwin shock in mice. Proc Natl Acad Sci USA 1995; 92: 1337.
AS. NF-κB antiapoptosis: induction of TRAF1 and TRAF2 and 2118. Waring PM, Waring LJ, Metcalf D. Circulating leukemia inhibi-
the c-IAP1 and c-IAP2 to suppress caspace-8 activation. Science tory factor levels correlate with disease severity in meningococ-
1998; 281: 1680. cemia. J Infect Dis 1994; 170: 1224.
Walter M. Lewko and Robert K. Oldham 271

2119. Waring PM, Wycherly K, Cary D, Nicola N, Metcalf D. 2137. Weiss T, Grell M, Siemienski K, et al. TNFR80-dependent
Leukemia inhibitory factor levels are elevated in septic enhancement of TNFR60-induced cell death is mediated by
shock and various inflammatory body fluids. J Clin Invest TNFR-associated factor 2 and is specific for TNFR60. J
1992; 90: 2031. Immunol 1998; 161: 3136–3142.
2120. Warren HS, Kinnear BF, Phillips JH, Lanier LL. Production of 2138. Wiethe C, Dittmar K, Doan D, et al. Provision of 4-1BB ligand
IL-5 by Human NK cells and regulation of IL-5 secretion by enhances effector and memory CTL responses generated by
IL-4, IL-10, and IL-12. J Immunol 1995; 154: 5144–5152. immunization with dendritic cells expressing a human tumor-
2121. Warringa RAJ, Koenderman L, Kok PTM, et al. Modulation associated antigen. J Immunol 2003; 170: 2919–2922.
and induction of eosinophil chemotaxis by granulocyte mac- 2139. Welch PA, Namen AE, Goodwin RG, et al. Human IL-7: a novel
rophage colony stimulating factor and interleukin 3. Blood T cell growth factor. J Immunol 1989; 143(11): 3562–3567.
1991; 77: 2694. 2140. Weller PF, Rand TH, Barrett T, et al. Accessory cell function of
2122. Wassink L, Veira PL, Smits HH, et al. ICOS expression by acti- eosinophils: HLA DR dependent MHC restricted antigen
vated human Th cells is enhanced by IL-14 and IL-23: Increased presentation and interleukin 1α formation. J Immunol 1993;
ICOS expression enhances the effector function of both TH1 150: 2554.
and Th2 cells. J Immunol 2004; 173: 1779–1786. 2141. Welte K, Chang YW, Mertelsmann R, et al. Purification of
2123. Waxman AB, Einarrson O, Seres T, et al. Targeted lung expres- human interleukin-2 to apparent homogeneity and its molecular
sion of interleukin-11 enhances murine tolerance of 100% oxy- heterogeneity. J Exp Med 1982; 156: 435.
gen and diminishes hyperoxia-induced DNA fragmentation. J 2142. Weng Y, Siciliano SJ, Waldburger KE, et al. Binding and func-
Clin Invest 1998; 101: 1970. tional properties of recombinant and endogenous CXCR3
2124. Weaver A, Chang J, Wrigley E, et al. Randomized comparison of chemokine receptors. J Biol Chem 1998; 273: 18288.
progenitor-cell mobilization using chemotherapy, stem cell fac- 2143. Wershil BK, Tsai M, Geissler EN, Zsebo KM, Galli SJ. The rat
tor, and filgrastrim or chemotherapy plus filgrastim alone in c-kit ligand, stem cell factor, induces c-kit-dependent mouse
patients with ovarian cancer. J Clin Oncol 1998; 16: 2601–2612. mast cell activation in vivo. Evidence that signaling through the
2125. Weber GF, Gaertner PC, Erl W, et al. IL-22-mediated tumor c-kit receptor can induce expression of cellular function. J Exp
growth reduction correlates with inhibition of ERK1/2 and AKT Med 1992; 175: 245.
phosphorylation and induction of cell cycle arrest in the G2-M 2144. Wesche H, Korherr C, Kracht M, et al. The interleukin-1 acces-
phase. J Immunol 2006; 177: 8266–8272. sory protein (IL-1RAcP) is essential for IL-1-induced activation
2126. Weber J, Yang JC, Topalian SL, et al. Phase I trial of subcutane- of interleukin-1 receptor associated kinase (IRAK) and stress-
ous interleukin 6 in patients with advanced cancer. J Clin Oncol activated protein kinases (SAP Kinases). J Biol Chem 1997;
1993; 11: 499–506. 272: 7727.
2127. Wegman TG, Lin H, Guilbert L, Mosmann TR. Bidirectional 2145. Wesche H, Wenzel WJ, Shillinglaw W, Li S, Cao Z. MyD88: an
cytokine interactions in the maternal–fetal relationship: is suc- adaptor that recruits IRAK to the IL-1 receptor complex.
cessful pregnancy a Th2 phenomenon? Immunol Today 1993; Immunity 1997; 7: 837.
18: 479. 2146. West WH, Tauer KW, Yannelli JR, et al. Constant infusion
2128. Wei CC, Chen WY, Wang YC. Detection of IL-20 and its recep- recombinant interleukin-2 in adoptive immunotherapy of
tor on psoriatic skin. Clin Immunol 2005; 117: 65–72. advanced cancer. N Engl J Med 1987; 316: 898.
2129. Wei L, Lawrence A, Elias KM, et al. IL-21 is produced by Th17 2147. Westendorf JJ, Jelinek DF. Growth regulatory pathways in
cells and drives IL-17 production in a STAT3-dependent man- myeloma. Eidence for autocrine oncostatin M expression. J
ner. J Biol Chem 2007; 282: 34605–34610. Immunol 1996; 157: 3081–3088.
2130. Wei SJ, Liu JH, Blanchard DK, Djeu JY. Induction of IL-8 gene 2148. Wetzler M, Kurzrock R, Estrov Z, et al. Altered levels of
expression in polymorphonuclear leukocytes by recombinant interleukin-1β and interleukin-1 receptor antagonist in chronic
IL-2. J Immunol 1994; 142: 244. myelogenous leukemia: clinical and prognostic correlates.
2131. Wei X-Q, Leung BP, Arthur HML, McInnes IB, Liew FY. Blood 1994; 84: 3142–3147.
Reduced incidence and severity of arthritis in mice lacking 2149. Whitehead RP, Fleming T, MacDonald JS, et al. A phase II trial
IL-18. J Immunol 2001; 166: 517–521. of recombinant tumor necrosis factor in patients with metastatic
2132. Weikowski MT, Leach MW, Evans EW, et al. Ubiquitous colorectal adenocarcinoma: a Southwest Oncology Group
transgenic expression of the IL-23 subunit p19 induces mul- Study. J Biol Response Modifiers 1990; 9: 588–591.
tiorgan inflammation, runting, infertility and premature death. 2150. Whitehead RP, Unger JM, Goodwin JW, et al. Phase II trial
J Immunol 2001; 166: 7563–7570. of recombinant human interleukin-4 in patients with dissemi-
2133. Weil-Hillman G, Fisch P, Prieve AF, et al. Lymphokine-activated nated malignant melanoma: a Southwest Oncology Group
killer activity induced by in vivo interleukin 2 therapy: predomi- study. J Immunother 1998; 21: 440–446.
nant role for lymphocytes with increased expression of CD2 and 2151. Whitman SP, Archer KJ, Feng L, et al. Absence of the wild-type
Leu19 antigens but negative expression of CD16 antigens. allele predicts poor prognosis in adult de novo acute myeloid
Cancer Res 1989; 49: 3680. leukemia with normal cytogenetics and the internal tandem
2134. Weinberg AD, Bourdette DN, Sullivan TJ, et al. Selective deple- duplication of Flt3: a cancer and leukemia group B study.
tion of myelin-reactive T cells with the anti-Ox-40 antibody Cancer Res 2001; 61: 7233–7239.
ameliorates autoimmune encephalitis. Nat Med 1996; 2: 183. 2152. Widmer M. Acres R, Sassenfeld H, Grabstein K. Regulation of
2135. Weinberg K, Parkman R. Severe combined immunodeficiency cytolytic cell populations from human peripheral blood by B cell
due to a specific defect in the production of interleukin-2. N Eng stimulatory factor 1 (interleukin 4). J Exp Med 1987; 166: 1447.
J Med 1990; 322: 1718. 2153. Widmer MB, Morrissey PJ, Namen AE, et al. Interleukin 7
2136. Weiss GR, Margolin KA, Sznol M, et al. Phase II study of the stimulates the growth of fetal thymic precursors of cytolytic
continuous infusion of Interleukin 6 for metastatic renal cell cells: induction of effector function by interleukin 2 and inhibi-
carcinoma. J Immunother 1995; 18: 52–56. tion by interleukin 4. Int Immunol 1990; 2: 1055–1061.
272 Cytokines

2154. Wierda WG, Cantwell ML, Woods SJ, Rassenti LZ, Prussak 2172. Williams NS, Klem J, Puzanov IJ, et al. Natural killer cell dif-
CE, Kipps TJ. CD40-ligand (CD154) gene therapy for chronic ferentiation: insights from knockout and transgenic mouse mod-
lymphocytic leukemia. Blood 2000; 96: 2917–2924. els and in vivo systems. Immunol Rev 1998; 165: 47–61.
2155. Wiethe C, Dittmar K, Doan T, et al. Enhanced effector and 2173. Williams RO, Martinova-Mutafchieva L, Feldman M, Maini
memory CTL responses generated by incorporation of receptor RN. Evaluation of TNF-α and IL-1 blockade in collagen-induced
activator of NF-κB (RANK)/RANK ligand costimulatory mol- arthritis and comparison with combined anti-TNF-α/anti-CD4
ecules into dendritic cell immunogens expressing a human therapy. J Immunol 2000; 165: 7240–7245.
tumor-specific antigen. J Immunol 2003; 171: 4121–4130. 2174. Williamson BD, Carswell EA, Rubin BY, et al. Human tumor
2156. Wiggington JM, Lee JK, Wiltrout TA, et al. Synergistic engage- necrosis factor produced by human B cell lines. Synergistic
ment of an ineffective endogenous anti-tumor response and cytotoxic interaction with human interferon γ. Proc Nat Acad
induction of IFN-γ and Fas-ligand-dependent tumor eradication Sci USA 1983; 80: 5397–5401.
by combined administration of IL-18 and IL-2. J Immunol 2175. Wills-Karp M, Luyimbazi J, Xu X, et al. Interleukin-13: central
2002; 169: 4467–4474. mediator of allergic asthma. Science 1998; 282: 2258–2261.
2157. Wigginton JM, Kuhns DB, Back TC, et al. Interleukin 12 primes 2176. Wirtz S, Tubbe I, Galle PR, et al. Protection from lethal septic
macrophages for nitric oxide production in vivo and restores peritonitis by neutralizing the biological function of interleu-
depressed nitric oxide production by macrophages from tumor kin-27. J Exp Med 2006; 203: 1875–1881.
bearing mice: implications for antitumor activity of interleukin 2177. Wischhusen J, Jung G, Radovanovic I, et al. Identification of
12 and/or interleukin 2. Cancer Res 1996; 56: 1131. CD70-mediated apoptosis of immune effector cells as a novel
2158. Wigginton JM, Park J-W, Gruys ME, et al. Complete regression immune escape pathway of human glioblastoma. Cancer Res
of established spontaneous mammary carcinoma and the thera- 2002; 62: 2592–2599.
peutic prevention of genetically programmed neoplastic transi- 2178. Witowski J, Pawlaczyk K, Breborowicz A, et al. IL-17 stimu-
tion by IL-12/pulse IL-2: Induction of local T cell infiltration, lates intraperitoneal neutrophil infiltration through the release of
Fas/Fas ligand gene expression and mammary epithelial apop- GROα chemokine from mesothelial cells. J Immunol 2000;
tosis. J Immunol 2001; 166: 1156–1168. 165: 5814–5821.
2159. Wilcox RA, Chapoval AI, Gorski KS, et al. Expression of func- 2179. Wittig B, Marten A, Dorbic T, Weineck S, et al. Therapeutic
tional CD137 by murine dendritic cells. J Immunol 2002; 168: vaccination against metastatic melanoma by expression-modu-
4262–4267. lated and immunomodified autologous tumor cells: a first clini-
2160. Wilcox RA, Tamada K, Strome FE, et al. Signaling through NK cal phaseI/II trial. Human Gene Therapy 2001; 12: 267–278.
cell-associated CD137 promotes both helper functions for 2180. Woerly G, Roger N, Loiseau S, Capron M. Expression of Th1
CD8+ cytolytic T cells and responsiveness to IL-2 but not and Th2 immunoregulatory cytokines by human eosinophils. Int
cytolytic activity. J Immunol 2002; 169: 4230–4236. Arch Allergy Immunol 1999; 118: 95.
2161. Wilcoxen SC, Kirkman E, Dowdell KC, Stohlman SA. Gender- 2181. Wogensen L, Huang X, Sarvetnik N. Leukocyte extravasation
dependent IL-12 secretion is regulated by IL-10. J Immunol into the pancreatic tissue in transgenic mice expressing interleu-
2000; 164: 6237–6243. kin 10 in the islets of Langerhans. J Exp Med 1993; 178: 175.
2162. Wild JS, Sigounas A, Sur N, Siddiqui MS, et al. IFN-γ inducing 2182. Wogensen LD, Mandrup-Poulsen T, Markholst H, et al.
factor (IL-18) increases allergic sensitization, serum IgE, Th2 Interleukin-1 potentiates glucose stimulated insulin release in the
cytokines and air way eosinophilia in a mouse model of allergic isolated perfused pancreas. Acta Endocrinol 1988; 117: 302.
asthma. J Immunol 2000; 164: 2701. 2183. Wogun AW, van Gils FC, Wagemaker G. Flow cytometric
2163. Wiley JA, Geha R, Harmsen AG. Exogenous CD40 ligand detection of receptors for interleukin-6 on bone marrow and
induces a pulmonary inflammation response. J Immunol 1997; peripheral blood cells of human and rhesus monkeys. Blood
158: 2932–2938. 1993; 81: 2036–2043.
2164. Wiley SR, Cassiano L, Lofton T, et al. A novel TNF receptor 2184. Wolf D, Hallmann R, Sass G, et al. TNF-α-induced expression
family member binds TWEAK and is implicated in angiogene- of adhesion molecules in the liver is under control of TNFRI.
sis. Immunity 2001; 15: 857–846. Relevance for Con A-induced Hepatitis. J Immunol 2001; 166:
2165. Wiley SR, Goodwin RG, Smith CA. Reverse signaling via 1300–1307.
CD30 ligand. J Immunol 1996; 157: 3635–3639. 2185. Wolf SF, Sieburth D, Sypek J. Interleukin 12: A key modulator
2166. Wiley SR, Schooley K, Smolak PJ, et al. Identification and char- of immune response. Stem Cells 1994; 12: 154–168.
acterization of a new member of the TNF family that induces 2186. Wolf SF, Temple PA, Kobayashi M, et al. Cloning of cDNA for
apoptosis. Immunity 1995; 3: 673. natural killer cell stimulatory factor, a heterodimeric cytokine
2167. Willems F, Marchant A, Delville JP, et al. Interleukin-10 inhibits with multiple biologic effects on T and natural killer cells. J
B7 and intracellular adhesion molecule-1 expression on human Immunol 1991; 146: 3074–3081.
monocytes. Eur J Immunol 1994; 24: 1007. 2187. Wolk K, Kunz S, Asadullah K, et al. Immune cells as sources
2168. Willerford DM, Chen J, Ferry FA, Davidson L, Ma A, Alt FW. and targets of the IL-10 family members. J Immunol 2002; 168:
Interleukin-2 receptor α chain regulates the size and content of 5397–5402.
the peripheral lymphoid compartment. Immunity 1995; 3: 521. 2188. Wolk K, Sabat R. Interleukin-22: a novel T- and NK-derived
2169. Williams DE, Eisenman J, Baird A, et al. Identification of a cytokine that regulates the biology of tissue cells. Cytokine
ligand for the c-kit proto-oncogene. Cell 1990; 63: 167–174. Growth Factor Rev 2006; 17: 267–380.
2170. Williams DE, Morrissey PJ, Mochizuki DY, et al. T cell growth 2189. Wolk K, Witte E, Hoffmann U, et al. IL-22 induces lipopolysac-
factor P40 promotes the proliferation of myeloid cell lines and charide-binding protein in hepatocytes: a potential systemic role
enhances erythroid burst formation by normal murine bone mar- of IL-22 in disease. J Immunol 2007; 178: 5973–5981.
row cells in vitro. Blood 1990; 76: 906–911. 2190. Wolk K, Witte K, Witte E, et al. Maturing dendritic cells are an
2171. Williams N, Eger RR, Jackson HN, et al. Two factor require- important source of IL-29 and IL-20 that may cooperatively
ment for murine megakaryocyte colony formation. J Cell increase the innate immunity of keratinocytes. J Leukocyte
Physiol 1982; 110: 101. Biol 2008; 83: 000–000.
Walter M. Lewko and Robert K. Oldham 273

2191. Wolk K, Witte E, Wallace E, et al. Il-22 regulates the expression 2210. Xia D, Li F, Xiang J. Engineered fusion hybrid vaccine of
of genes responsible for antimicrobial defense, cellular differen- IL-18 gene-modified tumor cells and dendritic cells induces
tialtion, and mobility in keratinocytes: a potential role in psoria- enhanced antitumor immunity. Cancer Biother Radiopharm
sis. Eur J Immunol 2006; 36: 1309–1323. 2004; 19: 322–330.
2192. Wong BR, Josien R, Choi Y. TRANCE is a TNF family member 2211. Xiang J. Expression of costimulatory 4-1BB ligand induces sig-
that regulates dendritic cell and osteoclast function. J Leukocyte nificant tumor regression and protective immunity. Cancer
Biol 1999; 65: 715. Biother Radiopharm 1999; 14: 353–361.
2193. Wong BR, Josien R, Lee SY, et al. TRANCE (Tumor necrosis 2212. Xie MH, Aggarwal S, Ho WH, et al. Interleukin (IL)-22, a novel
factor [TNF]-related activation-induced cytokine), a new TNF human cytokine that signals through the interferon related pro-
family member predominanctly expressed in T cells, is a den- teins CRF2 and IL-22R. J Biol Chem 2000; 275:
dritic cell-specific survival factor. J Exp Med 1997; 186: 2075. 31335–31339.
2194. Wong BR, Rho J, Arron J, et al. TRANCE is a novel ligand of 2213. Xin X, Cai Y, Matsumoto K, Agui T. Endothelin-induced inter-
the tumor necrosis factor receptor family that activates c-Jun leukin-6 production by rat aortic endothelial cells. Endocrinology
N-terminal kinase in T cells. J Biol Chem 1997; 272: 25190. 1995; 136: 132.
2195. Wong CK, Cheung PF, Ip WK and Lam CW. Interleukin-25- 2214. Xu D, Chan WL, Leung BP, et al. Selective expression and
induced chemokines and interleukin-6 release from eosinophils functions of interleukin 18 receptor on T helper (Th) type I but
is mediated by p38 mitogen-activated protein kinase, c-Jun not Th2 cells. J Exp Med 1998; 188: 1485.
N-terminal kinase, and nuclear factor-κB. Am J Respir Cell Mol 2215. Xu DP, Sauter BV, Huang TG, et al. The systemic administra-
Biol 2005; 33: 186–194. tion of Ig-4-1BB ligand in combination with IL-12 gene transfer
2196. Wood N, Bourque K, Donaldson DD, et al. IL-21 effects on eradicates hepatic colon carcinoma. Gene Ther 2005; 12:
human IgE production in response to IL-4 or IL-13. Cell 1526–1533.
Immunol 2004; 231: 135–145. 2216. Xu H, Okamoto A, Ichikawa J, et al. TWEAK/Fn14 interaction
2197. Wood N, Whitters MJ, Jacobson BA, et al. Enhanced interleu- stimulates human bronchial epithelial cells to produce IL-8 and
kin (IL)-13 responses in mice lacking IL-13 receptor α2. J Exp GM-CSF. Biochem Biophys Res Commun 2004; 318:
Med 2003; 197: 703–709. 422–427.
2198. Woods JM, Katschke KJ, Tokuhira M, et al. Reduction of 2217. Xu Z, Buckley MJ, Evans CH, Agarwal S. Cyclic tensile strain
inflammatory cytokines and prostaglandin E2 by IL-13 gene acts as an antagonist of IL-1β actions in chondrocytes. J
therapy in rheumatoid arthritis synovium. J Immunol 2000; 165: Immunol 2000; 165: 453–460.
2755–2763. 2218. Yacoub A, Mitchell C, Lebedeva IV. mda-7 inhibits growth and
2199. Worth LL, Jaffe N, Benjamin RS, et al. Phase II study of recom- enhances radiosensitivity of glioma cells in vitro via JNK sig-
binant interleukin 1α and etoposide with relapsed osteosarcoma. naling. Cancer Biol Ther 2003; 347–252.
Clin Cancer Res 1997; 3: 1721–1720. 2219. Yaden Y, Kuang WS, Yang-Feng T, et al. Human protooncogene
2200. Wu C-Y, Ferrante J, Gately K, et al. Characterization of IL-12Rβ c kit, a new cell surface receptor tyrosine kinase for an unidenti-
chain-deficient mice. J Immunol 1997; 159: 1658. fied ligand. EMBO J 1987; 6: 3341–3351.
2201. Wu C-Y, Wang X, Gadina M, et al. IL-12 receptor β2 2220. Yamada K, Takane N, Otabe S, et al. Pancreatic β-cell-selective
(IL-12Rβ2)-deficient mice are defective in IL-12-mediated sig- production of tumor necrosis factor induced by interleukin-1.
naling despite the presence of high affinity IL-12 binding. J Diabetes 1993; 42: 1026–1031.
Immunol 2000; 165: 6221–6228. 2221. Yamaguchi K, Kinosaki M, Goto M, et al. Characterization of
2202. Wu C, Demeure C, Kiniwa M, et al. IL-12 induces production structural domains of human osteoclastogenesis inhibitory fac-
of IFN-γ by neonatal human CD4 T cells. J Immunol 1993; 151: tor. J Biol Chem 1998; 273: 5117.
1938–1949. 2222. Yamagushi Y, Suda T, Suda J, et al. Purified Interleukin 5 sup-
2203. Wu DMH, Zhang Y, Parada NA, et al. Processing and release of ports the terminal differentiation and proliferation of purified
IL-16 from CD4+ but not CD8+ T cells is activation dependent. eosinophil precursors. J Exp Med 1988; 167: 43.
J Immunol 1999; 162: 1287–1293. 2223. Yamamoto M, Yoshizaki K, Kishimoto T, Ito H. IL-6 is required
2204. Wu K, Bi Y, Sun K, et al. IL-10-producing type 1 regulatory T for the development of Th1 cell-mediated murine colitis. J
cells and allergy. Cellular Molec Immunol 2007; 4: 269–275. Immunol 2000, 164: 4878–4882.
2205. Wu Q, Sun Y, Wang J, Lin X, et al. Signal via lymphotoxin-β on 2224. Yamanaka R, Honma J, Tsuchiya N, et al. Tumor lysate and
bone marrow stromal cells is required for an early checkpoint of IL-18 loaded dendritic cells elicits TH1 response, tumor-spe-
NK cell development. J Immunol 2001; 166: 1684–1689. cific CD8+ cytotoxic T cells in patients with malignant glioma.
2206. Wu Q, Wang Y, Wang J, et al. The requirement for membrane J Neurooncol 2005; 72: 107–113.
lymphotoxin for the presence of dendritic cells in lymphoid tis- 2225. Yamasaki K, Taga T, Hirata Y, et al. Cloning and expression of
sues. J Exp Med 1999; 190: 629. the human interleukin 6 (BSF-2/IFNβ2) receptor. Science 1988;
2207. Wu S, Boyer CM, Whittaker RS, et al. Tumor necrosis factor as 241: 825–828.
an autocrine and paracrine growth factor for ovarian cancer: 2226. Yang FC, Agematsu K, Nakazawa T, et al. CD27/CD70 interac-
Monokine induction of tumor cell proliferation and tumor tion directly induces natural killer cell killing activity.
necrosis factor α production. Cancer Res 1993; 53: 1939. Immunology 1996; 88: 289–293.
2208. Wurster AL, Rodgers VL, Satoskar AR, et al. Interleukin 21 is a 2227. Yang Y-C, Ciarletta AB, Temple PA, et al. Human IL-3 (multi-
T helper (Th) cytokine that specifically inhibits the differentia- CSF): identification by expression cloning of a novel hematopoi-
tion of naive Th cells into interferon γ producing Th1 cells. J etic growth factor related to murine IL-3. Cell 1986; 47: 3–10.
Exp Med 2002; 196: 967–977. 2228. Yang Y, Mercep M, Ware CF, Ashwell JD. FAS and activation-
2209. Wuthrich RP, Jenkins TA, Snyder TL. Regulation of cytokine induced ligand mediated apoptosis of T cells hybridomas: inhi-
stimulated vascular cell adhesion molecule-1 expression in bition of Fas ligand expression by retinoic acid and
renal tubular epithelium cells. Transplantation 1993; 55: 172. glucocorticoids. J Exp Med1995; 181: 1673.
274 Cytokines

2229. Yang YC, Riccaciardi S, Ciarletta A, et al. Expression cloning 2247. Yokota T, Coffman RL, Hagiwara H, et al. Isolation and charac-
of a cDNA encoding a novel human hematopoietic growth fac- terization of lymphokine cDNA clones encoding mouse and
tor: Human homologue of murine T cell growth factor P40. human IgA-enhancing factor eosinophil colony-stimulating
Blood 1989; 74: 1880–1884. activities: relationship to interleukin 5. Proc Natl Sci (USA)
2230. Yao L, Pike SE, Setsuda J, Parekh J, et al. Effective targeting of 1987; 84: 7388–7392.
tumor vasculature by angiogenesis inhibitors vasostatin and 2248. Yokota T, Lee F, Rennick D, et al. Isolation and characterization
interleukin-12. Blood 2000; 96: 1900–1905. of a mouse cDNA clone that expresses mast cell growth factor
2231. Yao Z, Fanslow WC, Seldin MF, et al. Herpes virus saimiri activity in monkey cells. Proc Natl Acad Sci USA 1984; 81:
encodes a new cytokine IL-17 which binds a novel cytokine 1070–1074.
receptor. Immunity 1995; 3: 811. 2249. Yokota T, Otsuka T, Mosmann T, et al. Isolation and character-
2232. Yao Z, Painter SL, Fanslow FC, et al. Interleukin-17: a novel ization of a human interleukin cDNA clone, homologous to
cytokine derived from T cells. J Immunol 1995; 155: 5483. mouse B-cell stimulatory factor 1, that expressed B cell and T
2233. Yao Z, Spriggs MK, Derry JMJ, et al. Molecular characteriza- cell stimulating activities. Proc Natl Acad Sci USA 1986; 83:
tion of the human interleukin (IL)-17 receptor. Cytokine 1997; 5894–5898.
9: 794. 2250. Yoneda J, Kuniyasu H, Crispens MA, et al. Expression of angio-
2234. Yasamura S, Lin WC, Weidemann E, et al. Expression of inter- genesis-related genes and progression of human ovarian carci-
leukin 2 receptors on human carcinoma cell lines and tumor nomas in nude mice. J Natl Cancer Inst 1998; 90: 447.
growth inhibition by interleukin-2. Int J Cancer 1994; 59: 2251. Yoshimoto T, Bendelac A, Watson C, Hu-Li J, Paul WE. Role of
225–234. NK1.1+ T cells in a Th2 response and in immunoglobulin E
2235. Yasuda H, Shima N, Nakagawa N, et al. Osteoclast differentia- production. Science 1995; 270: 1845.
tion factor is a ligand for osteoprotegerin/osteoclastogenesis- 2252. Yoshimoto T, Paul WE. CD4 pos NK1.1 pos T cells promptly
inhibitory factor and is identical to TRANCE/RANKL. Proc produce interleukin-4 in response to in vivo challenge with anti-
Natl Acad Sci USA 1998; 95: 3597. CD3. J Exp Med 1994; 179: 1285.
2236. Ye P. Rodriguez FH, Kanaly S, et al. Requirement of Interleukin 2253. Yoshimoto T, Takeda K, Takara T, et al. IL-12 upregulates IL-18
17 receptor signaling for lung CXC chemokine and granulocyte receptor expression on T cells, Th1 cells, and B cells: synergism
colony-stimulating factor expression, neutrophil recruitment with IL-18 for IFN-γ production. J Immunol 1998; 161: 3400.
and host defense. J Exp Med 2001; 194: 519. 2254. Yoshimoto T, Tsutsui H, Tominaga K, et al. IL-18, although
2237. Ye Q, Fraser CC, Gao W. Modulation of LIGHT-HVEM costim- antiallergic when administered with IL-12, stimulates IL-4 and
ulation prolongs cardiac allograft survival. J Exp Med 2002; histamine release by basophils. Proc Natl Acad Sci USA 1999;
195: 795–800. 96: 1362.
2238. Yee NS, Langen H, Besmer P. Mechanism of kit ligand, phorbol 2255. Yoshimoto T, Wang W-R, Yoneto T, et al. Role of IL-16 in
ester and calcium-induced down-regulation of c-kit receptors in delayed type hypersensitivity reaction. Blood 2000; 95:
mast cells. J Biol Chem 1993; 268: 14189. 2869–2874.
2239. Yeh K-Y, McAdam AJ, Pulaski BA, et al. IL-3 enhances both 2256. Yoshimura A, Ohkubo T, Niguchi T, et al. A novel cytokine-in-
presentation of exogenous particulate antigen in association ducible gene CIS encodes an SH2-containing protein that binds
with Class I major histocompatibility antigen and the generation to tyrosine-phoshorylated interleukin-3 and erythropoietin
of primary tumor specific cytotolytic T lymphocytes. J Immunol receptors. EMBO J 1995; 14: 2816.
1998; 160: 5773–5780. 2257. Yoshimura T, Matsushima K, Tanaka S, et al. Purification of a
2240. Yellin MJ, Sippel K, Inghirami G, et al. CD40 molecules induce human monocyte-derived neutrophil chemotactic factor that has
down-regulation and endocytosis of T cell surface T cell-B cell peptide sequence similarity to other host defense cytokines.
activating molecules/CD40L: potential role in regulating helper Proc Natl Acad Sci USA 1987; 84: 9233–9237.
effector functions. J Immunol 1994; 152: 598. 2258. Yoshimura T, Takeda A, Hamano S, et al. Two-sided roles of
2241. Yellin MJ, Winniloff S, Fortune SM, et al. Ligation of CD40 on IL-27: Induction of Th1 differentiation on Naïve CD4+ T cells
fibroblasts induces CD54 (ICAM-1) and CD106 (VCAM-1) versus suppression of proinflammatory cytokine production
upregulation and IL-6 production and proliferation. J Leukocyte including IL-23-induced IL-17 on activated CD4+ T cells par-
Biol 1995; 58: 209. tially through STAT3-dependent mechanism. J Immunol 2006;
2242. Yen D, Cheung J, Scheerens H, et al. IL-23 is essential for T 177: 5377–5385.
cell-mediated colitis and promotes inflammation via IL-17 and 2259. Younes A, Consoli U, Snell V, et al. CD30 ligand is expressed in
IL-6. J Clin Invest 2006; 116: 1310–1316. lymphoma patients with CD30+ tumors. J Clin Oncol 1997; 15:
2243. Yiling L, Granelli-Piperno A, Bjorndahl JM, Phillips CA, 3355–3362.
Trevillyan JM. CD28-induced T-cell activation. J Immunol 2260. Younes A, Consoli U, Zhao S, et al. CD30 ligand is expressed in
1992; 149: 24. normal and malignant human B lymphocytes. Brit J Haematol
2244. Yin T, Schendel P, Yang Y-C. Enhancement of in vitro and in 1996; 93: 569–571.
vivo antigen-specific antibody responses by IL-11. J Exp Med 2261. Yousefi S, Hemmann S, Weber M, et al. IL-8 is expressed by
1992; 175: 211. human peripheral blood eosinophils. Evidence for increased
2245. Yin T, Tsang ML, Yang YC. JAK1 kinase forms complexes with secretion in asthma. J Immunol 1995; 154: 5481–5490.
interleukin-4 receptor and 4PS/insulin receptor substrate-1-like 2262. Yssel H, de Waal Malfyt R, Roncarolo MG, et al. IL-10 is pro-
protein and is activated by interleukin-4 and interleukin-9 in T duced by subsets of human CD4+ T cell clones and peripheral
lymphocytes. J Biol Chem 1994; 269: 26614–26617. blood T cells. J Immunol 1992; 149: 2378.
2246. Yokota T, Arai N, DeVries J, et al. Molecular biology of inter- 2263. Yssel H, Schneider PV, Lanier LL. Interleukin 7 specifically
leukin 4 and interleukin 5 genes and biology of their products induces the B7/BB1 antigen on human cord blood and periph-
that stimulate B cells, T cells and hematopoietic cells. Immunol eral blood T cells and T cell clones. Int Immunol 1993; 5:
Rev 1988; 102: 137–187. 753–759.
Walter M. Lewko and Robert K. Oldham 275

2264. Yu JS, Wei MX, Chjiocca A, et al. Treatment of glioma with by normal human keratinocytes and a human squamous cell car-
engineered interleukin 4 secreting cells. Cancer Res 1993; 53: cinoma cell line (HSC-1). J Dermatological Sci 1994; 7: 24–31.
3125–3128. 2282. Zhang S, Kaplan MH. The p38 mitogen-activated protein kinase
2265. Yu KY, Kwon B, Ni J, et al. A newly identified member of tumor is required for IL-12-induced IFN-γ expression. J Immunol
necrosis factor receptor superfamily (TR6) suppresses LIGHT- 2000; 165: 1374–1380.
mediated apoptosis. J Biol Chem 1999; 274: 13733–13746. 2283. Zhang X, Franco A, Myers K, et al. Relation of TRAIL receptor
2266. Yuan Q, Gurish MF, Friend DS, Austen KF, Boyce JA. and FLIP expression to TRAIL-induced apoptosis of melanoma.
Generation of a novel stem cell factor-dependent mast cell pro- Cancer Res 1999; 59: 2747.
genitor. J Immunol 1998; 161: 5143–5146. 2284. Zhang X, Sun S, Hwang I, Tough D, Sprent J. Potent and selec-
2267. Yuan X, Hu J, Belladona ML, et al. Interleukin-23-expressing tive stimulation of memory phenotype CD8+ T cells in vivo by
bone marrow-derived neural stem cell-like cells exhibit antitu- IL-15. Immunity 1998; 8: 591.
mor activity against intracranial glioma. Cancer Res 2006; 66: 2285. Zhang XG, Gu JJ, Yasukawa K, et al. Ciliary neurotrophic fac-
2630–2638. tor, interleukin 11, leukemia inhibitory factor, and oncostatin M
2268. Yumoto K, Ishijima M, Rittling SR, et al. Osteopontin defi- are growth factors for human myeloma cell lines using the inter-
ciency protects joints against destruction in anti-type II collagen leukin 6 transducer gp130. J Exp Med 1994; 179: 1337–1342.
antibody-induced arthritis in mice. Proc Natl Acad Sci USA 2286. Zhang Y, Center DM, Wu DMH, et al. Processing and activa-
2002; 99: 4556–4561. tion of pro-interleukin-16 by caspase-3. J Biol Chem 1998;
2269. Yun TJ, Chaudary PM, Shu GL, et al. OPG/FDCR-1, a TNF 273: 1144.
family member, is expressed in lymphoid cells and is upregu- 2287. Zhang Y, Kornfeld H, Cruickshank WW, et al. Nuclear translo-
lated by ligating CD40. J Immunol 1998; 161: 6113. cation of the N-terminal prodomain of interleukin-16. J Biol
2270. Yun TJ, Tallquist MD, Aicher A, et al. Osteoprotegerin, a crucial Chem 2001; 276: 1299–1303.
regulator of bone metabolism, also regulates B cell develop- 2288. Zheng L, Fisher G, Miller RE, et al. Induction of apoptosis in
ment and function. J Immunol 2001; 166: 1482–1491. mature T cells by tumor necrosis factor. Nature 1995; 377: 348.
2271. Zachariae C, Ternowitz T, Larsen CG, Nielsen V, Thestrup- 2289. Zheng M, Bocangel D, Doneske B, et al. Human interleukin 24
Pedersen K. Epidermal lymphocye chemotactic factor specifi- (MDA-7/IL-24) protein kills breast cancer cells via the IL-20
cally attracts OKT4-positive lymphocytes. Arch Dermatol Res receptor and is antagonized by IL-10. Cancer Immunol
1988; 280: 354–357. Immunother 2007; 56: 205–215.
2272. Zamir O, Hasselgren PO, O’Brien W. Thompson RC Fischer JE. 2290. Zheng T, Nathanson M, Elias JA. Histamine augments cytokine
Muscle protein breakdown during endotoxemia in rats and after stimulated interleukin-11 production by human lung fibroblasts.
treatment with interleukin-1 receptor antagonist (IL-1Ra). Ann J Immunol 1994; 153: 4742.
Surg 1992; 216: 381. 2291. Zheng Y, Danilenko DM, Valdez P, et al. Interleukin-22, a TH17
2273. Zang XG, Gu JJ, Lu ZY, Yasukawa GD, et al. Ciliary neu- cytokine, mediates IL-23-induced dermal inflammation and
rotrohpic factor, interleukin 11, leukemia inhibitory factor and acanthosis. Nature 2007; 445: 648–651.
oncostatin M (OSM) are growth factors for human myeloma 2292. Zheng Y, Ouaaz F, Bruzzo P, et al. NF-κB RelA (p65) is essen-
cell lines using the interleukin 6 signal transducer gp130. J Exp tial for TNF-α-induced Fas expression but dispensable for both
Med 1994; 179: 1337. TCR-induced expression and activation-induced cell death. J
2274. Zarling JM, Shoyab H, Marquardt H, et al. Oncostatin M: a Immunol 2001; 166: 4949–4857.
growth regulator produced by differentiated histiocyte lym- 2293. Zhou L-J, Tedder TF. A distinct pattern of cytokine expression
phoma cells. Proc Natl Acad Sci USA 1986; 83: 9739. by human CD83+ blood dendritic cells. Blood 1995; 86: 3295.
2275. Zeng R, Spolski R, Finkelstein SE, et al. Synergy of IL-21 and 2294. Zhou P, Devadas K, Tewari D, Jegorow A, Notkins AL. Processing,
IL-15 in regulating CD8+ T cell expansion and function. J Exp secretion and anti-HIV-1 activity of IL-16 with or without a signal
Med 2005; 201: 139–148. peptide in CD4+ T cells. J Immunol 1999; 163: 906–912.
2276. Zerafa N, Westwood JA, Cretney E, et al. TRAIL deficiency 2295. Zhou P, Goldstein S, Devadas K, Tewari D, Notkins AL. Human
accelerates hematological malignancies. J Immunol 2005; 175: CD4+ cells transfected with IL-16 cDNA are resistant to HIV-1
5586–5590. infection: inhibition of mRNA expression. Nat Med 1997; 3:
2277. Zetterstrom M, Sundgren-Anderson AK, Ostlund P, et al. 659–664.
Delineation of the proinflammatory cytokine cascade in fever 2296. Zhu M, Oishi K, Lee SC, Patterson PH. Studies using leukemia
induction. Ann NY Acad Sci 1998; 856; 48–52. inhibitory factor (LIF) knockout mice and a LIF adenovirus vec-
2278. Zhai Y, Hsu TL, Yu GL, Kwon BS, et al. LIGHT, a novel ligand tor demonstrate a key anti-inflammatory role for this cytokine in
for lymphotoxin β receptor and TR2/HVEM induces apoptosis cutaneous inflammation. J Immunol 2001; 166: 2049–2054.
and suppresses tumor formation via gene transfer. J Clin Invest 2297. Zilocchi C, Stopociaro A, Chiodoni C, et al. Interferon-γ rejec-
1998; 102: 1142–1151. tion of interleukin 12-transduced carcinoma cells required CD+
2279. Zhang GX, Gran B, Yu S, et al. Induction of experimental auto- T cells and granulocyte-macrophage colony stimulating factor.
immune encephalomyelitis in IL-12Rβ2-deficient mice: IL-12 J Exp Med 1998; 188: 133.
is not required in the pathogenesis of inflammatory demyelina- 2298. Zimmermann N, Hogan SP, Mishra A, et al. Murine eotaxin-2: A
tion in the central nervous system. J Immunol 2003; 170: constitutive eosinophil chemokine induced by allergen challenge
2153–2160. and IL-4 overexpression. J Immunol 2000; 165: 5839–5846.
2280. Zhang H, Merchant MS, Chua KS, et al. Tumor expression of 2299. Zitrogel L, Tahara H, Robbins PD, et al. Cancer immunotherapy
4-1BB ligand sustains tumor lytic T cells. Cancer Biol Ther of established tumors with IL-12. Effective delivery by geneti-
2003; 2: 576–586. cally engineered fibroblasts. J Immunol 1995; 155: 1393–1403.
2281. Zhang JZ, Maruyama K, Iwatsuki K, Kaneko F. Regulatory 2300. Zitzmann K, Band S, Bachs S, et al. Novel interferon-lambdas
effects of 1,25-dihydroxyvitamin D3 and a novel Vitamin D3 induce proliferative effects in neuroendocrine tumor cells.
analog MC903 on secretion of interleukin 1α (IL-1α) and IL-8 Biochem Biophys Res Commun 2006; 344: 1334–1341.
276 Cytokines

2301. Zoltnick A, Ransom J, Frank G, et al. Interleukin 4 is a growth interleukin; ILA, induced by lymphocyte activation
factor for activated thymocytes: possible role in T cell ontogeny. (4-1BB); IL-1Ra, IL-1 receptor antagonist; IL-1RAcP,
Proc Nat Acad Sci USA 1987; 84: 3856.
2302. Zoltnik A, Fisher M, Roehm N, and Zipori D. Evidence for
IL-1 receptor accessory protein; IL-1RAK, IL-1 receptor
effects of (B cell stimulatory factor 1) on macrophages: enhance- associated kinase; IL-1Rrp, IL-1 receptor related
ment of antigen presenting ability of bone marrow derived mac- protein (IL-18Rα); IL-TIF, interleukin 10-related T
rophages. J Immunol 1987; 138: 4275. cell-derived inducible factor (IL-22); IP-10, interferon-
2303. Zou J, Presky DW, Wu CY, Grubler U. Differential associations
induced protein-10; JAK, Janus kinase; -L (as a suffix),
between the cytoplasmic regions of the interleukin-12 receptor
subunits β1 and β2 and the Jak kinases. J Biol Chem 1997; 272: ligand; LAK, lymphokine activated killer; LIF, leuke-
6073–6077. mia inhibitory factor; LIGHT, lymphotoxin-like induc-
2304. Zsebo KM, Williams DA, Geissler EN, et al. Stem cell factor is ible protein that competes with glycoprotein D for
encoded at the S1 locus of the mouse and is the ligand for the binding herpesvirus entry mediator on T cells; LIT,
c-kit tyrosine kinase receptor. Cell 1990; 63: 312–224.
lymphocyte inhibitor of TRAIL; LN, lymphnode; LPS,
2305. Zsebo KM, Wypych J, McNiece IK, et al. Identification charac-
terization and biological characterization of hematopoietic stem lipopolysaccharide; LT-α, lymphotoxin-α (TNF-β),
cell factor form Buffalo rat liver conditioned medium. Cell LT-β, lymphotoxin-β; Macro, macrophage; MAPK,
1990; 63: 195–210. mitogen-activated protein kinase; MCP, macrophage
2306. Zsebo KM, Yuschenoff VN, Schiffer S, et al. Vascular endothe- chemoattractant protein; M-CSF, macrophage colony
lial cells and granulopoiesis: interleukin-1 stimulates release of
G-CSF and GM-CSF. Blood 1988; 71: 99.
stimulating factor; MHC, major histocompatibility
2307. Zuany-Amorim C, Ruffie C, Haile S, et al. Requirement for γ/δ T complex; Mig, monokine induced by IFN-γ; MIP,
cells in allergic airway inflammation. Science 1998; 280: 1265. macrophage inflammatory protein; MMP, matrix
2308. Zubiaga AM, Munoz E, Huber BT. IL-2 and IL-4 selectively metalloprotease; Mono, monocyte; neut, neutrophil;
rescue Th cell subsets from glucocorticoid-induced apoptosis. NF-, nuclear (transcription) factor as in NF-κB; NGF,
J Immunol 1992; 149: 107–112.
2309. Zucali JR, Dinarello CA, Oblon DJ, et al. Interleukin 1 stimu-
nerve growth factor; NK, natural killer cell; NNT, novel
lates fibroblasts to produce granulocyte-macrophage colony neurotrophin; nTreg, natural regulatory T cell; OSM,
stimulating activity and prostaglandin E2. J Clin Invest 1986; oncostatin M; PAMP, pathogen-associated molecular
77: 1857. patterns; OPG, osteoprotegerin; OPN, osteopontin;
2310. Zurawski G, de Vries JE. Interleukin 13, an interleukin 4 like
PBL, peripheral blood lymphocyte; PDGF, platelet
cytokine that acts on monocytes and B cells but not T cells.
Immunol Today 1994; 15: 19–29. derived growth factor; PG, prostaglandin; p, protein w/
mw (x10−3); PMA, phorbol myristate acetate; -R (as a
suffix), receptor; RANKL, receptor activator of NF-κB
ligand (TRANCE); RANTES, regulated on activation,
Abbreviations normal T cell expressed and secreted; rhu- (as a prefix)
ADC; antibody dependent cytotoxicity; AICD, activa- recombinant human; SAPK, stress activated protein
tion-induced cell death; AP, antigen presentation; baso, kinase; s- (as a prefix) soluble (not membrane bound);
basophil; BCGF, B cell growth factor; BCSF, B cell SCF, stem cell factor; SCID, severe combined immuno-
stimulatory factor; BM, bone marrow; CFR, cytokine deficiency disease; SDF, stromal derived factor; SLE,
family receptor; CFU, colony forming unit; CNTF, cili- systemic lupus erythematosus; SOCS, suppressors of
ary neurotrophic factor; CTL, cytotoxic T lymphocyte; cytokine signaling; STAT, signal transducers and activa-
DC, dendritic cell; DcR, decoy receptor; DR, death tors of transcription; Stk, stem cell tyrosine kinase
receptor; EAE, experimental autoimmune encephalitis; (flt-3), TACE, TNF-α converting enzyme; TCR, T cell
EGF, epidermal growth factor; ELR, glutamic acid-leu- receptor; TDAC, tumor derived activated T cell (TIL);
cine-arginine (a chemokine designation); eos, eosino- TGFβ, transforming growth factor-β; Th, helper T cell;
phil; EPO, erythropoietin; ETA-1, early T lymphocyte TIL, tumor infiltrating lymphocyte; TIMP, tissue inhibi-
activation protein (OPN); FISP, IL-4-induced secreted tor of metalloprotease; TLR, toll-like receptor; TNF,
protein; γc, common gamma chain (IL-2Rγ chain); tumor necrosis factor; TNFR, TNF receptor; TPO,
G-CSF, granulocyte colony stimulating factor; GM-CSF, thrombopoietin; TRADD, TNF Receptor 1-associated
granulocyte macrophage colony stimulating factor; gp, death domain; TRAF, TNF receptor-associated factor;
glycoprotein w/mw (x 10−3); HLA, human leukocyte TRAIL, TNF-related apoptosis-inducing ligand;
antigen; HMW-BCGF, high molec wt-B cell growth TRANCE, TNF-related activation induced cytokine
factor (IL-14); HVEM, Herpes virus entry mediator (RANKL); Treg, regulatory T cell; TWEAK, Tumor
(LIGHT R); IAP, inhibitor of apoptosis; ICAM, inter- necrosis factor-like weak inducer of apoptosis; tyk,
cellular adhesion molecule; ICE, IL-1β cleavage enzyme tyrosine kinase; VCAM; vascular cell adhesion molecule;
= caspase 1; IFN, interferon; Ig, immunoglobulin; IL, VLS, vascular leak syndrome.
9 Interferons: therapy for cancer
DAVID GOLDSTEIN, ROBERT JONES, RICHARD V. SMALLEY, AND ERNEST C. BORDEN

Stimulate the phagocytes. Drugs are a delusion. Find the germ of study of the role of interferons as anti neoplastic agents.
the disease; prepare from it a suitable antitoxin; inject it three times A review of these interferon studies remains a valuable
a day, a quarter of an hour before meals and what is the result? The
phagocytes are stimulated; they devour the disease and the patient
resource of guidelines on how to optimise clinical studies
recovers-unless, of course, he’s too far gone. in malignancy using novel non-traditional agents.
– Sir Ralph Bloomfield Bonnington in The Doctors Dilemma
George Bernard Shaw, 1902.

Isaacs and Lindemann, in England, first characterised


interferon (IFN) in 1957 and coined the word to signify
Nomenclature
a protein, elaborated by virus-infected cells, that func- Alpha interferon is produced following viral stimula-
tions to prevent their infection by a second virus [108]. tion. There are at least 14 subtypes of alpha interferon,
However, difficulties with chemical isolation and char- each immunologically related but differing by a number
acterization led to great skepticism about the molecule’s of amino acids [290].
existence; indeed, “the scientific community dubbed the Several nomenclature systems exist for identifying
discovery ‘imaginon’ ” [192]. these subtypes. The two commonly used, but unrelated,
Time and effort have proven Isaacs and Lindemann systems use either lettering A, B, C, D, etc., or number-
right. Interferons are now a well-characterized group of ing 1, 2, 3, etc. to define the subtypes. A newer pro-
proteins. They are a large family of four major immuno- posal, approved by the Nomenclature Committee of the
logical types, alpha, beta, gamma, and omega [199]. International Society for Interferon Research, has been
The alpha and beta interferons were collectively known recently published [199]. A corresponding proposal for
as the type I interferons while gamma interferon was nomenclature of the interferon-gene-induced proteins
referred to as type II interferon. There are multiple natu- exists.
ral alpha interferon molecules and one natural beta Three natural alpha interferon pharmaceutical prod-
interferon molecule. These type I interferons share a ucts are currently available for use in patients in various
common receptor which is present on nearly all cells in countries in the world. The original Finnish Red Cross
the body. The alpha interferons, originally derived from partially purified material developed by Cantell and sub-
leucocytes, and beta interferon, originally derived from sequently also produced by others from the buffy coat of
fibroblasts, are actually secreted by nearly all mamma- peripheral blood is available as Finnferon® in some
lian cells. Even though they share a common receptor, European countries. A highly purified natural alpha inter-
their activation pathway must differ since the transfec- feron produced by virally stimulated human lymphoma
tion of the human type I receptor gene into murine cells (Namalva) cells has been produced in pharmacologic
allows alpha but not beta expression [266]. Gamma quantity by Burroughs Wellcome Co (now Glaxo
interferon, on the other hand, is produced by T lympho- Wellcome) and is known by the trade name Wellferon®.
cytes upon specific antigen recognition. It is marketed in Japan, Canada, Mexico and Europe
All interferon molecules have antiviral and antiprolif- but not the United States for treatment of hairy cell leu-
erative capacity, and to some extent immunomodulatory kemia (HCL) and some viral disorders. Alferon N® is an
activity, although gamma is a stronger immunomodula- aqueous formulation of human alpha interferon proteins
tory molecule than the type I interferons [19]. The enor- with a specific activity of 2 × 108 IU/mg of protein and is
mous amount of clinical study of the interferons in manufactured in the United States by the Purdue
malignancy established the field of biologic therapy of Frederick Company. It has been approved in the United
cancer. The many new agents currently under investiga- States for the intralesional treatment of refractory or
tion to inhibit angiogenesis and the aberrant growth fac- recurring external condylomata acuminata in patients
tor networks associated with malignancy have all taken over 18 years of age. These natural alpha interferons
advantage of the insights provided during the clinical contain most, if not all, of the multiple alpha molecules.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 277
© Springer Science + Business Media B.V. 2009
278 Interferons: therapy for cancer

In addition to the natural alpha interferon products, found in the native molecule. Betaseron® is approved in
there are several pure single alpha molecules obtained the United States for the treatment of ambulatory patients
by recombinant technology that are available for clini- with relapsing-remitting multiple sclerosis. Biogen, Inc
cal use. Interferon alpha A (alpha 2) has been pharmaco- has produced a recombinant molecule from the natural
logically produced as recombinant alpha interferon by beta interferon gene, which, since it is produced in eukary-
three companies, Hoffman-LaRoche in Nutley, NJ; otic cells, is glycosylated. This product, with the trade
Schering Corporation based in Kenilworth, NJ, and name Avonex®, has been approved in the US for treatment
Boehringer Ingelheim in Germany. These products are of relapsing forms of multiple sclerosis.
known as Roferon A® (rIFN alfa 2a), Intron A® (rIFN Serono also has a beta interferon molecule interferon
alfa 2b) and Berofor® (rIFN alfa 2c), respectively. The beta-1a for multiple sclerosis. Two recombinant gamma
alpha subtype, interferon alpha C (alpha 10) has been interferon molecules have been cloned and were clini-
produced in pharmacologic quantity in Israel. cally developed, one by Genentech, Inc., and one by
Roferon A® is manufactured by Roche Laboratories Biogen, Inc.
using recombinant DNA technology employing a geneti- These will be referred to, respectively, as rIFN gamma
cally engineered E coli bacteria containing an interferon (Genentech) with a trade name of Actimmune® and rIFN
alpha 2 gene obtained from a human myeloid leukemia gamma (Biogen) with a trade name of Immuneron.
cell line; it has an approximate MW of 19,000 daltons Actimmune® is manufactured by bacterial fermentation
and a specific activity of 2 × 108 IU/mg protein and is of a strain of E coli containing the DNA, which encodes
approved for use in the United States for the treatment of for the human gamma interferon molecule. It has a
HCL and AIDS-related Kaposi’s sarcoma in patients 18 specific activity of 30 million units/mg protein, and is
years of age or older. Intron® A is produced by Schering approved for clinical use in the United States for reducing
Corporation and is obtained from the bacterial fermenta- the frequency and severity of serious infections associ-
tion of a strain of E coli which bears a genetically engi- ated with chronic granulomatous disease. Immuneron is
neered plasmid containing an interferon alpha 2 gene not yet approved for prescription use in the US. These
from human leukocytes. It has a specific activity of various products are listed in Table 1. The generic desig-
2 × 108 IU/mg protein and is approved in the United nations will be used throughout this chapter.
States for treatment of patients 18 years of age or older Intracellular signaling in response to binding of type
with HCL and for selected patients with condylomata one and two interferons to their respective receptors has
acuminata, AIDS related Kaposi’s sarcoma, chronic hepatitis been carefully dissected out. Both subtypes of interferon
non-A, non-B/C and with chronic hepatitis B. result in binding and phosphorylation of Janus activated
Pegylated interferon alpha has been extensively stud- kinase molecules (Tyk2, Jak 1 and 2) followed by phos-
ied in hepatitis [140, 152] and initially in renal carci- phorylation of signal transducers and activators of tran-
noma [165] and chronic myeloid leukemia [111]. It scription (Stats) which translocate to the nucleus and
reduces frequency of administration (once/week versus activate interferon stimulated genes [198, 113].
three) and has an improved toxicity profile. A recombi-
nant IFN beta, with a single amino acid substitution
separating it from the natural beta molecule, was devel- Table 1. Interferon nomenclature
oped by Triton Biosciences, Inc., Berkeley, CA, and is
IFN Generic Trade name
currently manufactured by Berlex and marketed by
Chiron Corporation (US) and Schering AG (Germany) Natural alpha interferon
under the trade name Betaseron® for treatment of mul- Cantell IFN IFN alfa (Le) Finnferon®
Lymphoblastoid IFN alfa N1 Wellferon®
tiple sclerosis. It is obtained by bacterial fermentation of Leukocyte derived IFN alfa N3 Alferon N®
a strain of E coli that bears a genetically engineered Recombinant alpha interferon
plasmid containing the altered gene for human inter- Alpha A rIFN alfa 2a Roferon A®
feron beta. The native gene was obtained from human Alpha A rIFN alfa 2b Intron A®
fibroblasts and altered in a way that substitutes serine Alpha A rIFN alfa 2c Berofor®
Alpha C rIFN alfa 10
for the cysteine residue found in position 17 in the native
Recombinant beta interferon
molecule (interferon betaser17). rIFN beta 1B Betaseron®
The recombinant protein is a highly purified molecule rIFN beta (Biogen) Avonex®
with 165 amino acids, a MW of approximately 18,500 Recombinant gamma interferon
daltons and a specific activity of approximately 32 MU/mg rIFN gamma (Genentech) Actimmune®
protein; it does not include the carbohydrate side chains rIFN gamma (Biogen) Immuneron‫‮‬
D. Goldstein et al. 279

Clinical use this biologic was rapidly approved in most countries for
the treatment of this previously untreatable malignancy.
In the early 1970s, work by Kari Cantell and co-workers
led to the production of sufficient quantities of alpha
interferon, made from buffy cell layers, to support lim-
Hairy Cell Ieukemia
ited clinical trials in patients with several types of malig- Hairy cell leukemia, a B cell malignancy, is a disease
nancies [253, 254, 50]. This work was expanded in with an exquisite sensitivity to alpha interferon and was
North America by the U.S. National Cancer Institute the first human malignancy to be so identified. Prior to
(NCI) which sponsored trials beginning in 1975 and by the development of the interferons, no adequate therapy
the American Cancer Society (ACS) which supported existed for this relatively rare disorder. Splenectomy was
trials beginning in 1978. the standard approach when treatment was necessary and
Despite very limited clinical information, interferon was useful for controlling pancytopenia for a period of
was heralded in the popular press at the time as a signifi- time but had no effect on the leukemic process. Quesada
cant new drug for the cure of cancer. The early clinical was the first to demonstrate the beneficial effect of alpha
trials with this partially purified natural alpha interferon interferon in this disease in 1982 [205].
demonstrated response rates in breast cancer, osteosar- After finding that a dose of 12 MU/m2 was poorly tol-
coma and lymphoma, comparable to those achievable erated, he and his colleagues at the MD Anderson
with chemotherapeutic agents. The wave of enthusiasm Cancer Center, arbitrarily settled on a regimen of 3 MU
and outpouring of venture capital rapidly led to the (1 MU = 106 units) administered daily subcutaneously.
development of DNA recombinant technology and, fol- Several other investigators followed suit and have
lowing the successful cloning in 1979 by Taniguchi et al. shown that high response rates are attainable with the
of beta-interferon [259] and the subsequent cloning of potential for a prolonged response duration, even with
the alpha interferon subtypes and gamma interferon, the lower doses.
production of recombinant interferon molecules. Seventy-five percent to 80% of patients will obtain
Furthermore, the isolation and purification of natural major clinical benefit with improvement in hematologic
alpha Interferon from Namalva cell cultures led to far parameters and a decrease in the leukemic (tumor cell)
greater availability of natural alpha interferon and the population. Treatment of several months’ duration is
number of clinical trials increased dramatically. required for maximal benefit and continued treatment is
The partially purified IFN alfa (Le), used in the early necessary to maintain clinical benefit. Equivalent effi-
trials sponsored by the NCI and ACS, had a relatively cacy has been shown with each of the alpha interferon
low specific activity and, since it was a supernate, was products and with beta interferon [69, 82, 207, 284, 285,
composed of not only a variety of subtypes of alpha 76, 77, 83, 84, 72, 280, 242]. Gamma interferon is inef-
interferon but multiple other cytokines as well. [95, 18] fective in this disease [209].
working with patients with breast cancer in the US [50] Following Quesada’s lead, multiple studies by Golomb
working with patients with ovarian carcinoma in et al. at the University of Chicago, established an objec-
Scandinavia, reported response rates of over 20% using tive anti-leukemic response (CR or PR) rate of 20–25%
this natural alpha interferon product. These results have and an improvement in hematologic parameters in another
not been reproduced in these tumors using rIFN alfa 60% for an overall major clinical benefit in 80–85% of
2a or 2b, raising the question of what other active patients [82, 83, 84]. Following cessation of therapy,
constituent(s) in this interferon preparation may have patients relapsed but remission could be successfully
induced these responses. reinduced [208, 84].
The studies by the groups at MD Anderson and the
University of Chicago with recombinant alpha inter-
B Cell Malignancy feron led to the approval of alpha interferon in the
United States for the treatment of HCL. A significant
Several B cell malignancies have been shown to be cost benefit associated with interferon treatment was
therapeutically sensitive to alpha interferon. Multiple demonstrated [190] and additional studies with natural
myeloma and non-Hodgkin’s lymphoma (NHL) were alpha interferon demonstrated its superiority over sple-
among the first malignancies to respond in the early nectomy, the inadequate standard of therapy prior to the
trials with the Cantell interferon. development of alpha interferon [244].
Hairy cell leukemia (HCL) was shown in the early During the mid-1980s, investigators in Innsbruck
1980s to be exquisitely sensitive to alpha interferon and began to explore the question of dose effect in patients
280 Interferons: therapy for cancer

with HCL and determined that a dose as low as 0.5 MU cycle to melphalan/prednisone which were given on
(500,000 U) was biologically active and immunologi- days 1–4 and demonstrated a benefit in overall response
cally stimulating, as measured by the production of and survival for patients with either IgA or Bence-Jones
neopterin [105, 69]. Subsequent comparative studies myeloma but not IgG myeloma [188]. This study was
with rIFN alfa 2c [72] and IFN alfa N1, showed the low the third in a series of trials by this group to demonstrate
dose was as effective as the standard dose in inducing a a clinical benefit from this natural alpha interferon in
return to normal of the platelet count (within 36–38 days patients with IgA myeloma. The explanation for benefit
median) and of the neutrophil count (within 90–130 only for patients with IgA myeloma in this series of trials
days median), as well as reducing the need for red cell is not apparent [227].
transfusion support and the incidence of infection. The A study of VMCP with or without alpha interferon
standard dose, however, was clinically and statistically randomised 240 patients, there was no difference in
more effective in its antileukemic effect, i.e., reducing response rate but fewer patients receiving interferon
hairy cell infiltration in the marrow [242]. progressed and the median duration of response was 6
However alpha interferon has been supplanted by months longer (12.4 months versus 18.3 months, p < 02)
cladribine [289, 26]and pentostatin [112] as the treat- for patients receiving alpha interferon [146]. A subse-
ment of choice, just 1 decade after it was shown to be the quent study evaluated the addition of alpha interferon to
first effective systemic agent for the treatment of this dis- a five-drug combination (VBMCP) [182]. In 628
ease. Its role as an additional agent has not proven to be patients the complete response rate was increased 18%
significant [153]. Of interest a new biologic agent – anti versus 10% and there was an increased time to progres-
cD22 monoclonal antibody combined with an exotoxin sion with rIFN alfa2, 30 months versus 25. Of interest
has shown activity in cladribine resistant disease [128]. the best response was seen in the IgA subgroup. A small
Thus the promise shown by interferon that a biologic Australian study of adding interferon alpha 2a to inten-
agent could have an anti tumour impact and modify the sive combined chemotherapy failed to show a benefit in
natural history of a malignant disease has been kept. It either response rate or overall survival [114].
is likely that more targeted biologic therapy will remain Two meta-analyses have now examined the clinical
an integral part of the treatment of this disease. advantage of adding interferon to chemotherapy. Ludwig
et al. performed a global analysis on 30 trials evaluating
a total of 3,948 randomized patients, 17 of which added
Multiple Myeloma interferon and concluded there was a slight (6.6%)
Interest in the treatment of multiple myeloma with alpha advantage in response rate and median relapse free sur-
interferon was initially stimulated by the pilot studies of vival (4.8 months) and overall survival 3.1 months
Mellstedt et al. in 1979 and the ACS-supported trial [146]. The subsequent individual patient meta-analysis
reported in 1980 [160, 187]. Each study demonstrated of 24 trials with 4,012 patients confirmed the slightly
objective anti-tumor responses in a small number of higher response rates (57.5% versus 53.1%) and pro-
previously heavily treated patients. A number of phase gression free survival – 33% versus 24% at 3 years, a
II trials of both natural and recombinant alpha interferon 22% reduction in risk.
were subsequently performed and cumulatively sug- The median time to progression was increased by 6
gested that about 20% of patients with multiple months and the risk of death decreased by 8% [92].
myeloma, refractory to cytotoxic chemotherapy, might However, any advantage of adding interferon to induc-
obtain an objective response to a variety of doses and tion chemotherapy in this disease is too small to be
schedules [38, 276, 181, 25, 36, 180, 206, 37]. clinically meaningful and is probably not worth the
Because of these encouraging results in previously added financial cost (US$42,236.19/life year saved) or
treated patients, alpha interferon was compared with cyto- side effects. Open to further exploration however is the
toxic chemotherapy in previously untreated patients but persistent intriguing observation of the possible benefits
chemotherapy proved superior in terms of both response in patients with IgA/Bence-Jones myeloma.
rate and duration of response [4, 147]. Several trials were Several trials were next established to evaluate the
organized to evaluate the addition of alpha interferon to use of alpha interferon as a maintenance agent in patients
various combinations of cytotoxic chemotherapy in terms with myeloma. Following the induction of a response
of response induction [35, 227, 114, 182]. with cytotoxic chemotherapy, various groups random-
One of these a large Swedish trial involving over 300 ized patients to receive alpha interferon or no further
patients evaluated the addition of IFN alfa (Le) admin- therapy until the onset of progressive disease [151, 223,
istered on days 1–5 and days 22–26 of every 4-week 195, 279, 20, 16, 146, 225].
D. Goldstein et al. 281

In addition, the Nordic Study Group randomized retrospective analysis of 473 patients that received
patients at the start of treatment; those randomized to maintenance IFN compared to 419 patients who did not
receive alpha interferon received it continuously [91]. that overall survival and progression free survival were
These studies are summarized in Table 2. The US significantly better – 78 months versus 47 months and
cooperative group trial and the trial in Germany showed 29 versus 20 months. The retrospective nature of the
no benefit from interferon therapy. The other seven trials report requires cautious interpretation [15]. A third study
have demonstrated either a prolonged time to progression from the Nordic Group also used maintenance inter-
or duration of plateau phase in the interferon maintained feron following high dose therapy and showed a benefit
group. Ludwig et al. also performed a global analysis for for this program over a historic control but the role of
these maintenance studies also and concluded that alpha interferon itself in prolonging survival cannot be deter-
interferon both relapse free survival by 4.4. months and mined [137].
overall survival by 7.9 months at a cost of US$18,114.95 In summary, alpha interferon is an active agent in
(Figures 1 and 2) [146]. The individual patient meta myeloma. Current data support its greatest usefulness
analysis showed an increase in progression free survival when used as an agent capable of prolonging remission
at 3 years of 27% versus 19%, a 34% reduction in risk of in patients with low tumor burden; in some of these
progression and a 12% decreased risk of death [92]. patients it may also improve survival.
Median survival is increased by 4 months.
Such an effect also needs to be balanced against not
only cost but also quality of life. In that regard a recent
Non-Hodgkin’s lymphoma (NHL)
study suggests that after 12 months there is no signifi- Several clinical trials conducted in the late 1970s with
cant difference in quality of life in two Nordic mainte- the Cantell interferon suggested efficacy in patients with
nance studies [282]. A recent study with pegylated low and, to a lesser extent, intermediate grade NHL
interferon suggests it may further improve quality of life [161, 95, 144, 104].
in this setting [240]. Single agent trials in the 1980s with the more highly
A randomised trial of maintenance alpha interferon, purified recombinant and natural alpha interferons con-
following high dose melphalan consolidation with bone firmed the efficacy of alpha interferon in patients with
marrow transplant rescue has also been performed. rIFN NHL [64, 177, 136].
alfa 2b had a beneficial effect on prolonging remission, Two single institutions and the CALGB, in three sep-
especially in those patients who achieve a complete arate trials, combined alpha interferon with an alkylat-
remission 46 versus 27 months at 52 months of follow ing agent in patients with low grade NHL and
up and in improving survival at 52 months 95% versus demonstrated the combination to be relatively non-toxic
75%. However by 77 months most patients had died and and well tolerated. The combination induced a response
the benefit was not sustained [40]. In addition the in 50–75% of patients, more readily in previously
European marrow transplant registry has reported in a untreated individuals [29, 28, 189, 30].
The group at MD Anderson, in a single institution
non-randomized study, evaluated the use of IFN alfa N1
as maintenance therapy, inducing a response with CHOP
Table 2. IFN versus CT in CML
plus Bleomycin and then treating patients with IFN alfa
N1 until relapse [157]. They concluded, based on his-
IFN effect on Dur
torical controls, that alpha interferon prolonged the
Group/reference Comparison of Resp survival
duration of response but did not improve survival.
Italian Alpha IFN versus CT + + Subsequently a large number of randomised trials have
study group
(202) been reported in complete or abstract form evaluating,
German study Alpha IFN versus + + by direct comparison, the addition of alpha interferon to
group (75, 76) busulfan cytotoxic chemotherapy in the initial treatment of the
Alpha IFN versus HU + Neg disease.
Japanese study Alpha IFN versus + +
In studies using less intensive regimens such as
group (128) busulfan
UK study group Maintenance with α + + chlorambucil or CVP there has been no benefit on over-
(7) IFN versus no all survival and at best a marginal improvement in pro-
maintenance gression free survival in one study [197, 96, 216]. There
HU = Hydroxyurea have been two large prospectively randomized trials
282 Interferons: therapy for cancer

Study start year, Prog’sions/Patients Statistics O.R. & Cl* Odds Redn.
code and name IFN None (O-E) Var. (IFN : None) (SD)

Interferon in Induction:
85G GATLA 3-M-85 13/16 14/15 -2.6 6.3 34% (33): P = 0.5
86I Rome IFN 1 15/21 16/19 -4.2 6.5 45% (29): P = 0.1
86J MGCS 1986 97/105 61/69 -2.9 37.9 -5% (17): P = 0.6

87E EMSG 2 (1) 32/70 37/56 -7.9 16.7 35% (19): P = 0.6
88A EOOG 9486 123/161 121/153 -13.9 59.2 21% (12): P = 0.07
89H KIF, Avicenne 43/97 42/95 1.3 21.2 -7% (22): P = 0.8

90D NMSG 04–90 83/125 89/134 -16.4 44.6 31% (15): P = 0.01
90H ALSG Myeloma II 14/24 21/26 -8.0 7.9 64% (22): P = 0.016

90J Ital NHLSG (I) 11/28 15/21 -4.7 6.0 54% (20): P = 0.04
91B GMM (I), Mexico 20/40 14/40 0.7 6.9 -51% (40): P = 0.6

Subtotal 451/685 440/638 -52.7 213.4 22% (6)


(65.8%) (69.0%) reduction
P = 0.0003
Test for heterogeneity between trails: c20 = 15.8; P = 0.07

Interferon in maintenance:
85D Ital. MMSG M84 35/50 44/51 -14.0 18.0 54% (15): P = 0.001

87C SWOG 8624 78/107 83/104 -8.1 39.9 15% (14): P = 0.2
87D NCI-C MY6 66/89 80/92 -21.1 33.9 45% (15): P = 0.0003
87H MGWS (extended) 45/51 62/64 -23.5 23.6 63% (15): P < 0.0003
89B EMSG 2 (M) 22/46 37/54 -8.8 14.6 45% (15): P = 0.09
89E GMTG MM02 41/52 56/65 -5.0 24.0 10% (16): P = 0.3
89F Royal Marsden 31/42 33/42 -6.0 15.5 20% (21): P = 0.1
89A MRC-MYEL-6e 118/143 116/140 -12.8 57.8 20% (12): P = 0.00
89E CMN (M). Mexico 9/13 7/8 0.6 1.1 -21% (120): P = 0.5

90B PETHEMA 38/50 40/42 -8.7 18.4 39% (19): P = 0.01


90K Ital. NHLSG (M) 30/44 26/48 0.5 14.3 -4% (27): P = 0.9

91E GERM 36/70 46/66 -10.0 19.4 40% (16): P = 0.02

Subtotal 549/787 635/776 -116.7 -260.5 34% (5)


(71.6%) (81.8%) reduction
P < 0.00001
Test for heterogeneity between trials: c211= 21.7; P = 0.03

Total 1000/1452 1075/1414 -169.4 493.9 29% (4)


(68.9%) (76.0%) reduction

95% CI for total 98% CI for individual trials


0.0 0.5 1.0 1.5 2.0
IFN None
better better
Test for heterogeneity (22 trials): c231 = 41.0; P = 0.008 Effect P < 0.0001
Test for heterogeneity between subtotals: c21 = 3.5; P = 0.08

Figure 1. Meta-analysis of impact of interferon on progression free survival (adapted from report in the Br J Haematol, ref.
227)
D. Goldstein et al. 283

0 1 2 3 4 5 6+ years
100

Estimated percentage still progression free


90

80

70

60

50

40
32.6%

30
22.9%
20.0%
20 23.8% 5.3 % SD 2.0
(logrank
- allocated IFN (% ± s.d.) 16.2% 2P < 0.00001)
10 - allocated none (% ± s.d.) 14.7%

0
0 1 2 3 4 5 6+ years
Progressions/period–years
IFN 412/1214 237/770 184/450 57/287 27/176 28/224
None 561/1102 319/558 115/300 45/129 16/113 16/114

Figure 2. Progression free survival for the addition of interferon to standard treatment of multiple myeloma (adapted from
report in the Br J Haematol, ref. 227)

performed by cooperative groups in the United States and an improved overall survival (not reached versus 5.6
and Europe evaluating the effect of alpha interferon years, 10% of patients withdrew because of IFN related
added to four-drug induction cytotoxic chemotherapy toxicity and 28% required dose reduction).
[244, 246]. Both trials added alpha interferon to a The initial ECOG publication reported an improve-
CHOP-like induction regimen; the Group d’Etude des ment in survival but a subsequent analysis a year later,
Lymphomes Folliculaires (GELF) treated only patients although demonstrating a consistent 10% increase in
with follicular lymphoma but with bulky (>7 cm) dis- survival over a 5 year period, did not show statistical
ease while the ECOG in the United States studied significance [8]. A study of quality of life has similar to
patients with bulky or symptomatic low grade and inter- the myeloma study supported a benefit in terms of qual-
mediate grade NHL. The ECOG used cyclophosph- ity adjusted life years for IFN treatment [32].
amide, Oncovin®, prednisone and Adriamycin® (COPA) By contrast a large SWOG study in 571 patients com-
as the chemotherapy regimen, administering alpha inter- paring eight cycles of ProMACE 9 day 1-MOPP (day 8)
feron for 5 days (D22–26) of every 28 day cycle for a with and without interferon alpha as maintenance for 2
total of 8–10 months. years showed no benefit in either progression free or
The GELF used cyclophosphamide, VM 26, predni- overall survival [62].
sone and Adriamycin® monthly for 6 months and then In addition to these reported trials, there have been
every 2 months for an additional 12 months giving alpha two small trials examining the issue of dose, one show-
interferon three times a week for the entire 18 months. ing a dose response [68, Gams, 1990, personal commu-
Although the two trials were very similar in design, nication] the other no statistical difference in response
there were some important differences. Both groups rates. The question of influence of dose and schedule
treated patients with bulky, symptomatic disease, all fol- remains unanswered because of the very small numbers
licular lymphoma in one, and follicular and diffuse in of patients.
the other while the aggressiveness and length of treat- A meta-analysis only reported in abstract form [215]
ment with both chemotherapy and alpha interferon was – summarised eight randomised trails involving 1,756
different. patients. It suggested that maintenance treatment in trials
The GELF demonstrated a better response rate in the with more intense initial therapy showed a 14% survival
group receiving interferon, but had an unusually low advantage at 5 years (74% versus 60%) and 19% at 8
response rate in the chemotherapy-only arm. The GELF years, but no benefit for lower initial intensity treatment.
study demonstrated an improved duration of response Thus Alpha interferon may be an active therapeutic
and time to treatment failure 2.9 years versus 1.5 years agent in patients with NHL, low grade, but its role has
284 Interferons: therapy for cancer

yet to be clarified. Optimal dose and schedule have yet response was seen in earlier stages (Ia, b, Iia), frequency
to be defined and the question of whether higher, less of complete response was maximal by 6 months. 57%
tolerable doses are needed for maximal effect remains relapsed within 1 year but 175 had a very prolonged
unanswered. Additionally, alpha interferon may be most complete response with a mean of 31 months [115]. One
active in patients with lower tumor burdens, i.e. with or randomized trial has been reported comparing the rela-
following CHOP-like regimens. tive value of the combination of PUVA and IFN to retin-
In high grade NHL a small randomised study showed oic acid and IFN [249]. The PUVA combination had a
no benefit to 1 year of maintenance therapy following higher complete response 70% versus 38% and overall
initial therapy [10]. response 80% versus 60%.
However a recent survey has shown some evidence Once again the recent reporting of an IL-2 toxin conju-
of activity in relapsed high grade disease [9] and trials in gate with efficacy in the resistant disease setting, although
the post transplant setting for relapsed high grade at a cost in terms of toxicity [249] suggests that biologic
patients are ongoing. The whole role of IFN is now therapy will have an ongoing role. Combination of IFN
uncertain following the approval of anti CD-20 mono- with emerging new therapeutics is continuing [226].
clonal antibody (mabthera – see chapter). Several small pilot trials have evaluated the use of
Its activity and excellent toxicity profile for relapsed alpha interferon in patients with CLL [64, 229, 177,
low grade lymphoma both alone and conjugated with 193]. Alpha interferon appears to have a mild to moder-
radionucleide and the increasing data on its use as initial ate ability to decrease the population of circulating leu-
therapy in both low grade and high grade NHL [88] kemic cells, especially in patients with early or minimal
make it a very attractive biologic agent. disease who have not received prior therapy. However,
However given that patients may ultimately relapse none of these studies strongly suggest a clinically sig-
following mabthera therapy the role of IFN alpha to nificant benefit from alpha interferon as a single agent.
enhance the effectiveness of this therapy requires fur- ATLL has been etiologically associated with infec-
ther study [41]. In addition novel directions in combina- tion with HTLV-1, a human retrovirus endemic in south-
tion to avoid chemotherapy are being examined [120]. ern Japan and the Caribbean basin [201]. All three
interferons, alpha, beta, and gamma have been shown to
have an anti-tumor effect occasionally durable, in a
Other lymphomas small percentage of patients with the acute form of this
Several small phase II studies have been performed disease. A multi-institutional group studied the two drug
in-patients with cutaneous T cell lymphoma (CTCL), combination of rIFN alfa 2b and AZT in 19 patients and
chronic lymphatic leukemia (CLL), and adult T cell leu- induced a major response in 11 patients, including a
kemia- lymphoma (ATLL). Alpha interferon has been complete response in five [78]. Some of these patients
shown to have an anti-tumor effect in patients with these had disease resistant to cytotoxic chemotherapy. Several
disorders. Bunn et al. initially showed that rIFN alfa 2a of the responses were durable.
induced responses in patients with CTCL. Responses
were seen in both cutaneous and extracutaneous sites
[21, 22].
Chronic Myeloid leukemia (CML)
These encouraging results were followed by a CML is a triphasic disease, consisting of chronic, accel-
confirmatory report in a small series from Duke and erated, and blastic elements. Management options
Northwestern [185]. Three complete and ten partial include stem cell transplants, alpha-interferon, the
responses (response rate-59%) were induced and the tyrosine kinase inhibitor (STI571) and chemotherapy.
suggestion of a dose–response relationship was demon- The choice of therapy depends on the phase of the dis-
strated. Investigators from Northwestern University ease and the characteristics of the patient. Interferon has
then combined rIFN alfa 2a with psoralen plus ultraviolet most effect in the chronic phase as initially found by
light irradiation (PUVA) in a Phase I dose escalation Talpaz et al. at MD Anderson [256]. This group con-
trial in which the dose of alpha interferon was escalated ducted a series of uncontrolled trials, which initially
from 6 to 30 MU IM tiw [132]. demonstrated the ability of interferon alphã2a to control
This escalation was based on the steep dose–response leucocytosis and subsequently demonstrated its ability
relationship observed in the earlier trials. A complete to reduce the size of the Ph positive clone and to induce
response was obtained in 12 patients (80%). Several a clinical and histologic CR. Large multi-institutional
phase II trials in patients with cutaneous T cell lym- randomized trials have since demonstrated the superior
phoma have confirmed this high response rate [133, efficacy of alpha interferon compared to cytotoxic treat-
222, 283]. In one long term follow up report the best ment in terms of haematological response and survival
D. Goldstein et al. 285

and, until recently, it was established as the treatment of studies from Japan and the UK have reached similar
choice for patients with CML who were not eligible for conclusions [179].
a bone marrow transplant. In the Japanese study, newly diagnosed patients with
CML in chronic phase were randomized to receive
either alpha Interferon or busulfan. Although a complete
Interferon Monotherapy; Early Studies
cytogenetic response occurred in two patients receiving
Based upon the demonstration of in vitro antiprolifera- busulfan (not previously noted with this agent), there
tive activity of alpha interferon against CFUs [183, 174, was a statistically and clinically significant difference in
210, 12]. Talpaz and co-investigators initiated a series favor of alpha interferon in terms of major cytogenetic
of trials in the early 1980s initially using partially puri- response (13 of 80 patients) and in predicted 5-year sur-
fied alpha interferon in patients with CML and subse- vival rate. The UK study using a different approach
quently with recombinant alpha interferon molecules. demonstrated superiority of alpha interferon over no
Their initial observations that alpha interferon could maintenance therapy after induction of a response by
reduce the size of the malignant clone of cells was sub- either busulfan or hydroxyurea [6].
sequently confirmed; treatment with alpha interferon
reduced the number of Ph positive cells in over half of
Interferon in combination with cytotoxic
the responding patients.
About 20% of patients obtaining a conversion to a drugs in the treatment of CML
cytogenetically normal marrow, confirmed by molecular Interferon has been used in combination with cytarabine
studies [255, 256, 257, 288]. This is a true pathologic CR. (ara-C) following work showing ARA-C could selec-
Such an effect had met with only limited success using tively suppress CML clones in vitro and also demonstrate
aggressive cytotoxic chemotherapy [241]. It is now antitumour activity when given as a low dose continuous
clear that patients gaining a major partial or complete infusion in vivo.
cytogenetic response have improved survival. The MD Anderson experience of combining inter-
feron alpha with ara-C has been addressed by Kantajaran
Randomized Studies of Interferon et al. They initially reported on a group of 60 patients
with advanced phases of CML in 1992 showing that a
Monotherapy Versus Chemotherapy study group receiving daily alpha interferon and inter-
A number of prospectively randomized studies have mittent ara-C had a better CHR compared to historical
confirmed the superiority of interferon monotherapy controls receiving interferon alone. A subsequent report
over chemotherapy in the treatment of early chronic on 140 patients with Ph positive early chronic phase
phase CML (Table 2). CML in 1999 [117] showed that the schedule of ara-C
The Italian Cooperative Group first demonstrated may also be important. The study group received com-
alpha interferon’s superiority over hydroxyurea with a bination treatment of IFN-alpha (5 × 106 U/m2 daily)
karyotypic response rate of 30% in the interferon group and low dose ara-C (10 mg/m2 daily), compared to his-
versus 5% in the chemotherapy group and significantly torical controls receiving interferon (5 × 106 U/m2 daily).
improved median survival in patients given interferon A significantly improved CHR (92% versus 84%) was
[265]. A prospectively randomized study by the German seen with low dose continuous versus intermittent ara-C
CML Study Group compared the use of IFN alpha 2a, and although there was a trend to an improved major
given at a daily dose of up to 9 MU SC, with standard cytogenetic response this was not statistically significant
cytotoxic chemotherapy either busulfan or hydroxyurea (50% versus 38%, p = 0.06).
[98, 99]. The number of patients with a reduction in the Two randomized studies using a combination of
Ph1 positive clone and the time to progression to accel- interferon with cytotoxic drugs have also been reported.
erated or blast crises were both increased in the inter- Guilholt et al. (240) reported on 721 patients with previ-
feron group compared to either cytotoxic agent. ously untreated early phase CML. Patients received
The overall survival of patients was superior in the hydroxyurea (50 mg/kg/day) with interferon (5 × 106 U/
group of patients treated with alpha interferon ther- m2 daily) with or without cytarabine (20 mg/m2/day for
apy but statistical significance was demonstrable only 10 days each month). There was a significantly improved
when interferon was compared to busulfan. This high CHR (66% versus 55%) and also a survival advantage
daily dose was poorly tolerated, however, and 16% of at 3 years (86% versus 79%) in the patients given the
patients had to discontinue alpha interferon therapy. ara-C. An Italian study [220] randomized 540 patients
The financial cost of interferon therapy was also with Ph positive chronic phase CML to receive daily
substantially higher than for chemotherapy. Further interferon alone or in combination with ara-C (40 mg/
286 Interferons: therapy for cancer

kg/day subcutaneously for 10 days/month). The com- tial thrombocytosis and polycythemia rubra vera [238,
bined treatment group had a significantly increased 79, 94, 135, 258, 261]. Control of the markedly elevated
major and complete cytogenetic response rate (28% ver- platelet count, decreasing the risk of resultant life-
sus 19%) and Kaplan Meier calculated survival benefit threatening complications can be obtained in nearly all
of 85% versus 80% at 3 years. patients with this disorder. The red cell mass, in patients
Interferon is clearly highly active in the treatment of with PRV, can also be reduced leading to symptomatic
chronic phase CML being able to induce major cytoge- improvement [236]. Pegylated interferon can provide
netic responses reflecting a real survival benefit over these benefits with some reduction in toxicity [224].
conventional cytotoxic therapy. Combining it with ara-C
appears to increases activity further. However there are
a number of issues that should be considered. Firstly
one should assess the relative benefits of carrying out a
Solid Tumors
stem cell transplant. Alpha interferon has a demonstrable beneficial effect in
The full details required cannot be covered in the context patients with some solid tumors; patients with malignant
of this chapter but in young patients with a good perfor- melanoma, renal cell carcinoma, and AIDS related Kaposi’s
mance status a transplant should still be regarded as the sarcoma have benefited by treatment with alpha interferon.
treatment of choice. In addition the dosing and duration of Alpha interferon has been approved in the US for the treat-
interferon treatment to achieve maximal anti-tumour effi- ment of two of these tumors, AIDS-related Kaposi’s sar-
cacy must be balanced with its toxic side effects. coma and for high risk patients with malignant melanoma
Although their studies were uncontrolled, the MD following surgical removal of the primary lesion.
Anderson group has amassed a significant amount of
experience and believes that relatively high doses,
administered daily, are required for maximum beneficial
Kaposi’s Sarcoma
effect [118]. However, a number of their patients require Trials in patients with Kaposi’s sarcoma (AIDS) have
dose reduction because of lassitude, neurologic problems indicated that about 40% of patients will obtain an
and/or thrombocytopenia and neutropenia. anti-tumor response [130]. There has been the sugges-
By contrast a subsequent analysis of randomised trials tion of a dose–response effect, with better results com-
of the MRC and HOVON showed no benefit for high ing with treatment in the range of 20–50 MU/m2 [43,
dose versus low dose [127]. With regard to duration it 90, 212, 270].
appears that interferon only very rarely elicits a com- Most of these studies indicate benefit is most likely in
plete molecular response even when a complete cytoge- patients with stage II and III disease, in patients with the
netic response is achieved, so there was a real motivation absence of a history of opportunistic infection, and in
to obtain a more efficacious but less toxic therapy. patients with a relatively good lymphocyte (>15,000/
The advent of the tyrosine kinase inhibitor Glivec has mm3) and helper lymphocyte (CD4) count (>400/mm3).
filled that niche [45] reported in patients with chronic Patients with advanced stage disease and/or poor
phase CML that had failed interferon therapy showing immune status are less likely to respond [73].
that of 54 patients receiving more than 300 mg 53 had An analysis of 364 patients has suggested prolonged
complete haematological responses, with 29 obtaining survival for patients with the highest CD4 counts [56].
cytogenetic responses (17 of which were major or com- Studies of interferon in combination with chemotherapy
plete) with very little in the way of toxicity. This success have not shown any benefit over interferon alone [129].
has lead to the rapid FDA approval of Glivec in the Studies evaluated the combination of interferon with
treatment of interferon resistant disease. Subsequent AZT and suggested synergy against the HIV virus and
studies in first line therapy led to its adoption as the possibly also against the tumor, which is itself virally
standard of care [176]. induced by HHV-8 [155]. A prospective randomized
However the development of resistance to Glivec trial has shown that moderate doses of IFN, at least
therapy occurs both in chronic phase disease and blast 8 mU/m2 are needed [232]. A small randomised trial
crisis and the mechanisms of resistance have been anal- against chemotherapy has further supported its use with
ysed at a molecular level [85]. Despite new targeted an increase in survival of 24 versus 13 months [186].
agents, interferon may well still have an important role Unfortunately with the exception of those patients with
as an additional therapeutic option. high CD4 counts, the length of response is only approxi-
Alpha interferon also has clinical utility in patients mately 6 months and, a significant beneficial effect on
with other myeloproliferative disorders including essen- survival in the majority of patients remains undefined.
D. Goldstein et al. 287

Once again this indication may be of limited use as with interferon produced a tumour response rate 53%
the dramatic impact of effective combination antiretro- greater than with DTIC alone (95% CI 1.10–2.13) but
viral therapy has sharply reduced the incidence of kapo- this was not translated into a survival benefit.
sis sarcoma and should always be the first approach in a There have also been studies that have used interferon
new patient with Kaposis Sarcoma. Furthermore excel- in combination with interleukin 2 as additional therapy
lent palliation without the toxicity of IFN is now possi- to conventional cytotoxic drugs. Although there is fre-
ble with liposome encapsulated doxorubicin (Doxil). quently an increased response with the addition of
Immunotherapy, statistically significant survival advan-
tages are again lacking [119, 217, 55]. This conclusion is
Melanoma also supported by an additional meta-analysis which also
The role of interferon 2 alpha in the treatment of mela- examined chemotherapy combined with both interferon
noma has been investigated in the context of metastatic and interleukin-2 similarly showing improved response
disease and also as adjuvant therapy following the rates without a survival gain [110].
removal of lesions at high risk of recurrence. Thus, considering the considerable toxicity afforded by
Although there are a number of trials that have such therapy, further studies are unlikely to be of value.
addressed the role of interferon in metastatic disease most
of the current emphasis has been on defining the exact
role of interferon in the adjuvant setting and this will form
Adjuvant Therapy
the main point of discussion in this section. A number of trials in the last few years have addressed
the role of alpha interferon as adjuvant therapy after sur-
gical removal of high risk lesions. These have been
Advanced Disease designed to test whether interferon improves survival
Response rates in patients with advanced melanoma and if so which disease stage will benefit and how
vary from 5% to 27%, with an average of 15% [39, 122] intense the treatment must be.
and an intravenous schedule has allowed higher doses to The most highly influential of these was the ECOG
be used with less associated toxicity [122]. Metastatic E1684 trial which included patients with a T4 lesion,
malignant melanoma is a difficult malignancy to treat. with or without the presence of local lymph nodes, and
The response rates for single agent alpha interferon any other primary lesion with proven lymph node
compare favorably with those of single agent cytotoxic involvement (resected IIB or III) [125]. After surgery
agents, but response rates with single cytotoxic agent patients were randomized to receive high dose inter-
therapy are low compared to combinations of cytotoxic feron, consisting of an induction phase of 20 MU/m2 for
drugs [156]. Thus there has been interest in combining 5 days every week for 4 weeks followed by a mainte-
interferon with cytotoxic drugs. An early study random- nance phase of 10 MU/m2 for 3 days a week over 48
izing patients to receive DTIC 200 mg/m2 intravenously weeks in total, or observation. The Kaplan-Meier analy-
for 5 days every 4 weeks with or without interferon sis showed a statistically significant improvement in
(given as 15 MU/m2 for 5 days a week over 3 weeks and disease free survival (26% versus 37%) and overall sur-
then 10 MU/m2 sc three times a week) showed a statisti- vival (37% versus 46%) in the treatment group and led
cally significant improvement in response rate and median the FDA to approve the use of interferon alpha in this
survival in the interferon group. patient group. However there was substantial toxicity
Unfortunately a larger follow up study as well as a experienced in the treatment group and it was hoped
number of other trials have failed to demonstrate a sta- that a less toxic equally efficacious protocol could be
tistically significant advantage of adding interferon to a generated.
variety of cytotoxic regimens [58]. Thus there was considerable interest in the results of
Considering the additional toxicity afforded by inter- the WHO melanoma program where lower less toxic
feron it would therefore appear it has relatively little to doses were employed. This study included patients with
offer in the metastatic setting. However a recent meta- stage III disease using a 3 MU dose of drug three given
analysis of 3,273 patients in 20 randomised trials was times weekly compared with observation.
recently carried out assessing single-agent DTIC versus Unfortunately, although data is still being generated
combination chemotherapy with or without immuno- from this trial, no global overall or relapse free survival
therapy in metastatic melanoma [107]. benefit has yet been demonstrated indicating a low dose
This included 926 patients in five trials that utilized protocol may be ineffective in this patient group [24].
interferon and showed that the combination of DTIC This has been further confirmed in two studies of low
288 Interferons: therapy for cancer

dose interferon alone [97, 126] and two in combination clearly highly persuasive and if such data is also made
with interleukin-2 and retinoic acid respectively. This available from the E1690 trial it would reinforce the
has been reinforced by a further three arm ECOG study, advantages of adjuvant high dose interferon treatment.
E1690, evaluating the efficacy of high dose interferon, In spite of the above data there are two studies using
as documented in the 1684 study, compared with a lower low dose interferon have indicated some therapeutic
dose (3 MU/day three times weekly for 2 years) and efficacy in certain patient groups. For example Grob et
simple observation [123]. This study again showed a al. [89] compared 18 months of low dose interferon with
significant improvement in relapse free survival between observation alone in 489 patients with resected disease
the high dose treatment and observation alone. of >1.5 cm depth but with no nodal involvement. They
However there was no relapse free survival advantage showed a statistically significant improvement in dis-
in the low dose treatment compared to observation. Further ease free survival and a trend towards overall survival.
support for high dose adjuvant interferon was forthcoming Similar results were also published by Pehamberger et al.
in the recently published ECOG study, E1694. using a year of interferon treatment (196). Those patients
This trial was designed to compare a high dose inter- without nodal involvement may therefore represent a group
feron with a markedly less toxic GM2 ganglioside vac- that could be treated with a less aggressive protocol.
cine, thought to be active in melanoma, to see if the The length of treatment required is uncertain as a
vaccine offered a more tolerable alternative. Here the shorter course would clearly improve the toxicity.
interferon arm was clearly superior after an interim anal- Although no randomised trials are testing node positive
ysis thus leading to premature closure of the study [124]. patients with very short interferon courses the E1697 is
All these studies therefore advocate the use of high dose currently looking at 1 month of HDI versus observation
interferon in the adjuvant setting for patients with posi- in T3 N0 resected disease. By contrast a trial – EORTC
tive nodal disease. However there are a number of issues 18952 comparing two intermediate doses of IFN alpha
that are cause for discussion. Firstly in the 1690 study 2b in high risk melanoma patients showed no benefit
where high dose, low dose and observation arms were [49a]. Recent data on pegylated interferon alfa-2b
compared there was no statistically significant overall shows similar benefits but with possible reduced toxic-
survival advantage in having the high dose treatment ity [49b]
compared to observation alone. This was is in contrast to A recent meta-analysis concluded that the use of HDI
the earlier E1684 study where a clear overall survival should be considered in the adjuvant treatment of resected
benefit was apparent. Of note the overall survival in the stage IIB and stage III melanoma because of improve-
observation arm in the later E1690 study approached that ments in disease free and 2 year mortality [268].
of the treatment arm in the E1684 study. The important questions as to whether the full
This has not been completely rationalised but one extended course is required, and whether one may be
possibility considers the availability of salvage inter- able to better select patients who will particularly ben-
feron treatment in those patients relapsing in the obser- efit from treatment remain unanswered. However for
vation arm of E1690. This was not available for patients stage IIA disease a low dose protocol may be appropri-
relapsing in the E1684 study and thus most of the obser- ate. In metastatic disease the additional issue of at best
vation arm patients in the E1690 study differed from very modest survival benefits versus additional toxic-
those in E1684 as they would have received interferon ity mean that treatment should be tailored on an indi-
at some point. vidual patient basis and clearly selection must be
It is possible therefore that any overall survival ben- careful.
efit apparent for the adjuvant treatment in E1690 may
have been diluted. Intuitively this may argue that the use
of interferon could be delayed until relapse considering
Renal Carcinoma
the toxicity it causes in the adjuvant setting. However Alpha interferon role in this malignancy may well now
one must remember that although other studies show be only of historical significance. It has an anti-tumor
interferon does have activity in metastatic melanoma effect in about 12–15% of patients with metastatic
this has not been translated into a survival benefit (see renal cell carcinoma (RCC) [281, 204] and may be
previous section). In addition a follow up study address- more effective in patients with less bulky disease and
ing quality of life issues in the 1684 trial using Q-TWIST when given in higher doses [154, 61, 168, 273, 228,
analysis revealed that in spite of toxicity the adjuvant 231, 211].
treatment group had more quality of life adjusted sur- A sufficient number of randomised trials have now
vival time than the observation group [33]. This is been reported to allow a clear picture of the role of
D. Goldstein et al. 289

single agent interferon. Interferon either alone or in The response rate to interferon of 3% was surpris-
combination with vinblastine is more effective than ingly low however. The benefits of combining interferon
medroxyprogesterone [34] or vinblastine [204]. As a with any other agent appear limited [173, 66, 204, 7,
result a Cochrane Meta analysis [281] analysed 42 164, 46, 165, 166]. The role of interleukin-2 is addressed
studies involving 4,216 patients and showed an average in great detail in another chapter. However it is instruc-
response rate of 10.2% with 3.2 5 CR, a median survival tive to review the results of a recent randomised trial of
time of 11.6 months and 2 year survival of 22%. They interferon alfa versus IL-2 monotherapy versus the
estimated a pooled survival hazard ratio of 0.78 for IFN combination compared to medroxyprogesterone [171].
treated patients. They suggested that IFN alfa 10 MU In 492 patients no survival benefit was shown for either
S.C. three times per week should be the control arm for drug [172].
future studies (Table 3). The recent very impressive results with the antiangio-
A large randomised study of interferon gamma genic agents such as the VEGF inhibitors and their con-
[80] showed that this cytokine is inactive. It also sistent superiority over interferon has made them the
demonstrated that spontaneous remissions do occur standard of care [167, 54, 106]. However at least one
with significant frequency 6% (CI 2.5–13.2%), higher study showed a benefit to the combination of bevaci-
than previously reported and a 15.7 month median zumab and interferon which means it remains an option
survival was noted for this early stage metastatic for exploration but the lack of efficacy of the combination
group [53]. This suggested that the activity of inter- of temsirolimus and interferon compared to temsirolimus
feron alfa in this disease needs to be assessed against alone means that issues of toxicity may differ with each
that standard, at least in good performance early stage drug combination. Other approaches such as dendritic
disease. The role of interferon as adjuvant therapy cell vaccines [103, 131] and autologous transplantation
post nephrectomy showed no survival benefit [203, [27] may still be the subject of combined therapy also.
264, 200].
The benefit of nephrectomy as part of a combined
surgical-biotherapeutic approach has been examined.
Other Solid Tumors
241 patients were randomised to either interferon alone Aside from the three solid tumors listed above, there
5 mU three times per week or nephrectomy plus inter- have been trials in many other types of malignancy.
feron. The survival in patients allocated to nephrectomy There are some encouraging leads. rIFN alfa 2a has been
was superior 12.5 months versus 8.1 months [63]. shown to enhance the cytotoxic effect of 5-FU in vitro
and, in pilot clinical trials, the combination induced a
partial response in over 60% of patients [5, 274, 275].
Table 3. Recent randomised Trials of IFN alpha in renal cell However, a prospective randomized trial evaluating
carcinoma the addition of alpha interferon to 5 FU in patients with
12 advanced colorectal cancer has demonstrated no benefit,
months 24 only added toxicity, from the addition of alpha inter-
RR MST survival months
feron [102].
N (%) (weeks) (%) (%)
By contrast the development of combination therapy
VBL #260 81 2.5 37.8 38 19
with oxaliplatin, irinotecan and the addition of bevaci-
IFN + VBL 79 16.5 67.6 56 38
IFN #121 53 11 34 N/A zumab and cetuximab to them has opened new avenues
IFN + VBL 66 24 24 N/A and will be covered in other chapters. Other trials have
IFN #263 82 12 N/A demonstrated clinical benefit from interferon therapy in
IFN + VBL 83 8 N/A patients with carcinoid tumors [163, 178]. Improvement
MEGACE 176 7 24 32 13
IFN #261 174 14 36 43 22
in symptomatology associated with a decrease in
IFN #269 139 6 88 22 10 5-hydroxyindoleacetic acid levels has been reported in
IFN + 145 12 128 28 19 approximately 50% of patients but objective regression
Cisretinoicacid is much less frequent.
IFN #275 123 4 34 N/A In addition trials using interferon with other agents
IFN + 123 4 50
Nephrectomy such as octreotride have indicated that combined treat-
IFN #267 147 8 56 12 ment may improve efficacy [67]. The role of other tar-
IFN + IL-2 140 19 72 20 geted agents such as bevacizumab is being explored and
MEAN IFN 11 36 combination with these VEGF targeting agents may
Response need to be explored. There has also been encouraging
290 Interferons: therapy for cancer

results using interferon with 13-cis retinoic acid as bio- but independent of, 2,5 A synthetase [230]. Whether
adjuvant therapy in locally advanced head and neck these are, in fact, central to antiproliferative as well as
squamous cell carcinoma following experimental evi- antiviral activity remains speculative [260].
dence that these agents may work synergistically in this Because of its high degree of sensitivity to the Type I
tumour [141, 233]. interferons, HCL would appear to be an ideal disease in
Once again the positive results with cetuximab and which to determine a mechanism of action for interferon’s
chemotherapy may overtake this work (review else- antitumor effect. Type I interferon receptors are present on
where). Anti-tumor activity has also been demonstrated hairy cells and are down regulated with therapy [13, 59].
with a similar drug combination in patients with A lack of demonstrable down regulation in vivo has
squamous cell carcinoma of the cervix [142]. Although been associated with lack of response [14]. Immunologic
it is relatively ineffective in patients previously treated recovery as manifested by a return of NK activity and
with radiation therapy [278]. normalization of T and B cell counts has been docu-
mented [175] but responses occur without improvement
in NK activity [70,71]. Further, hairy cells were not sus-
ceptible to NK cytotoxicity in vitro [235]. It has also
Mode of Action been shown that alpha interferon, when cultured with
Alpha interferon has an antiproliferative effect, antiviral peripheral blood mononuclear cells of patients with
effect, an immune augmenting effect, and a differentiation HCL, gives rise to multi-lineage colonies composed of
effect and an anti-angiogenic effect. Any one or combina- myeloid and erythroid progenitors – evidence for the
tion of these effects may induce an anti-tumor response. existence of circulating hematopoietic stem cells respon-
Gamma interferon (Type II IFN) is a more potent sive to a differentiation effect of interferon [162].
immune stimulator of monocyte function and class II It has also been shown that the mononuclear cells of
HLA activity than are either alpha or beta interferon HCL are defective in their ability to release tumor necro-
(Type I IFNs) [269]; however, on a weight basis, the sis factor (TNF) and that interferon overcomes this –
Type I IFNs are ten times as potent as gamma interferon perhaps interferon and TNF then act synergistically as
in their antiviral effect. antiproliferative or cytotoxic agents [2, 3]. The low dose
– standard dose randomized study with IFN alpha N-1
in patients with HCL supports both of these theories;
Antiproliferative both doses were comparable in their ability to induce an
In vitro studies and murine models have been used to increase in platelet and neutrophil counts (differentia-
demonstrate the anti-proliferative action of interferons tion) while the standard dose was more effective as an
[87, 262]. Decreased tumorigenicity has also been shown antileukemic (antiproliferative) agent [242]. This is the
in cells pretreated with human interferon [17, 87]. only prospectively randomized clinical evidence avail-
Whether these models are applicable to human tumors able to support a dose-response effect. Ford et al. and
in vivo remains conjectural. subsequently Pagannelli et al. have shown that HCL
Cell cycle analysis has shown that interferon causes proliferation in vitro required B cell growth factor
extension of all phases of the cell cycle and prolonga- (BCGF) and Pagannelli et al. have shown that Type I but
tion of the overall cell generation time [121, 194, 252]. not Type II interferon inhibits the effect of BCGF [191,
In some cases, an accumulation of cells in G0 has been 65]. These observations strongly suggest that interfer-
observed, accompanied by a decrease in transition to G1. on’s primary anti-tumor effect is an antiproliferative
This decrease in growth rate may be incompatible with one, at least in this tumor setting. Most recently a down
cell life [262]. The critical antiproliferative mechanisms regulation of telomerase activity in human malignant
operative at the cellular level have not been elucidated, heamatopoietic cell lines may have identified a novel
but it is possible that they may be mediated by the same antiproliferative mechanism [286].
inhibitors of DNA and RNA synthesis that are found in
virus-infected cells.
Specifically, induction of 2′5′-oligoadenylate (2,5 A)
Antiviral Activity
synthetase leads to endoribonuclease activation, which That interferons have antiviral properties is well estab-
in turn inhibits RNA transcription by degrading mRNA lished and alpha interferon is approved in many coun-
linked to dsRNA [213, 230]. In addition, a protein kinase tries for the treatment of several forms of viral hepatitis.
and a phosphodiesterase pathway represent mechanisms Whether this effect is the basis for interferon’s antitu-
of inhibition of protein synthesis which are parallel to, mor effect in tumors possibly related to viral etiologies,
D. Goldstein et al. 291

such as Kaposi’s sarcoma associated with AIDS and feron. However, intravenous administration was used for
ATLL, is unknown. the high dose, and intramuscular administration for the
Although other tumors may be of viral origin, most low dose, and the schedule of therapy was also different
prominently cervical carcinoma, there has not been a (alternate weeks for low-dose versus monthly for high-
major investigative effort mounted in this area and cur- dose). Both uncontrolled factors, i.e., route of adminis-
rently the antiviral effect of alpha interferon remains tration and schedule, might have caused the differences
unexploited in the treatment of malignancy. noted. An earlier study with lymphoblastoid interferon
showed a dose-related decrease in NK activity over a
6-week period, after an initial stimulatory effect; but in
Immune Modulation that instance the treatment was given three times a week
Support for immune stimulation as a mechanism of [134]. This suggests that an intermittent schedule may
action is found in experiments with animals, in which result in different patterns of response.
tumors known to be interferon-resistant in vitro have Since interferon consistently stimulates NK-cells in
regressed when the animal received systemic treatment vitro, the inability to reproduce consistent effects of
with interferon [42, 87]. A variety of immune changes interferon on NK-cell activity in clinical practice sug-
have been described, but the most relevant appear to gests a need to explore confounding in vivo effects, such
be the effects on natural killer (NK)-cells and as scheduling, dose, or sampling method (peripheral
macrophages. blood versus tumor site). Furthermore, both Type I and
In vitro work with human lymphocytes shows that Type II interferons have been shown to induce resis-
interferon increases the cytotoxic potential of NK-cells tance to NK activity in vitro. The controversy surrounding
by recruitment of pre-NK-cells, increasing activity of NK-cell function after interferon treatment suggests that
activated cells, and by augmenting NK-cell-mediated it may not be a useful immunologic marker.
antibody-dependent cell-mediated cytotoxicity [100, Macrophages, more consistently than NK-cells, are
194]. Alpha and beta interferons also appear to cause NK affected by gamma interferon; indeed, gamma inter-
activation at a lower dose and over a shorter time interval feron has all the activities of a macrophage-activation
than does gamma interferon [263]. In clinical trials, both factor (MAF) [42, 269]. Gamma interferon acts to
the dose and route of interferon administration have been increase the number and density of Fc receptors, which
shown to effect NK-cell activity significantly. in turn are associated with an increase in antigen-pre-
Several trials have suggested that low-dose inter- senting function. Enhancements of phagocytosis and
feron results in more marked NK-cell activity [51, 134, also of antiviral activity and cytotoxicity occur [194,
47, 48]. This is a confusing issue, however, since oth- 252]. Thus, assays of macrophage/monocyte function
ers have reported widely differing effects on NK-cell may be of more relevance for assessment of gamma
activity, varying from a consistent increase [52, 202, interferon action, whereas NK or antiviral activity may
145] to a consistent decrease [143, 148, 149, 149, 247] be the best choice for alpha or beta interferons.
in addition to individual variation without discernable In addition to functional studies, changes in serum
pattern [170]. Much of the controversy may be related molecules, particularly neopterin, tryptophan and beta 2
to the dose of interferon used. Unpublished work from microglobulin have been found to be very consistent
the NCI by Varesio et al. [267] has suggested a bell- markers of biologic response to interferons. Their rela-
shaped curve for both the antiproliferative effect and tionship to the degree of immune modulation in vivo is
immune augmentation. These curves do not precisely unclear but recent studies with alpha, beta and gamma
overlap. interferons have shown the optimal biologic dose to be
A few clinical studies have examined the long-term well below the maximum tolerated dose [71, 72, 81].
immunologic effects of alpha interferon treatment; The ability to increase cell-surface antigen expression
again, these differ with respect to whether there is an presents another aspect of interferon action that may be
increase [134, 237] or decrease [148, 149, 150] in NK important in tumor control. Augmentation has been
activity. A study by Silver et al. [237] (p. 175) showed demonstrated for the expression of Fc receptors on
that low dose alpha interferon led to an increase in NK lymphocytes and of Class I and II (Ia) MHC antigens
activity within 48 h, which was, however, not sustained [42, 194, 252] on several other cell types as well.
despite continued administration. In addition to augmentation of expression, interferon
Repeated high-dose interferon gave a more sustained has also been shown to induce HLA expression in a
increase of NK activity in the long term, even though it variety of normal cells [219]. In neoplastic cells, inter-
did not lead to the same initial rise as low-dose inter- feron has been shown to induce HLA antigen expression
292 Interferons: therapy for cancer

and augment tumor-associated antigen (TAA) expres- 3T3-Li cells into adipocytes and of human monocytes to
sion in several cell lines [74, 86, 75, 23]. This work has macrophages has been inhibited [219, 252].
been expanded and augmentation has been demon- Other evidence supporting an effect on differentiation
strated in vitro in tumor cells obtained from malignant includes an increased expression of HLA antigens,
effusions and following the intraperitoneal administra- enhanced excitability of nerve cells, and decreased beat-
tion of gamma interferon to patients [81, 93]. ing frequency of myocardial cells after interferon treat-
The increased expression of TAA occurring concur- ment [219]. Interestingly, sodium butyrate, a well known
rently with enhancement of macrophage antigen-pre- differentiating agent, enhances this effect of interferons in
senting function, may improve the endogenous antitumor vitro [219]. An important finding – which may suggest
activity of macrophages. one cellular mechanism for inducing differentiation – is
The actions of interferon in promoting tumor antigen the evidence of decreased c-myc and c-Ha-ras gene
expression suggest another major line of inquiry, namely, expression following interferon treatment, with as much
the combination of interferon with monoclonal antibody as a 60% decrease in mRNA formation [31]. With 3T3
(MoAb) therapy, with or without linkage to a toxin [218]. fibroblasts, this decrease in mRNA has been associated
One of the major stumbling blocks to MoAb therapy has with a reversion to normal cell type. Thus, interferon-
been modulation or poor expression of tumor antigens; if induced differentiation appears to be a significant effect
pretreatment with interferon can increase antigen expres- and may be part of the antineoplastic action of interferon.
sion, then the effectiveness of subsequent MoAb treat-
ment might be greatly enhanced. However, the risk of
similar expression in nontarget areas must be kept in
Anti-Angiogenesis
mind, since it is often the level of expression rather than Inhibition of experimental angiogenesis by interferons
the novel nature of the antigen which characterizes the was first demonstrated in a mouse tumor model [234].
tumor cell. At least one early trial has demonstrated This observation was confirmed in infants with life-
potential therapeutic effect [277]. threatening hemangiomas [57]. Hemangioma regression
The group at Stanford University and IDEC Pharma- and significant clinical benefit was demonstrated in
ceutical Corporation in California has performed an more than 80% of seriously effected infants.
interesting trial. These investigators previously reported Successful angiogenesis is essential for the expansion
the use of anti-idiotype antibodies in patients with B cell of any malignant tumour. It is a complex process involv-
lymphomas, obtaining a response in five of ten patients ing extensive interplay between cells, soluble factors
including 1 CR [158, 159]. Patients ultimately failed and extracellular matrix component. The mechanism
because of the emergence of a dominant clone of anti- through which interferons mediate this inhibition is
gen (idiotype) negative cells [158, 159]. An animal unclear but inhibition fibroblast growth factor (FGF)
model was developed to evaluate therapeutic modalities pathways appears to play some part. For example IFN-
in this setting. Using this model, it was shown that a alpha can inhibit FGF induced endothelial cell prolifera-
synergistic effect was obtained with the combination of tion and both IFN-alpha and beta can down-regulate the
anti-idiotype antibodies and interferon [11]. rIFN alfa expression of FGF in human carcinoma cells [101, 239].
2a, 12 MU/m2 SC tiw, was combined with anti-idiotype In addition experiments in nude mice have shown that
therapy and administered to twelve patients. IFN-alpha can lower the expression of FGF and this
Responses were obtained in nine (with 2 CR), not sub- correlates with a decreased blood vessel density and an
stantially different from the results with anti-idiotype inhibition of growth of ectopically implanted tumours
therapy alone. Interferon failed to prevent the emergence in these animals [44].
of idiotype negative clones. Currently ongoing studies In the clinical setting interferons have been shown to
evaluating the combination of alpha interferon and anti be effective in producing antiangiogenic effects in a
CD-20 monoclonal antibody may be clinically more number of tumours. These include Kaposi’s sarcoma
encouraging [41]. [56], bladder carcinoma [248] and giant cell tumour of
the mandible [116]. However the intense interest in
angiogenesis has lead to the development of a number
Differentiation of newer antiagiogenic agents [139]. Although a num-
Interferons have been shown to have a variety of effects on ber of these have become standard of care, the pleiotro-
differentiation. In vitro differentiation has been enhanced pic activities of interferon in combination may still need
in mouse myeloid leukemia cells and in erythroblasts of to be understood to add to the overall antineoplastic
the Friend leukemia system, whereas conversion of mouse effect of some targeted agents [287]
D. Goldstein et al. 293

Side Effects reported to have neutralizing antibody [109]. Antibody


formation is a significantly less frequent phenomenon in
While side effects of the Interferons can be debilitating, patients treated with either rIFN alfa 2b or IFN alfa N1
most appear to be reversible upon cessation of therapy. [251, 271, 272]. Of the three allelic genes used in the
As one might expect, high doses give rise to more severe development of recombinant interferon alfa 2, the gene
manifestations than low doses. The major documented used to produce rIFN alfa 2b is by far the more common
short term side effects are fevers, headache, and myal- allele in North American individuals [138].
gia, while fatigue, a long term side effect, can be severe
enough to be dose-limiting.
Gastrointestinal side effects, in particular, anorexia,
nausea, vomiting and/or diarrhea, are neither universal
Summary
nor severe, but may lead to weight loss, which can be This review of the clinical effects of the interferons indi-
profound with high-dose or prolonged moderate-dose cates that alpha interferon, when used as a single agent,
administration. Elevated serum transaminase (but not has antitumor effects in a large number of malignancies,
clinical hepatitis) has also been noted [134, 245, 237]. perhaps more than any single chemotherapeutic agent
Hypotension with higher doses of both alpha and gamma does. However, like many cytotoxic agents, the use of
interferons can be dose limiting. Both granulocytopenia interferons in combination with cytotoxic drugs, other
and thrombocytopenia have occurred, but are rapidly biologics, or antiviral agents remains under explored.
reversible [237, 15] and rarely dose limiting unless the Clinically, the interferons proved to be a significant addi-
patient has had extensive prior radiation. Margination of tion to our therapeutic armamentarium and served well
leukocytes rather than direct marrow suppression as the prototype for subsequent more targeted biological
appears responsible. therapeutics. Interferon has overcome two misleading
Central nervous system toxicities, including paresthe- labels at opposite ends of the spectrum: of “imaginon”
sias, weakness, somnolence, decreased attention span, when most scientists doubted its existence, and more
short-term memory impairment, confusion, depression, recently that of “magic bullet,” when the public was led
personality change, and even coma at very high doses, to believe it might be the cure-all for cancer. In the con-
have all been seen. EEG abnormalities, including a text of cancer treatment, interferons have been the proto-
slowing of the dominant alpha rhythm and diffuse slow type of the so-called “fourth arm” of therapy; surgery,
waves, have been documented [214, 245, 1, 60]. radiotherapy, chemotherapy and biotherapy [184].
The potential of depression to limit beneficial therapy
such as adjuvant treatment of melanoma has led to a
randomised study of paroxetine, which substantially
reduced the incidence from 45% to 11%. Although only References
a small number of patients were studied, its prophylactic 1. Adams, F., J. R. Quesada, et al. (1984). “Neuropsychiatric mani-
festations of human leukocyte interferon therapy in patients with
use may well improve the therapeutic index for higher
cancer.” J Am Med Assoc(252): 938–941.
dose IFN therapy [169]. 2. Aderka, D., Y. Levo, et al. (1987). “Reduced production of tumor
Exacerbation of coronary artery disease has been necrosis factor (TNF) by mononuclear cells of hairy cell leuke-
reported. Although cardiac toxicity may be life-threatening, mia patients and improvement following leukemia therapy.”
serious cardiac toxicity due to interferon is unusual. Cancer(60): 2208–2212.
3. Aderka, D., R. Michalevicz, et al. (1988). “Recombinant inter-
A significant history of coronary artery disease is a rela-
feron alpha-C for advanced hairy cell leukemia.” Cancer(61):
tive contraindication for interferon therapy, particularly 2207–2213.
at high doses. Life-threatening acute pulmonary toxicity, 4. Ahre, A., M. Bjorkholm, et al. (1984). “Human leukocyte inter-
renal failure, and unexplained sudden death have also feron and intermittent high dose melphalan-prednisone adminis-
been noted on rare occasions. tration in the treatment of multiple myeloma. A randomized
clinical trial from the myeloma group of central Sweden.” Cancer
Evidence has increased indicating that long term Treat Rep(68): 1331–1338.
treatment with interferons results in the production of 5. Ajani, J., A. H. Rios, et al. (1989). “Phase I and II Studies of
antibodies to interferon. In a study of 51 patients with the combination of recombinant human interferon-alpha and
hairy cell leukemia, neutralizing antibody was detected 5-fluorouracil in patients with advanced colorectal carcinoma.”
in 16 patients, six of whom developed some degree of J of Biol Resp Modif(8): 14–46.
6. Allan, N. C., S. M. Richards, et al. (1995). “On behalf of the UK
clinical resistance [250]. Medical Research Council’s Working Parties for Therapeutic Trials
In a previous review of more than 617 patients who in Adult Leukaemia – UK Medical Research Council randomized
received intramuscular interferon alfa 2a, 25% were multicentre trial of interferon alpha N1 for chronic myeloid leukaemia:
294 Interferons: therapy for cancer

Improved survival irrespective of cytogenetic response.” Lancet 25. Case, D. C. J., H. L. Sonneborn, et al. (1986). “Phase II study of
1392–1397. rDNA alpha-2 interferon (Intron A) in patients with multiple
7. Allen, M. J., M. Vaughan, et al. (2000). “Protracted venous infu- myeloma utilizing an escalating induction phase.” Cancer Treat
sion 5-fluorouracil in combination with subcutaneous interleukin-2 Report(70): 1251–1254.
and alpha-interferon in patients with metastatic renal cell cancer: a 26. Cheson, J. M., D. A. Sorensen, et al. (1998). “Treatment of hairy
phase II study.” Br J Cancer(83): 980–985. cell leukemia with 2-chlorodeoxyadenosine via the Group C
8. Andersen, J. and R. V. Smalley (1993). “Interferon alfa plus che- protocol mechanism of the National Cancer Institute: a report of
motherapy for non-Hodgkin’s lymphoma: Five-year follow up.” 979 patients BD.” J Clin Oncol(16): 3007–3015.
New England J Med(329): 1821–1822. 27. Childs, R., A. Chernoff, et al. (2000). “Regression of metastatic
9. Armitage, J. O. and B. Coiffier (2000). “Activity of interferon renal cell carcinoma after non-myeloablative allogeneic peripheral
alpha in relapsed patients with diffuse large B-cell and peripheral blood stem cell transplantation.” N Engl J Med(343): 750–758.
T-cell Non-Hodgkin’s Lymphom.” Ann Oncology(11): 359–361. 28. Chisesi, T., G. Capnist, et al. (1987). “Interferon alfa-2b and
10. Aviles, A., S. Cleto, et al. (2001). “Interferon alfa 2b as mainte- chlorambucil in the treatment of non-Hodgkin’s lymphoma.”
nance therapy in poor risk diffuse large B-cell lymphoma in com- Invest New Drugs(5): 535–540.
plete remission after intensive CHOP-BLEO regimens.” Eur J 29. Clark, R. H., N. V. Dimitrov, et al. (1984). “Leukocyte interferon as
Haematol(66): 94–99. a possible biological response modifier in lymphoproliferative dis-
11. Basham, T. Y., M. Kaminski, S., et al. (1986). “Synergistic antitumor orders resistant to standard therapy.” J Biol Resp Mod(3): 613–619.
effect of interferon and anti-idiotype monoclonal antibody in 30. Clark, R. H., N. V. Dimitrov, et al. (1989). “A phase II trial of
murine lymphoma.” J Immunol(137): 3019. intermittent leukocyte interferon and high dose chlorambucil in
12. Bergsagel, D. E., R. H. Haas, et al. (1987). “Interferon alfa-2b in the treatment of non-Hodgkin’s lymphoma resistant to conven-
the treatment of chronic granulocytic leukemia.” Invest New tional therapy.” Am J Clin Oncol(12): 75–77.
Drugs(5): 9–17. 31. Clemens, M. (1985). “Interferons and oncogenes.” Nature(313):
13. Billard, C., F. Sigaux, et al. (1986). “Treatment of hairy cell leukemia 531–532.
with recombinant alpha interferon: II. In vivo down-regulation of 32. Cole, B. F., P. Solal-Celigny, et al. (1988). “Quality-of-life-adjusted
alpha interferon receptors on tumor cells.” Blood(67): 821–826. survival analysis of interferon alfa-2b treatment for advanced
14. Billard, C., R. A. Diez, et al. (1989). “Lack of in vivo down- follicular lymphoma: an aid to clinical decision making.” J Clin
regulation of interferon alpha receptors in non-Hodgkin lymphoma Oncol(16): 2339–2344.
cells from patients unresponsive to interferon-alpha therapy.” 33. Cole, B. F., R. D. Gelber, et al. (1996). “Quality-of-life-adjusted
J Interf Res(9(Suppl 2) ): S202. survival analysis of interferon alpha-2b adjuvant treatment of high
15. Bjorkstrand, B., H. Svensson, et al. (2001). “Alpha-interferon risk resected cutaneous melanoma: An Eastern Cooperative
maintenance treatment is associated with improved survival after Oncology Group study.” J Clin Oncol(14): 2666.
high-dose treatment and autologous stem cell transplantation in 34. Collaborators, M. R. C. R. C. (1999). “Interferon-alpha and sur-
patients with myeloma: a retrospective registry study from the vival in metastatic renal carcinoma: Early results of a randomized
European group for blood and marrow transplantation.” Bone controlled trial.” Lancet(353): 14–17.
Marrow Transplant (EBMT)(27): 511–515. 35. Cooper, M. R. (1988). “Interferons in the management of multiple
16. Blade, J., J. F. San Miguel, et al. (1998). “Maintenance treatment myeloma.” Sem Oncol(15): 21–25.
with interferon alpha-2b in multiple myeloma; a prospective ran- 36. Cooper, M. R., A. Fefer, et al. (1986). “Alpha-2-interferon/mel-
domised study from PETHEMA.” Leukemia(12): 1144–1148. phalan/prednisolone in previously untreated patients with multiple
17. Borden, E. (1979). “Interferons, rationale for clinical trials in neo- myeloma: A Phase I-II trial.” Cancer Treat Report(70): 473–476.
plastic disease.” Ann Intern Med(91): 472–479. 37. Costanzi, J. J. and R. B. Pollard (1987). “The use of interferon in
18. Borden, E. C., J. F. Holland, et al. (1982). “Leukocyte-derived the treatment of multiple myeloma.” Sem Oncol(14): 24–28.
interferon (alpha) in human breast carcinoma. The American 38. Costanzi, J., J, M. R. Cooper, et al. (1985). “Phase II study of
Cancer Society Phase II trial.” Ann Intern Med(97): 1–6. recombinant alpha 2 interferon in resistant multiple myeloma.” J
19. Borden, E. C., G. C. Sen, et al. (2007). “Interferons at age 50: past, Clin Oncol(3): 654–659.
current and future impact on biomedicine.” Nat Rev Drug 39. Creagan, E. T., D. L. Ahmann, et al. (1984). “Phase II study of
Discov(6(12) ): 975–990. recombinant leukocyte A interferon (RIFN-Alpha-A) in dissemi-
20. Browman, G. P., D. Bergsagel, et al. (1995). “Randomized trial of nated malignant melanoma.” Cancer 1(54): 2844–2849.
interferon maintenance in multiple myeloma:A study of the 40.Cunningham, D., R. Powles, et al. (1998). “A randomised trial of mainte-
National Cancer Institute of Canada Clinical Trials Group.” J Clin nance interferon following high-dose chemotherapy in multiple
Oncol(13): 2354–2360. myeloma; long term follow-up results.” Br J Haematol(102): 495–502.
21. Bunn, P. A., K. A. Foon, et al. (1984). “Recombinant leukocyte A 41. Davis, T. A., G. Maloney, et al. (2000). “Combination Immunotherapy
interferon: An active agent in advanced cutaneous T-cell lymphomas.” of Relapsed or Refractory Low-Grade or Follicular Non-Hodgkin’s
Ann Intern Med(101): 484–487. Lymphoma with Rituximab and Interferon – 2a.” Clin Cancer Res
22. Bunn, P. A. J., D. C. Ihde, et al. (1989). “The role of recombinant (6): 2644–2652.
interferon alfa-2a in the therapy of cutaneous T-cell lymphoma.” 42. DeMaeyer-Guignard,J.andE.DeMaeyer(1985).“Immunomodulation
Cancer(57): 1689–1695. by interferons: Recent developments.” Academic Press(6): 69–86.
23. Carrel, S., A. Schmidt-Kessen, et al. (1985). “Recombinant inter- 43. Dewit, R., C. A. B. Boucher, et al. (1988). “Clinical and virological
feron-gamma can induce the expression of HLA-DR and DC-or effects of high-dose recombinant interferon alpha in disseminated
DR-negative melanoma cells and enhance the expression of HLA- AIDS-related Kaposi’s sarcoma.” Lancet(2): 1214–1217.
ABC and tumor-associated antigen.” Eur J Immunol(15): 118–123. 44. Dinney, C. P. N., D. R. Bielenberg, et al. (1998). “Inhibition of
24. Cascinelli, N. (1995). “Evaluation of efficacy of adjuvant rIFNa basic fibrobroblast growth factor expression, angiogenesis and
2A in melanoma patients with regional node metastases.” Proc Am growth of human bladder carcinoma in mice by systemic inter-
Soc Clin Oncol(14): 1296. feron alpha administration.” Cancer Res(58): 808.
D. Goldstein et al. 295

45. Druker, B. J., M. Talpaz, et al. (2001). “Efficacy and safety of a 63. Flanigan, C., B. A. Blumnetsein, et al. (2000). “Cytoreduction
specific inhibitor of the BCR-ABL tyrosine kinase in chronic nephrectomy in metastatic renal cancer: The results of South west
myeloid leukemia.” N Eng J Med(344): 1031–1037. Oncology group trial 8949.” Proc Am Soc Clin Oncol(19): 2a.
46. Dutcher, J. P., T. Logan, et al. (2000). “Phase II trial of interleukin 2, 64. Foon, K. A., S. A. Sherwin, et al. (1984). “Treatment of advanced
interferon alpha and 5-fluorouracil in metastatic renal cell cancer: non-Hodgkin’s lymphoma with recombinant leukocyte A inter-
a cytokine working group study.” Clin Cancer Res September feron.” N Engl J Med(311): 1148–1152.
2000 (6(9) ): 3442–3450. 65. Ford, R. J., D. Kwok, et al. (1986). “Production of B cell growth
47. Edwards, B. S., M. J. Hawkins, et al. (1984). “Comparative in vivo and factor(s) by neoplastic B cells from hairy cell leukemia patients.”
in vitro activation of human NK cells by two recombinant interferons Blood(67): 573–577.
alpha differing in anti-viral activity.” Cancer Res(44): 3135–3139. 66. Fosså, S., G. Martinelli, et al. (1992). “Recombinant interferon-alpha-2a
48. Edwards, B. S., J. A. Merritt, et al. (1985). “Low doses of inter- with or without vinblastine in metastatic renal cell carcinoma: Results of
feron alpha result in more effective clinical natural killer cell a European multi-centre phase III study.” Ann Oncol(3): 301–305.
activation.” J Clin Invest(75): 1908–1913. 67. Frank, M., K. J. Klose, et al. (1999). ”Combination therapy with
49a. Eggermont, A. M., S. Suciu, et al. (2005). “Post-surgery adjuvant octreotride and alpha-interferon: effect on tumour growth in meta-
therapy with intermediate doses of interferon alfa 2b versus obser- static endocrine gastropancreatic tumours.” Am J Gastroenerol(94):
vation in patients with stage IIb/III melanoma (EORTC 18952): 1381–1387.
randomised controlled trial.” Lancet(366(9492) ): 1189–1196. 68. Gams, R., D. Gordon, et al. (1984). “Phase II trial of human poly-
49b. Eggermont, A. M., S. Suciu, M. Santinami, et al. (2008). “Adjuvant clonal lymphoblastoid interferon in the management of malignant
therapy with pegylated interferon-2b versus observation alone in lymphomas.” Proc Am Soc Clin Oncology(3): 65.
resected stage III melanoma: final results of EORTC 18991, a ran- 69. Gastl, G., H. Denz, et al. (1985). “Treatment with low dose human
domised phase III trial.” Lancet(12): 372 (9633): 117–126. recombinant interferon-alpha-2 ARG induces complete remission
50. Einhorn, N., K. Cantel, et al. (1982). “Human leukocyte inter- in patients with hairy cell leukemia.” Onkologie(8): 143–144.
feron.” Am J Clin Oncol(5): 167–172. 70. Gastl, G., W. Aulitzky, et al. (1986). “Alpha interferon induces
51. Einhorn, S., A. Ahre, et al. (1982). “Interferon and natural killer remission in hairy cell leukemia without enhancement of natural
activity in multiple myeloma. Lack of correlation between inter- killing.” Blut(52): 273–279.
feron-induced enhancement of natural killer activity and clinical 71. Gastl, G., W. Aulitzky, et al. (1986). “A biological approach
response to human interferon-alpha.” Int J Cancer 30(I): 167–172. to optimize interferon treatment in hairy cell leukemia.”
52. Einhorn, S., H. Blomgren, et al. (1978). “Interferon and spontane- Immunobiology(172): 262–268.
ous cytotoxicity in man.” Acta Med Scand(204): 477–483. 72. Gastl, G., M. Werter, et al. (1989). “Comparison of clinical efficacy
53. Elhilali, M. M., M. Gleave, et al. (2000). “Placebo-associated and toxicity of conventional and optimum biological response modify-
remissions in a multicentre, randomized, double-blind trial of ing doses of interferon alpha-2C in the treatment of hairy cell leukemia:
interferon gamma-1b for the treatment of metastatic renal cell car- A retrospective analysis of 39 patients.” Leukemia(3): 453–460.
cinoma.” BJU Int 2000 (86(6) ): 613–618. 73. Gelmann, E. P., O. T. Preble, et al. (1985). “Human lymphoblas-
54. Escudier, B., A. Pluzanska, et al. (2007). “Bevacizumab plus interferon toid interferon treatment of Kaposi’s sarcoma in the acquired
alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, immune deficiency syndrome.” Am J Med(78): 737–741.
double-blind phase III trial.” Lancet(370(9605) ): 2103–2111. 74. Giacomini, P., A. Aguzzi, et al. (1984). “Modulation by recombi-
55. Eton, O., S. Legha, et al. (2000). “Phase III randomized trial of nant DNA leukocyte (a) and fibroblast (b) interferons of the
cisplatin vinblastine and dacarbazine (CVD) plus interleukin-2 expression and shedding of HLA- and tumor-associated antigens
(IL2) and interferon-alpha-2b (INF) versus CVD in patients with by human melanoma cells.” J Immunol(133): 1649–1655.
metastatic melanoma. 2000.” Proc Am Soc Clin Onco(l 19): 2174. 75. Giacomini, P., L. Imberti, et al. (1985). “Immunochemical analysis
56. Evans, L. M., L. M. Itri, et al. (1989). “Alfa interferon 2a in the treat- of the modulation of human melanoma-associated antigens by DNA
ment of AIDS related Kaposi’s sarcoma.” J Interf Res(9): S207. recombinant immune interferon.” J Immunol(135): 2887–2894.
57. Ezekowitz, R. A. B., J. B. Mulliken, et al. (1992). “Interferon alfa- 76. Gibson, J., K. Cameron, et al. (1988). “Clinical response of hairy
2a therapy for life-threatening hemangiomas of infancy.” N Eng J cell leukaemia to interferon-α: results of an Australian study.” Med
Med(326): 1456. J of Aust(149): 293–296.
58. Falkson, C. I., J. Ibrahim, et al. (1998). “Phase III trial of dacarba- 77. Gibson, J., K. Gallagher, et al. (1988). “Peripheral blood lymphocyte
zine with tamoxifen in patirnts with metastatic malignant melanoma: subsets and natural killer cell number and function during α-interferon
an Eastern Cooperative Oncology Group study.” J Clin Oncol(16): treatment for hairy cell leukemia.” Aust New Zeal J Med(18): 897–898.
1743–1751. 78. Gill, P. S., W. J. Harrington, et al. (1995). “Treatment of adult
59. Faltynek, C. R., G. L. Princler, et al. (1986). “Relationship of the T-cell leukemia-lymphoma with a combination of interferon alfa
clinical response and binding of recombinant interferon alpha in and zidovudine.” N Engl J Med(332): 1744–1748.
patients with lymphoproliferative diseases.” Blood(67): 1077–1082. 79. Gisslinger, H., H. Ludwig, et al. (1989). “Long-term interferon
60. Farkilla, M., M. Ilvanainen, et al. (1984). “Neurotoxic and other therapy for thrombocytosis in myeloproliferative diseases.”
side effects of high-dose interferon in amyotrophic lateral sclerosis.” Lancet(1): 634–637.
Acta Neurol Scand(69): 42–46. 80. Gleave, M. E., M. Elhilali, et al. (1998). “Interferon gamma-1b
61. Figlin, R. A., J. B. deKernion, et al. (1985). “Treatment of renal cell compared with placebo in metastatic renal cell carcinoma.” New
carcinoma with alpha (human leukocyte) interferon and vinblastine in Engl J Med(338): 1265–1271.
combination: A phase I-II trial.” Cancer Treat Report(69): 263–267. 81. Goldstein, D., F. Guadagni, et al. (1989). “Clinical and biological
62. Fisher, R. I., B. W. Dana, et al. (2000). “Interferon Alfa Consolidation effects of intraperitoneal interferon gamma in a phase 1A/b trial in
After Intensive Chemotherapy Does Not Prolong the Progression- ovarian carcinoma.” J Interf Res(9(2) ): S115.
Free Survival of Patients With Low-Grade Non-Hodgkin’s 82. Golomb, H. M., A. Jacobs, et al. (1986). “Alpha-2 interferon ther-
Lymphoma: Results of the Southwest Oncology Group. Randomized apy of hairy cell leukemia: A multi-center study of 64 patients.”
Phase III Study 8809.” J Clin Oncol (18): 2010–2020. J Clin Oncol(4): 900–905.
296 Interferons: therapy for cancer

83. Golomb, H. M., A. Fefer, et al. (1988). “Report of a multi- 102. Hill, M., A. Norman, et al. (1995). “Royal Marsden phase III trial
institutional study of 193 patients with hairy cell leukemia of fluorouracil with or without interferon alfa 2b in advanced col-
treated with interferon-alfa 2b.” Sem in Oncol(15): 7–9. orectal cancer.” J Clin Onc(13): 1297–1302.
84. Golomb, H. M., M. J. Ratain, et al. (1988). “Randomized study of 103. Holtl, L., C. Reiser, et al. (1999). “Cellular and humoral immune
the duration of treatment with interferon alfa-2B in patients with responses in patients with metastatic renal cell carcinoma after vacci-
hairy cell leukemia.” J Natl Cancer Inst(80): 369–373. nation with antigen pulsed dendritic cells.” J Urol (161(3)): 777–782.
85. Gore, M. E., M. Mohammed, et al. (2001). “Clinical resistance to 104. Horning, S. J., T. L. Merigan, et al. (1985). “Human interferon
STI-571 cancer therapy by BCR-ABL gene mutation or amplifi- alpha in malignant lymphoma and Hodgkin’s disease.” Cancer
cation.” Science(293): 876–880. Res(56): 1305–1310.
86. Greiner, J. W., P. H. Hand, et al. (1984). “Enhanced expression of 105. Huber, C., Batchelor. J. R., et al. (1984). “Immune response-
surface tumor associated antigen on human breast and colon associated production of neopterin.” (160): 310–316.
tumor cells after recombinant human leukocyte a-interferon treat- 106. Hudes, G., M. Carducci, et al. (2007). “Temsirolimus, interferon
ment.” Cancer Res(44): 3208–3211. alfa, or both for advanced renal-cell carcinoma.” N Engl J
87. Gresser, I. (1985). “How does interferon inhibit tumor growth?” Med(356(22) ): 2271–2278.
Interferon(6): 93–126. 107. Huncharek, M., J. F. Caubet, et al. (2001). “Single agent DTIC
88. Grillo-Lopez, A. J., C. A. White, et al. (2000). “Rituximab: the versus combination therapy with or without immunotherapy in
first monoclonal antibody approved for the treatment of lym- metastatic melanoma: a meta-analysis of 3273 patients from 20
phoma.” Curr Pharm Biotechnol(1(1) ): 1–9. randomized trials.” Melanoma Research(11): 75–81.
89. Grob, J. J., B. Dreno, et al. (1998). “Randomised trial of interferon 108. Isaacs, H. and J. Lindemann (1957). “Virus interference 1. The
alpha-2a as adjuvant treatment in resected primary melanoma thicker interferon.” Proc R Soc Oncol Biol(147): 257–262.
than 1.5 mm without clinically detectable node metastases. French 109. Itri, L. M., M. Campion, et al. (1987). “Incidence and clinical
Co-operative Group on Melanoma.” Lancet(351): 1905–1910. significance of neutralizing antibodies in patients receiving
90. Groopman, J., M. Gottlieb, et al. (1984). “Recombinant alpha-2 recombinant interferon alfa 2a by intramuscular injection.”
interferon therapy for Kaposi’s sarcoma associated with the acquired Cancer(59): 668–674.
immunodeficiency syndrome.” Ann Intern Med(100): 671–676. 110. Ives, N. J., R. Stowe, L., et al. (2007). “Chemotherapy compared
91. Group, N. M. S. (1996). “Interferon-α 2b added to melphalan- with biochemotherapy for the treatment of metastatic melanoma
prednisone for initial and maintenance therapy in multiple meta-analysis of 18 trials involving 2621 patients.” J Cin Onco(1):
myeloma.” Ann Intern Med(124): 212–222. 5426–5434.
92. Group, T. M. T. C. (2001). “Interferon as therapy for multiple 111. Jabbour, E., H. Kantarjian, et al. (2007). “PEG-IFN-alpha-2b
myeloma. An individual patient data overview of 24 randomized therapy in BCR-ABL-negative myeloproliferative disorders: final
trials and 4012 patients.” Br J Haematol(113): 1020–1034. result of a phase 2 study.” Cancer Res(110(9) ): 2012–2018.
93. Guadagni, F., J. Schlom, et al. (1989). “Selective Interferon- 112. Johnston, J. B., E. Eisenhauer, et al. (2000). “Long-term outcome
Induced Enhancement of Tumor-Associated Antigens on a following treatment of hairy cell leukemia with pentostatin; a
Spectrum of Freshly Isolated Human Adenocarcinoma Cells.” J National Cancer Institute of Canada study.” Semin Onco(27
Natl Cancer Inst(81): 502–512. (Suppl. 5) ): 32–36.
94. Gugliotta, L., G. P. Bagnara, et al. (1989). “In vivo and in vitro inhib- 113. Jonasch, E. and F. G. Haluska (2001). “Interferon in Oncological
itory effect of alpha interferon on megakaryocyte colony growth in Practice; Review of Interferon biology, Clinical applications, and
essential thrombocythaemia.” Br J Hematol(71): 177–181. toxicities.” The Oncologist(6): 34–55.
95. Gutterman, J. U., A. R. Blumenschein, et al. (1980). “Leukocyte 114. Joshua, D. E., R. Penny, et al. (1997). “Australian leukemia study
interferon-induced tumor regression in human metastatic breast group myeloma II: A randomised study of intensive combination
cancer, multiple myeloma and malignant lymphoma.” Ann Intern chemotherapy with or without interferon in patients with
Med(93): 399–406. myeloma.” Br J Haematol(97): 38–45.
96. Hagenbeek, A., P. Carde, et al. (1998). “Maintenance of remis- 115. Jumbou, O., J. M. N’Guyen, et al. (1999). “Long-term follow-up
sion with human recombinant interferon alfa-2a in patients with in 51 patients with mycosis fungoides and Sezary syndrome
stages III and IV low-grade malignant non-Hodgkin’s lymphoma. treated by interferon-alfa.” Br J Dermatol(140(3) ): 427–431.
European Organization for Research and Treatment of Cancer 116. Kaban, L. B., J. B. Mulliken, et al. (1999). “Antiangiogenic therapy
Lymphoma Cooperative Group.” J Clin Oncol(16): 148. of a recurrent giant cell tumour of the mandible with interferon
97. Hancock, B. W., K. Wheatley, et al. (2004). “Adjuvant interferon in alpha-2a.” Pediatrics(103): 1145.
high risk melanoma; the AIM high study – UKCCR randomized 117. Kantarajan, H. M., S. O’Brien, et al. (1999). “Treatment of Philadelphia
study of adjuvant low-does extended duration interferon Alfa-2a in chromosome-positive early chronic phase myelogenous leukemias
high risk resected malignant melanoma.” J Clin Oncol(22): 53–61. with daily doses of interferon alpha and low-dose cytarabine.”
98. Hehlmann, R., B. Anger, et al. (1988). “Randomized study on the J Clin Oncol(17): 284–293.
treatment of chronic myeloid leukemia (CML) in chronic phase 118. Kantarjian, H. M., T. L. Smith, et al. (1995). “Prolonged survival
with busulfan versus hydroxyurea versus interferon-alpha.” in chronic myelogenous leukemia after cytogenetic response to
Blut(56): 87–91. interferon alpha therapy.” Ann Int Med(122): 254–261.
99. Hehlmann, R., H. Heimpel, et al. (1994). “Randomized comparison 119. Keiholz, U., S. H. Goey, et al. (1997). “Interferon alpha-2a and
of interferon alpha with Busulfan and hydroxyurea in chronic interleukin-2 with or without cisplatin in metastatic melanoma: a
myelogenous leukemia.” Blood(84): 4064–4077. randomized trial of the European Organisation for Research and
100. Herberman, R. B. and J. R. Ortaldo (1981). “Natural killer cells: Treatment of Cancer Melanoma Cooperative Group.” J Clin
Their role in defenses against diseas.” Science(214): 24–30. Oncol(15): 2579–2588.
101. Heyns, A. P., Eldor. A., et al. (1985). “The antiproliferative effect 120. Kimby, E., J. Jurlander, et al. (2008). “Nordic Lymphoma Group.
of interferon and the mitogenic activity of growth factors are Long-term molecular remissions in patients with indolent lym-
independent cell cycle events.” Exp Cell Res(161): 297. phoma treated with rituximab as a single agent or in combination
D. Goldstein et al. 297

with interferon alpha-2a: a randomized phase II study from the 139. Liekens, S., E. De Clercq, et al. (2001). “Angiogenesis: regula-
Nordic Lymphoma Group.” Leuk Lymphoma(49(1) ): 102–112. tors and clinical applications. Biochemical Pharmacology.”
121. Kirchner, H. (1984). “Interferons, a group of multiple lymphokines.” Biochemical Pharmacology(61): 253–270.
Semin Immunopathol(7): 347–374. 140. Lindsay, K. L., C. Trepo, et al. (2001). “A randomized, double-blind
122. Kirkwood, J. M., M. S. Ernstoff, et al. (1985). “Comparison of trial comparing pegylated interferon alfa-2b to interferon alfa-2b as
intramuscular and intravenous recombinant a-2 interferon in initial treatment for chronic hepatitis C.” Hepatology(34(2) ):
melanoma and other cancers.” Ann Intern Med(103): 32–36. 395–403.
123. Kirkwood, J. M., J. G. Ibrahim, et al. (2000). “High-and low-dose 141. Lingen, M. W., P. J. Polverini, et al. (1998). “Retinoic acid and
interferon alpha-2b in high-risk melanoma: first analysis of inter- interferon alpha act synergistically as antiangiogenic antitumour
group trial E1690/S9111/C9190.” J Clin Oncol(18): 2444–2458. agents against human head and neck squamous cell carcinoma.”
124. Kirkwood, J. M., J. G. Ibrahim, et al. (2001). “High dose inter- Cancer Res(58): 5551–5558.
feron alpha-2b significantly prolongs relapse-free and overall 142. Lippman, S. M., D. R. Parkinson, et al. (1992). “13-cis-retinoic
survival compared with the GM2-KLH/QS-21 vaccine in patients acid plus interferon alfa 2a:Effective combination therapy for
with resected stage IIB-III melanoma: results of intergroup trial advanced squamous cell carcinoma of the cervix.” J Nat Ca
E1694/S9512/C509801.” J Clin Oncol(19): 2370–2380. Inst(84): 235–240.
125. Kirkwood, J. M., M. H. Strawderman, et al. (1996). “Interferon 143. Lotzova, E., C. A. Savary, et al. (1983). “Regulation of natural
alpha-2b adjuvant therapy of high-risk resected cutaneous lym- killer cell cytotoxicity by recombinant leukocyte interferon clone
phoma: the Eastern Cooperative Oncology Group Trial EST A.” J Biol Resp Modif(2): 482–498.
1684.” J Clin Oncol (14): 7–17. 144. Louie, A. C., J. G. Gallagher, et al. (1981). “Followup observa-
126. Kleberg, U. R., S. Suciu, et al. (2004). “Final results of the EORTC tions on the effect of human leukocyte interferon in non-Hodg-
18871/DKG 80–1 randomised phase III trial. rIFN-alpha2b versus kin’s lymphoma.” Blood(58): 712–718.
rIFN-gamma versus ISCADOR M versus observation after sur- 145. Lucero, M. A., H. Magdalenat, et al. (1982). “Comparison of effects
gery in melanoma patients with either high risk primary or regional of leukocyte and fibroblast interferon on immunological parameters
lymph node metastasi.” Eur J Cancer(40): 390–402. in cancer patients.” Eur J Cancer Clin Oncol(18): 243–251.
127. Kluin-Nelemans, H. C., G. Buck, et al. (2004). “Randomised 146. Ludwig, H. and E. Fritz (2000). “Interferon in multiple myeloma-
comparison of low dose versus high dose interferon-alpha in summary of treatment results and clinical implications.” Acta
chronic myeloid leukemia: prospective collaboration of 3 joint Oncol(39): 815–821.
trials by the MRC and hOVON groups.” Blood(103): 4408–4415. 147. Ludwig, H., A. Cortelezzi, et al. (1986). “Recombinant interferon
128. Kreitman, R. J., W. H. Wilson, et al. (2001). “Efficacy of the anti- alfa-2C versus polychemotherapy (VMCP) for treatment of multiple
CD22 recombinant immunotoxin BL22 in chemotherapy resis- myeloma: a prospective randomized trial.” Eur J Ca Clin
tant hairy cell leukemia.” New Engl J Med(345): 241–247. Onc(22): 1111–1116.
129. Krigel, R. L., C. M. Slywotzky, et al. (1988). “Treatment of epi- 148. Maluish, A. E., J. R. Ortaldo, et al. (1983a). “Depression of natural
demic Kaposi’s sarcoma with a combination of interferon-alpha-2b killer cytotoxicity after in vivo administration of recombinant
and etoposide.” J Biol Resp Modif(7): 359–364. leukocyte interferon.” J Immunol(131): 503–507.
130. Krown, S. E. (1987). “The role for interferon in the therapy of 149. Maluish, A. E., J. R. Ortaldo, et al. (1983b). “Changes in
epidemic Kaposi’s sarcoma.” Semin Oncol(2): 27–33. immune function in patients receiving natural leukocyte inter-
131. Kugler, A., G. Stuhler, et al. (2000). “Regression of human meta- feron.” J Biol Resp Modif(2): 418–422.
static renal cell carcinoma after vaccination with tumor cell-dendritic 150. Maluish, A. E., R. Leavitt, et al. (1984). “Effects of recombinant
cell hybrids.” Nat Med(6(3) ): 332–336. interferon-alpha on immune function in cancer patients.” J Biol
132. Kuzel, T. M., K. Gilyon, et al. (1990). “Interferon alfa-2a com- Resp Modif(2): 470–481.
bined with phototherapy in the treatment of cutaneous T-cell lym- 151. Mandelli, F., G. Avvisati, et al. (1990). “Maintenance treatment
phoma.” J Natl Cancer Inst(82): 203–207. with recombinant interferon alpha 2b recombinant in multiple
133. Kuzel, T. M., H. H. Roenigk, et al. (1995). “Effectiveness of myeloma patients responding to conventional induction chemo-
interferon alpha 2a combined with phototherapy for mycosis therapy.” N Engl J Med(322): 1430–1434.
fungoides and the Sézary syndrome.” J Clin Onc(13): 257. 152. Mangia, A., G. L. Ricci, et al. (2005). “A randomized controlled
134. Laszlo, J., A. T. Huang, et al. (1983). “Phase I study of pharma- trial of pegylated interferon alpha-2a (40 KD) or interferon alpha-
cological and immunological effects of human lymphoblastoid 2a plus ribavirin and amantadine vs interferon alpha-2a and bavirin
interferon given to patients with cancer.” Cancer Res(43): in treatment-naïve patients with chronic hepatitis C.” J Viral
4458–4466. Hepat(12(3) ): 292–299.
135. Lazzarino, M., A. Vitalle, et al. (1989). “Interferon alpha-2b as 153. Marotta, G., A. Frassoldati, et al. (2006). “Furthermore role of
treatment for Philadelphia-negative chronic myeloproliferative interferon-alpha administration after 2-deoxycoformycin in the
disorders with excessive thrombocytosis.” Br J Haematol(72): treatment of hairy cell leukemia patient.” Eur J Haematol(77(2) ):
173–177. 109–113.
136. Leavitt, R. D., V. Ratanatharathorn, et al. (1987). “Alfa-2b inter- 154. Marumo, K., M. Murai, et al. (1984). “Human lymphoblas-
feron in the treatment of Hodgkin’s Disease and Non-Hodgkin’s toid interferon therapy for advanced renal cell carcinoma.”
lymphoma.” Sem Oncol(14(Suppl 2) ): 18–23. Urology(24): 567–571.
137. Lenhoff, S., M. Hjorth, et al. (2000). “Impact on survival of high 155. Mauss, S. and H. Jablonowski (1995). “Efficacy, safety, and tol-
dose therapy with autologous stem cell support in patients erance of low-dose, long-term interferon-alpha 2b and zidovu-
younger than 60 years with newly diagnosed multiple myeloma: dine in early-stage AIDS-associated Kaposi’s sarcoma.” J Acquir
a population based study.” Blood(95): 7–11. Immune Defic Syndr Hum Retrovirol(10(2) ): 157–162.
138. Liao, M. J., N. Lee, et al. (1994). “Distribution of interferon alpha 156. McClay, E., M. Mastrangelo, et al. (1992). “Effective com-
2 genes in humans.” J Interferon Res(14): 183–185. bination chemo/hormonal therapy for malignant melanoma:
298 Interferons: therapy for cancer

Experience with three consecutive trials.” Int J Cancer(50): 176. O’Brien, S. G., F. Guilhot, et al. (2003). “Imatinib compared with
553–556. interferon and low-dose cytarabine for newly diagnosed chronic-
157. McLaughlin, P., F. Cabanillan, et al. (1993). “CHOP-Bleo plus phase chronic myeloid leukemia.” N Engl J Med (348(11) ):
interferon for stage IV low-grade lymphoma.” Ann Onc(4): 994–1004.
205–211. 177. O’Connell, M. J., J. P. Colgan, et al. (1986). “Clinical trials of
158. Meeker, T., Lowder. J., et al. (1985). “Emergence of idiotype recombinant leukocyte A interferon as initial therapy for favor-
variants during treatment of B-cell lymphomas with anti-idiotype able histology Non-Hodgkin’s lymphomas and chronic lympho-
antibodies.” N Eng J Med(312): 1658. cytic leukemia. An Eastern Cooperative Oncology Group Pilot
159. Meeker, T. C., J. Lowder, et al. (1985). “A clinical trial of anti- Study.” J Clin Oncol(4): 128–136.
idiotype therapy for B cell malignancy.” Blood(65): 1349. 178. Oberg, K., G. Alm, et al. (1989). “Treatment of malignant carci-
160. Mellstedt, H., A. Ahre, et al. (1979). “Interferon therapy in noid tumors with recombinant interferon alfa-2b:Development of
myelomatosis.” Lancet(1): 245–247. neutralizing IFN antibodies and possible loss of antitumor activity.”
161. Merigan, T. L., K. Sikorak, et al. (1978). “Preliminary observations J Nat Ca Inst(81(7)): 531-535.
on the effect of human leukocyte interferon in non-Hodgkin’s 179. Ohnishi, K., R. Ohno, et al. (1995). “A randomized trial comparing
lymphoma.” N Engl J Med(299): 1449–1453. interferon-α with busulfan for newly diagnosed chronic myelog-
162. Michalevicz, R. and M. Revel (1987). “Interferons regulate the in enous leukemia in chronic phase.” Blood(86): 906–916.
vitro differentiation of multilineage lympho-myeloid stem cells 180. Ohno, R. and K. Kimura (1986). “Treatment of multiple myeloma
in hairy cell leukemia.” Proc Natl Acad Sci(84): 2307–2311. with recombinant interferon alfa-2a.” Cancer(57): 1685–1688.
163. Moertel, C. G., J. Rubin, et al. (1989). “Therapy of metastatic 181. Oken, M. M., R. A. Kyle, et al. (1985). “A Phase II trial of inter-
carcinoid tumor and the malignant carcinoid syndrome with feron alpha-2 (rIFN-a2) in the treatment of resistant multiple
recombinant leukocyte alpha interferon.” J Clin Onc(7): 865. myeloma (MM).” Proc Am Soc Clin Oncol(4): 215.
164. Motzer, R. J., B. A. Murphy, et al. (2000). “Phase III Trial of 182. Oken, M. M., T. Leong, et al. (1999). “The addition of interferon
Interferon Alfa-2a With or Without 13-cis-Retinoic Acid for or high dose cyclophosphamide to standard chemotherapy in the
Patients With Advanced Renal Cell Carcinoma.” J Clin Oncol(18): treatment of patients with multiple myeloma: phase III ECOG
2972–2980. EST 9486.” Cancer(15): 957–963.
165. Motzer, R. J., t. A. Rakhi, et al. (2001). “ Phase I trial of 40-kd 183. Oladipupo-Williams, C. K., I. Svet-Moldavskaya, et al. (1981).
branched pegylated interferon alfa-2a for patients with advanced “Inhibitory effects of human leukocyte and fibroblast interferons
renal cell carcinoma.” J Clin Oncol(19): 1312–1319. on normal and chronic myelogenous leukemic granulocytic
166. Motzer, R. J., A. Rakhit, et al. (2001). “Randomized multicenter progenitor cells.” Oncology(38): 356–360.
phase II trial of subcutaneous recombinant human interleukin-12 184. Oldham, R. K. (1984). “ Biologicals and biologic response mod-
versus interferon-alpha 2a for patients with advanced renal cell ifiers. The fourth modality of cancer therapy.” Cancer Treat
carcinoma.” J Interferon Cytokine Res(21(4) ): 257–263. Report(68): 221–232.
167. Motzer, R. J., T. E. Hutson, et al. (2007). “Sunitinib versus interferon 185. Olsen, E. A., S. T. Rosen, et al. (1989). “Interferon alfa-2a in the
alfa in metastatic renal-cell carcinoma.” N Engl J Med(115–24): treatment of cutaneous T cell lymphoma.” J Am Acad Dermatol(20):
115–124. 395–407.
168. Muss, H. B., C. Welander, et al. (1985). “Interferon and doxorubi- 186. Opravil, M., B. Hirschel, et al. (1999). “Related Articles. A ran-
cin in renal cell carcinoma.” Cancer Treat Report(69): 721–722. domized trial of interferon-alpha2a and zidovudine versus bleo-
169. Musselman, D., D. H. Lawson, et al. (2001). “Paroxetine for the mycin and zidovudine for AIDS-related Kaposi’s sarcoma. Swiss
prevention of depression induced by high-Dose interferon Alfa.” HIV Cohort Study.” Int J STD AIDS(10(6) ): 369–375.
New Engl J Med (344): 961–966. 187. Osserman, E. T., W. F. Sherman, et al. (1980). “Preliminary
170. Neefe, J. R., J. E. Sullivan, et al. (1985). “Augmented immunity results of the American Cancer Society-sponsored trial of human
in cancer patients treated with alpha-interferon.” Cancer Res(45): leukocyte interferon in multiple myeloma.” Proc Am Assoc
874–878. Cancer Res(21): 161.
171. Negrier, S., B. Escudier, et al. (1998). “Recombinant human inter- 188. Österborg, A., M. Björkholm, et al. (1993). “Natural interferon-
leukin-2, recombinant human interferon alfa-2a, or both in meta- alpha in combination with melphalan/prednisone versus mel-
static renal-cell carcinoma.” N Engl J Med(338): 1273–1278. phalan/prednisone in the treatment of multiple myeloma stag.es
172. Negrier, S., D. Perol, et al. (2007). “For The French Immuno- II and III. A randomized study from the Myeloma Group of
therapy Intergroup. Medroxyprogesterone, interferon alfa-2a, Central Sweden.” Blood(81): 1428–1434.
interleukin 2, or combination of both cytokines in patients 189. Ozer, H., J. R. Anderson, et al. (1987). “Combination trial of subcu-
with metastatic renal carcinoma of intermediate prognosis: taneous interferon alfa-2b and oral cyclophosphamide in favorable
results of a randomized controlled trial.” Cancer Res(110(11) ): histology, non-Hodgkin’s lymphoma.” Invest New Drugs(5):
2468–2477. 527–523.
173. Neidhart, J. A., S. A. Anderson, et al. (1991). “Vinblastine Fails 190. Ozer, H., H. M. Golomb, et al. (1989). “Cost benefit analysis of
to Improve Response of Renal Cancer to Interferon Alfa-n1 interferon alfa 2-b in the treatment of hairy cell leukemia.” J Natl
(Wellferon); High Response Rate in Patients with Pulmonary Cancer Inst(8): 594–602.
Metastases.” J Clin Onc(9): 832–837. 191. Pagannelli, K. A., S. S. Evans, et al. (1986). “B cell growth factor-
174. Neuman, H. A. and A. A. Fauser(1982). “Effect of interferon on induced proliferation of hairy cell lymphocytes and inhibition by
pluripotent hemopoietic progenitors(CFU-GEMM) derived from Type I interferon in vitro.” Blood(67): 937–942.
human bone marrow.” Exp Hematol(10): 587. 192. Panem, S. (1985). “The Interferon Crusade.” The Brookings
175. Nielsen, B., M. Hokland, et al. (1989). “Immunological recovery Institute.
and dose evaluation in IFN-a treatment of hairy cell leukemia: 193. Pangalis, G. A. and E. Griva (1988). “Recombinant alfa-2b-inter-
Analysis of leukocyte differentiation antigens, NK and 2′,5′-oli- feron therapy in untreated, stages A and B chronic lymphocytic
goadenylate synthetase activity.” Eur J Haematol(42): 50–59. leukemia.” Cancer(61): 869–872.
D. Goldstein et al. 299

194. Paulnock, D. M. and E. C. Borden (1985). “ Modulation of immune 214. Rohatiner, A. Z. S., P. F. Prior, et al. (1983). “Central nervous
functions by interferons. In: Immunity to Cancer” 545–559. system toxicity of interferon.” Br J Cancer(47): 419–422.
195. Peest, D., H. Deicher, et al. (1995). “A comparison of polychemo- 215. Rohatiner, A. Z. S., et al. (1998). Proc AM Soc Clin Oncol(17): 11.
therapy and melphalan/prednisone for primary remission induc- 216. Rohatiner, A., C. G. A. Price, et al. (2001). “A randomized con-
tion, and interferon-alpha for maintenance treatment, in multiple trolled trial to evaluate the role of interferon as initial and main-
myeloma. A prospective trial of the German Myeloma Treatment tenance therapy in patients with follicular lymphoma.” Br J
Grou.” Eur J Cancer Clin Oncol(31A): 146–151. Cancer(85(1)): 29–35.
196. Pehamberger, H., H. P. Soyer, et al. (1998). “Adjuvant interferon 217. Rosenberg, S. A., J. C. Yang, et al. (1999). “Prospective random-
alpha-2a in resected primary stage II cutaneous melanoma.” J Clin ized trial of the treatment of patients with metastatic melanoma
Oncol(16): 1425–1429. using chemotherapy with cisplatin, dacarbazine, and tamoxifen
197. Peterson, B. A., G. Petroni, et al. (1997). “Cyclophosphamide vs. alone or in combination with interleukin-2 and interferon alpha-2b.”
Cyclophosphamide plus interferon alfa-2b in follicular low grade J Clin Oncol(17): 968–975.
lymphomas.” Proc Am Soc Clin Oncol(16): 41–47. 218. Rosenblum, M. G., B. L. Maxwell, et al. (1986). “In vivo sensitivity
198. Pfeffer, L. M., A. Dinarello. C, et al. (1998). “Biological proper- and resistance of chronic myelogenous leukemia cells to alpha
ties of recombinant alpha-interferons: 40th anniversary of the interferon: Correlation with receptor binding and induction of
discovery of interferons.” Cancer Res(15(58) ): 2489–2499. 2′,5′- oligoadenylate synthetase.” Cancer Res(46): 4848–4852.
199. Pitha, P. (1993). “Nomenclature of the human interferon genes. 219. Rossi, G. (1985). “Interferon and cell differentiation. In: Interferon.”
For the Nomenclature Committee of the International Society for Academic Press(6): 31–68.
Interferon Research.” J Int Res(13): 443–444. 220. Rosti, G., F. Bonifazi, et al. (1999). “Cytarabine increases karyo-
200. Pizzocaro, G., L. Piva, et al. (2001). “Interferon adjuvant to radical typic response in alpha interferon treated chronic myeloid leuke-
nephrectomy in Robson stages II and III renal cell carcinoma: a mia patients: results of a national prospective randomized trial of
multicentric randomized study.” J Clin Oncol(19(2) ): 425–431. the Italian Cooperative Study Group on CML.” Blood(94): 600A.
201. Poiesz, B. J., F. W. Ruscetti, et al. (1980). “Detection and isola- 221. Rozman, C., E. Montserrat, et al. (1988). “Recombinant alpha2
tion of type C retrovirus particles from fresh and cultured lym- interferon in the treatment of B chronic lymphocytic leukemia in
phocytes of a patient with cutaneous T-cell lymphoma.” Proc early stages.” Blood(71): 1295–1298.
Natl Acad Sci USA(77): 7415–7419. 222. Rupoli, S., S. Barulli, et al. (1999). “Low dose interferon-alpha2b
202. Pope, G. R., M. R. Hadam, et al. (1981). “Kinetics of natural combined with PUVA is an effective treatment of early stage mycosis
cytotoxicity in patients treated with human fibroblast interferon.” fungoides: results of a multicenter study. Cutaneous-T Cell Lymphoma
Cancer Immunol Immunother(11): 1–6. Multicenter Study Group.” Haematologica(84(9) ): 809–813.
203. Porzsolt, F. (1992). “Adjuvant therapy of renal cell cancer with 223. Salmon, S. E., J. J. Crowley, et al. (1994). “Combination chemo-
interferon alfa-2A.” Proc Amer Soc Clin Oncol(11): 202. therapy, glucocorticosteroids, and interferon alfa in the treatment
204. Pyrhonen, S., E. Salminen, et al. (1999). “Prospective randomized of multiple myeloma. A Southwest Oncology Group Study.” J
trial of interferon alfa-2a plus vinblastine versus vinblastine alone Clin Oncol(12): 2405–2414.
in patients,with advanced renal cell cancer.” J Clin Oncol(17(9) ): 224. Samuelsson, J., H. Hasselbalch, et al. (2006). “A phase II trial of
2859–2867. pegylated interferon alpha-2b therapy for polycythemia vera and
205. Quesada, J. R., E. M. Hersh, et al. (1984). “Alpha interfereon for essential thrombocythemia: feasibility, clinical and biologic effects,
induction of remission in hariy cell leukemia.” N Engl J and impact on quality of life.” Cancer Res(106(11) ): 2397–2405.
Med(310): 15–18. 225. Schaar, C. G., H. C. Kluin-Nelemans, et al. (2005). “Interferon
206. Quesada, J. R., R. Alexananian, et al. (1986). “Treatment ot mul- alpha as maintenance therapy in patients with multiple myleoma.”
tiple myeloma with trecombinanat alpha interferon.” Blood(67): Ann Oncol(3(16) ): 634–639.
275–278. 226. Scheinfeld, N. (2007). “A brief primer on treatments of cutaneous
207. Quesada, J. R., E. M. Hersh, et al. (1986). “Treatment of hairy T cell lymphoma, newly approved or late in development.” J
cell leukemia with recombinant alpha-interferon.” Blood(68): Drugs Dermatol(6(7) ): 757–760.
493–497. 227. Scheithauer, W., A. Cortelezzi, et al. (1989). “Combined alpha-
208. Quesada, J. R., R. Alexananian, et al. (1987). “Alpha interferons 2C-interferon/VMCP polychemotherapy versus VMCP as induc-
in hairy cell leukemia (HCL). A five year follow up in 100 tion therapy in multiple myeloma: A Prospective Randomized
patients.” J Interf Res (7): 678. Trial.” J Biol Resp Modif(8): 109–115.
209. Quesada, J. R., R. Alexananian, et al. (1988). “Recombinant 228. Schornagel, J. H., J. Verweij, et al. (1989). “Phase II Study of
interferon gamma in hairy cell leukemia, multiple myeloma, recombinant interferon alpha-A and vinblastine in advanced
Waldenstrom’s macroglobulinemia.” Am J Hematol (29): 1–4. renal cell carcinoma.” J Urol(142): 253–254.
210. Raefsky, E. L., L. C. Platanias, et al. (1985). “Studies of interferon 229. Schulof, R. S., M. J. Lloyd, et al. (1985). “Recombinant leukocyte A
as a regulator of hematopoietic cell proliferation.” J Immunol interferon in B-cell chronic lymphocytic leukemia: In vivo effects on
(135): 2507. autologous antitumor immunity.” J Biol Resp Modif(4): 310–323.
211. Trump D.L., Ravdin, P., M., E. C. Borden, et al. (1990). “Interferon 230. Senn, C. C. (1984). “Biochemical pathways in interferon action.”
alpha-N1 and continuous infusion vinblastine for treatment of Pharmacol Ther(24): 235–257.
advanced renal cell carcinoma.” J Biol Resp Mod. (9(1)): 108-111 231. Sertoli, M. R., I. Brunette, et al. (1989). “Recombinant alpha-20
212. Real, F. X., H. F. Oettgen, et al. (1986). “Kaposi’s sarcoma and interferon plus vinblastine in the treatment of metastatic renal
the acquired immunodeficiency syndrome. Treatment with high cell carcinoma.” Am J Clin Oncol(12): 43–45.
and low doses of recombinant leukocyte A interferon.” J Clin 232. Shepherd, F. A., R. Beaulieu, et al. (1998). “Prospective ran-
Oncol(4): 544–551. domised trial of two dose levels of interferon alfa with zidovu-
213. Revel, M., Kimch, A., et al. (1980). “Role of interferon-induced dine for the treatment of Kaposi’s sarcoma associated with human
enzymes in the antiviral and antimitogenic effects of interferon.” immunodeficiency virus infection: a Canadian HIV clinical trials
Ann NY Acad Sc(350): 459–473. network study. et a.” J Clin Oncol(16): 1736–1742.
300 Interferons: therapy for cancer

233. Shin, D. M., F. R. Khuri, et al. (2001). “Combined interferon-alpha, 253. Strander, H., U. Abramson, et al. (1978). “Adjuvant interferon
13-cis-retinoic acid, and alpha-tocopherol in locally advanced head treatment of human osteosarcoma.” Rec Results Ca Res(68): 40.
and neck squamous cell carcinoma: novel bioadjuvant phase 254. Strander, H., K. Cantell, et al. (1980). “Interferon treatment of
II trial.” J Clin Oncol(19): 3010–3017. osteogenic sarcoma: A clinical trial. In: Conference on Modulation
234. Sidky, Y. A. and E. C. Borden (1987). “Inhibition of angiogenesis of Host Immune Resistance in the Prevention and Treatment of
by interferons: Effects on tumor- and lymphocyte-induced vascu- Induced Neoplasms.” (28): 377–381.
lar responses.” Cancer Res 1987(47): 5155. 255. Talpaz, M., K. B. McCredie, et al. (1983). “Leukocyte interferon-
235. Sigaux, F., S. Castaigne, et al. (1987). “Alpha-interferon in hairy induced myeloid cytoreduction in chronic myelogenous leuke-
cell leukaemia: direct effects on hairy cells or indirect cytotoxicity.” mia.” Blood(62): 689–692.
Int J Cancer Suppl(1):2–8. 256. Talpaz, M., J. M. Trujillo, et al. (1985). “Suppression of clonal
236. Silver, R. T. (1993). “Interferon-alpha 2b:A new treatment for evolution in two chronic myelogenous leukemia patients treated
polycythemia vera.” Ann Intern Med: 1091–1092. with leukocyte interferon.” Br J Haematol(60): 619–624.
237. Silver, H. K. B., J. M. Connors, et al. (1985). “Prospectively random- 257. Talpaz, M., H. M. Kantarjian, et al. (1987). “Clinical investigation
ized toxicity study of high-dose versus low-dose treatment strategies of human alpha interferon in chronic myelogenous leukemia.”
for lymphoblastoid interferon.” Cancer Treat Rep(69): 743–750. Blood(69): 1280–1288.
238. Silver, R. T., M. Coleman, et al. (1987). “Interferon (rIFN) alfa 258. Talpaz, M., R. Kurzrock, et al. (1989). “Recombinant interferon-
2b has activity in treating chronic myeloid leukemia (CML) in alpha therapy of Philadelphia chromosome-negative myeloprolif-
rIFN gamma failures.” Proc Amer Soc Clin Onc(6): 149. erative disorders with thrombocytosis.” Am J Med(86): 554–558.
239. Singh, R. K., M. Gutman, et al. (1995). “Interferons alpha and 259. Taniguchi, T., S. Ohno, et al. (1980). “The nucleotide sequence of
beta downregulate the expression of basic fibroblast growth fac- human fibroblast interferon cDNA.” Gene(20): 11–15.
tor in human carcinoma.” Proc Natl Acad Sci USA(92): 4562. 260. Taylor-Papadimitriou, J., N. Ebsworth, et al. (1985). “Possible
240. Sirohi, B., R. Powles, et al. (2007). “An open, randomized, con- mechanisms of interferon-induced growth inhibition. In:
trolled, phase II, single centre, two-period cross-over study to Mediators in Cell Growth and Differentiation.” 283–298.
compare the quality of life and toxicity experienced on PEG 261. Tichelli, A., A. Gratwohl, et al. (1989). “Treatment of thrombocy-
interferon with interferon-alpha2b in patients with multiple tosis in myeloproliferative disorders with interferon alpha-2a.”
myeloma maintained on a steady dose of interferon-alpha2.” Ann Blut(58): 15–19.
Oncol 3(18(8) ): 1388–1394. 262. Toy, J. (1983). “The interferons.” Clin Exp Immunol(54): 1–13.
241. Smalley, R. V., J. Vogel, et al. (1977). “Chronic granulocytic 263. Trinchieri, G. and B. Perussia (1985). “Immune interferon: A
leukemia:Cytogenetic conversion of the bone marrow with cycle pleiotropic lymphokine with multiple effects.” Immunol Today
specific chemotherapy.” Blood(50): 107–113. 1(6): 131–136.
242. Smalley, R. V., S. S. Anderson, et al. (1991). “A randomized com- 264. Trump, D. I., P. Elson, et al. (1996). “Randomised controlled trial of
parison of a low dose and a standard dose of alpha interferon adjuvant therapy with lymphoblastoid interferon in resected high
(WellferonR) in Hairy Cell Leukemia.” Blood(78): 3133–3141. risk renal cell carcinoma.” Proc Am Soc Clin Oncol(15): 253.
243. Smalley, R. V., J. W. Andersen, et al. (1992). “Interferon Alfa 265. Tura, S. (1994). “Interferon alfa 2a as compared with conven-
Combined with Cytotoxic Chemotherapy for Patients with Non- tional chemotherapy for the treatment of chronic myeloid leuke-
Hodgkin’s Lymphoma – an ECOG Study.” New Eng J Med(327): mia.” New Engl J Med(330): 820–825.
1336–1341. 266. Uze, G., G. Lutfalla, et al. (1990). “Genetic transfer of a func-
244. Smalley, R. V., J. Connors, et al. (1992). “Splenectomy vs. Alpha tional human interferon-α receptor into mouse cells: Cloning and
Interferon: A Randomized Study in Patients with Previously expression of its cDNA.” Cell(60): 225–234.
Untreated Hairy Cell Leukemia.” Amer J Hematol(41): 13–18. 267. Varesio, L., E. Blasi, et al. (1985). “Recombinant IFN-gamma
245. Smedley, H. M. and T. Wheeler (1983). “Toxicity of IFN. In: activates macrophages to become tumoricidal.”
Interferon and cancer.” 203–210. 268. Verma, S., I. Quirt, et al. (2006). “Systematic review of systemic
246. Solal-Celigy, P., E. Lepage, et al. (1998). “Doxorubicin- adjuvant therapy for patients at high risk for recurrent melanoma.”
containing regimen with or without interferon alfa-2b for Cancer Res(106 (7) ): 1431–1442.
advanced follicular lymphomas: final analysis of survival and 269. Vilcek, J., H. C. Kelke, et al. (1985). “Structure and function of
toxicity in the Groupe d’Etude des Lymphomes Folliculaires.” J human interferon gamma. In: Mediators in Cell Growth and
Clin Oncol(16): 2332–2338. Differentiatinon.” 299–313.
247. Spinna, C. A., J. L. Dahey, et al. (1983). “Suppression of NK cell 270. Volberding, P., R. Valero, et al. (1984). “Alpha interferon therapy
cytotoxicity in the peripheral blood of patients receiving inter- of Kaposi’s sarcoma in AIDS.” Ann NY Acad Sci(437): 439–447.
feron therapy.” J Biol Resp Modif(2): 458–459. 271. von Wussow, P., M. Frund, et al. (1988). “Immunogenicity of dif-
248. Stadler, W. M., T. M. Kuzel, et al. (1997). “Metastatic bladder ferent types of interferons in the treatment of hairy cell leukemia.”
cancer: advances in treatment.” Eur J Cancer(33): 23. N Engl J Med(319): 1226.
249. Stadler, R., H. G. Otte, et al. (1998). “Prospective randomized 272. von Wussow, P., R. Hehlmann, et al. (1994). “Roferon (rIFN-
multicenter clinical trial on the use of interferon-2a plus acitretin α2a) is more immunogenic than Intron A (rIFN-α2b) in patients
versus interferon-2a plus PUVA in patients with cutaneous T-cell with chronic myelogenous leukemia.” J Interferon Res(14):
lymphoma stages I and II.” Blood(92(10) ): 3578–3581. 217–219.
250. Steis, R. G., J. W. Smith, et al. (1988). “Resistance to recombinant 273. Vugrin, D., L. Hood, et al. (1985). “A Phase II trial of high-dose
interferon alfa 2a in hairy cell leukemia associated with neutral- human lymphoblastoid alpha interferon in patients with advanced
izing anti-interferon antibodies.” N Engl J Med(318): 1409–1413. renal carcinoma.” Cancer Treat Rep(69): 817–820.
251. Steis, R. G., W. J. Urba, et al. (1988). “Immunogenicity of different 274. Wadler, S., E. L. Schwartz, et al. (1989). “5-fluorouracil and
types of interferons in the treatment of hairy-cell leukemia.” recombinant alpha-2a-interferon: An active regimen against
N Engl J Med(319): 1227. advanced colorectal carcinoma.” J Clin Oncol(7): 1769–1775.
252. Stewart, W. E. and D. K. Blanchard (1985). “Interferons, cytostatic 275. Wadler, S. and P. H. Wiernik (1990). “Clinical update on the role
and immunomodulatory effects.” Immunity to Cancer Eds Reif of fluorouracil and recombinant interferon alfa-2a in the treat-
AE, Mitchell MS (Academic Press, Orlando, Fl, USA) 295–307. ment of colorectal carcinoma.” Sem Onc(17(Suppl 1) ): 16–21.
D. Goldstein et al. 301

276. Wagstaff, J., P. Loynds, et al. (1985). “A phase II study of rDNA photochemotherapy: a prospective controlled trial.” J Am Acad
human alpha 2 interferon in multiple myeloma.” Cancer Treat Dermatol(44(2) ): 253–260.
Rep(69): 495–498. 284. Worman, C. P., D. Catovsky, et al. (1987). “The UK experience
277. Weiner, L. M., P. J. Moldofsky, et al. (1988). “Antibody delivery withhuman lymphoblastoid interferon in HCL: A report of the
and effector cell activation in a Phase II trial of recombinant gamma first 50 cases.” Leukemia(1): 320–322.
interferon and the murine monoclonal antibody CO-17–1-A in 285. Worman, C. P., A. B. W. Nethersell, et al. (1987). “Natural IFN-a
advanced colorectal carcinoma.” Cancer Res(49): 1609–1621. therapy in hairy-cell leukaemia (Namalva-Type IFN-Wellferon).”
278. Weiss, G. R., P. Y. Liu, et al. (1998). “13-cis-retinoic acid or Klin Wochenschr(65): 685–687.
all-trans-retinoic acid plus interferon-alpha in recurrent cervical 286. Xu, D., S. Erickson, et al. (2000). “Interferon alpha down-regu-
cancer: a Southwest Oncology Group phase II randomized trial lates telomerase reverse transcriptase and telomerase activity in
1998.” Gynecol Oncol(71): 386–390. human malignant and nonmalignant hematopoietic cells.”
279. Westin, J., S. Rödjer, et al. (1995). “Interferon alfa-2b versus no Blood(96): 4313–4318.
maintenance therapy during the plateau phase in multiple 287. Yang, J. L., X. J. Qu, et al. (2007). “Erlotinib (OSI-774)-induced
myeloma. A randomized study.” Br J Haematol(89): 561–568. inhibition of transitional cell carcinoma of bladder cell line growth is
280. Wiernik, P. H., B. Schwartz, et al. (1990). “Successful treatment enhanced by interferon-alpha.” BJU Int 2000(99(6) ): 1539–1545.
of hairy cell leukemia with beta ser interferon.” Am J Hematol. 288. Yoffe, G., M. Blick, et al. (1987). “Molecular analysis of inter-
281. Wirth, M. P. (1993). “Immunotherapy for metastatic renal cell feron-induced suppression of Philadelphia chromosome in
carcinoma Urol.” Clin North Am(20): 283–295. patients with chronic myeloid leukemia.” Blood(69): 961–963.
282. Wisloff, F. and N. Gulbrandsen (2000). “Health related quality of 289. Zinzani, P. L., Magagnoli. M., et al. (2000). “Long term-term
life and patient’s perceptions in interferon-treated multiple follow-up of hairy cell leukemia patients treated with 2-chlorode-
myeloma patients.” Nordic Myeloma Study Group Acta Onco(39): oxyadenosine.” Haematologica 2000 (85(9) ): 922–925.
809–813. 290. Zoon, K. C., R. Hu, et al. (1986). “Further Studies on the Purification
283. Wollina, U., A. Looks, et al. (2001). “Treatment of stage II cutane- and Partial Characterization of Human Lymphoblastoid Interferon
ous T-cell lymphoma with interferon alfa-2a and extracorporeal Alphas. In: The Biology of the Interferon System.” 55–58.
10 Monoclonal antibody therapy
ROBERT O. DILLMAN

Abstract Monoclonal antibodies were the first suc-


cessful targeted anti-cancer therapeutics in the modern
Background and Rationale
era. In the 1970s elucidation of hybridoma and molecular for Antibody Therapy
biology technologies established mechanisms to pro-
duce and modify monoclonal antibodies with therapeu- Historical aspects 1900–1980
tic anti-cancer potential. In the 1980s early investigation Monoclonal antibodies (Mab) were the first successful
with murine monoclonal antibodies in animal models anti-cancer targeted therapeutics in the modern era
and patients established most of the principles of anti- [168]. A chronological history of monoclonal antibody
body therapy including immunologic and regulatory development is shown in Table 1. At the end of the
mechanisms of anti-cancer effects, and the use of immu- nineteenth century antisera and antibodies were discov-
noconjugates in which antibodies act as carriers. These ered as part of the independent work of Emil Behring
early studies also showed that most of the toxicities and Kitasato Shibegan that led to developing diptheria
associated with monoclonal antibodies relate to the antitoxin. By immunizing an animal with foreign anti-
target antigen, and less often immune interactions with gens contained on cells, early immunologists recog-
the antibody itself. In 1997 the United States Food and nized that they could produce antisera against antigens
Drug Administration approved rituximab (Rituxan) for on the surface of cells. The potential to utilize antibod-
the treatment of B-cell lymphoma, making it the first ies as therapeutic agents was first espoused in the early
monoclonal antibody to receive regulatory approval for twentieth century. The German immunologist, bacteri-
the treatment of human malignancy. As of early 2008, ologist, and 1908 Nobel Prize winner Paul Ehrlich used
there were six commercially available unconjugated the term “passive immunization” to describe the use of
monoclonal antibodies with marketing indications for antisera and antibodies in the treatment of disease as
treatment of human malignancy. In addition to ritux- opposed to the “active immunization” using cell or
imab for B cell malignancies, this list includes alemtu- antigen-based vaccines. Ehrlich, whose 150th birthday
zumab (Campath) for B and T cell malignancies, was 14 March 2004, is generally considered the father
trastuzumab (Herceptin) which inhibits Her-2 overex- of antibody therapy and is credited for the term “magic
pressing breast cancers, anti-epidermal growth factor bullets” to describe the potential for antibodies to spe-
receptor (EGFR) antibodies cetuximab (Erbitux) and cifically target bacteria or cancer cells as opposed to
panitumumab (Vectibix) which inhibit various epithelial normal cells [201, 644].
solid tumors including colorectal cancer, and bevaci- Before 1975, it was virtually impossible to isolate a
zumab (Avastin) which binds to vascular endothelial specific antibody. Initial therapeutic endeavors were
growth factor (VEGF) and is probably useful to treat with “antisera”, a collection of heterogeneous anti-
most malignancies, including colorectal, lung, and bodies from multiple clones, typically derived from
breast cancers. These products have been most useful animal serum. Animals were repeatedly immunized
when combined with chemotherapy because of additive with preparations of human cells which induced an
or synergistic anti-tumor effects. immune response. Animal blood was collected several
Keywords Alemtuzumab • bevacizumab • cetuximab weeks later at a time when a primary immune response
• panitumumab • rituximab • trastuzumab was estimated to occur, and the serum fraction isolated.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 303
© Springer Science + Business Media B.V. 2009
304 Monoclonal antibody therapy

Table 1. Milestones in the development of monoclonal antibodies for the treatment of cancer
1908 Paul Ehrlich, German immunologist states concept of “magic bullets” for seeking out cancer cells, awarded Nobel Prize
1927 Publication describing serotherapy of 10 patients with chronic myelogenous leukemia
1975 Kohler and Milstein report “hybridoma” methodology to produce monoclonal antibodies; awarded Nobel Prize in 1986
1980 First publication of treatment of a cancer patient (lymphoma) with a monoclonal antibody
1982 First published report of a complete clinical remission in a patient (lymphoma) treated with a monoclonal antibody,
murine anti-idiotype
1984 First U.S. FDA approval of a therapeutic monoclonal antibody, murine anti-CD3 muromonab-CD3 (Orthoclone OKT3) to
prevent renal allograft rejection
1992 First U.S. FDA approval of a radiolabeled anti-cancer monoclonal antibody, murine anti-B72.3 indium-labeled monoclonal
antibody, 111In-satumomab pendetide (Oncoscint), for radioimmunodetection of colon and ovarian cancer
1995 Approval in Germany of murine monoclonal antibody edrecolomab (Panorex) for the adjuvant treatment of colon cancer
in Germany
1997 U.S. FDA approval of chimeric anti-CD20 monoclonal antibody rituximab (Rituxan) for the treatment of B-cell lymphoma,
first indication to treat human malignancy
1998 FDA approval of humanized anti-HER2 monoclonal antibody trastuzumab (Herceptin) for the treatment of breast cancer,
first indication to treat a solid tumor
2000 U.S. FDA approval of humanized anti-CD33 immunotoxin, gemtuzumab ozgomicin (Mylotarg), for the treatment of acute
myelogenous leukemia, first approval of antibody-targeted immunotoxin
2002 U.S. FDA approval of murine anti-CD20 radiolabeled monoclonal antibody Y-90 ibritumomab tiuxetan (Zevalin) for B-cell
lymphoma, first approval of a radiolabeled antibody for treatment of malignancy
2003 U.S. FDA approval of murine anti-CD20 radiolabeled monoclonal antibody I-131-tositumomab (Bexxar) for B-cell
lymphoma
2004 U.S. FDA approval of bevacizumab (Avastin) for colorectal cancer
2004 U.S. FDA approval of cetuximab (Erbitux) for colorectal cancer
2006 U.S. FDA approval of panitumumab (Vectibix) for colorectal cancer

Such sera were then modified by repeated absorption In 1975 Hans Kohler and Caesar Milstein published a
with normal human tissues that were presumed to not paper in which they described the hybridoma technology,
contain tumor associated antigens. This process which allowed isolation of cloned cells that could
increased the specity of the final product which consti- secrete a single type of antibody [400]. For this work
tuted an antiserum. The degree of reactivity of this they were awarded Nobel Prizes in 1986. Following
immunoglobulin collection was tested in bioassays publication of this paper numerous academic laborato-
using serial dilution titrations, and the product was ries and early biotechnology companies began making
characterized on the basis of the dilutional titers, such Mab by immunizing rodents with human cells and anti-
as 1:64 or 1:256. Typically, large animals such as gens. The potential advantages of Mab over antiserum
horse, sheep, or goats were immunized in order to as a therapeutic product were readily apparent as sum-
yield large volumes of anti-serum. Because each four- marized in Table 2.
legged biofactory was biologically unique, it was
impossible to reproduce a given antiserum exactly,
even in the same animal. There were always issues of
purity and the impossibility of exactly reproducing any
given serum that inevitably consisted of multiple anti- Table 2. Comparative advantages of monoclonal Antibodies
vs. Antisera
bodies with various specificities and affinities. In as
much as antibodies were genetically determined for an Monoclonal Antibody Antisera
individual animal, the supply of any given antiserum Purified antigen to optimize Cell or crude antigen
was limited, and therefore there was no ability to per- satisfactory
Absorptions for specificity Screening for specificity
form large clinical trials with any specific antiserum.
Heterogeneous Homogeneous
Some antisera, such as anti-thymocyte globulin, are Variable lots Reproducible lots
still in clinical use today, but the biotechnology indus- Variable affinities Uniform affinity
try is trying to replace as many as possible with single Variable specificities Unique specificity
or combinations of Mabs. Limited quantities Unlimited production
Robert O. Dillman 305

Antisera Trials 1925–1980 with some antibodies, antitumor effects are observed in
immunodeficient mice even in the absence of an interac-
Between 1925 and 1980 numerous clinical studies were tion with complement and/or effector cells, suggesting
performed using antisera. These pioneering studies with that antitumor effects can be mediated by regulatory
heterologous and isologous antisera have been reviewed effects. Third, immune-mediated antitumor effects vary
in more detail elsewhere [137, 607, 795]. Such trials to some extent with the Ig subclass and isotype of the
were limited by the quantity of purified anti-tumor anti- Mab. Fourth, just as with chemotherapy, antitumor
sera available, the purity of such preparations, and dif- effects are more difficult to achieve in the presence of
ficulties producing similar lots of new antibody. large tumor burdens. Fifth, combining antibodies to dif-
Nevertheless, some antitumor effects were noted. One ferent antigens is a promising strategy to overcome
of the earliest reports by Lindstrom in 1927 described problems of heterogeneity [14, 16, 534]. Sixth, animal
ten chronic myelogenous leukemia (CML) patients models are of little use in predicting toxicities in humans
whom he treated with 15 courses of rabbit antisera. A for Mabs that react with antigens that are more prevalent
decline in peripheral blood myeloid cells was noted in in humans or only expressed in humans.
five patients, but there were also significant side effects
that were attributed to impurities in the preparation,
although these may well have been secondary to anti-
Human Trials with Murine Monoclonal
body binding to circulating CML cells. In the 1970s Antibodies 1980–1989
anti-thymocyte globulin (ATG) was used by several Clinical trials in man with murine Mab began in 1980 in
investigators to treat patients with T-cell malignancies hematopoietic malignancies and subsequently in solid
[198, 224, 815]. All eight patients described in these tumors. Most of the clinical investigation during the
reports had some improvement in their Sezary syndrome 1980s involved pilot and phase I trials with mouse
and/or lymphadenopathy. Therapy was complicated by Mab, and relatively few patients were actually studied.
hypotension and chills. Prophylaxis with diphenhy- Perhaps the first written report of monoclonal antibody
dramine and steroids seemed to prevent these side therapy was that by Nadler et al. who treated a man with
effects during subsequent treatments, but this may have B cell lymphoma with a murine Mab called AB89 [518].
been because circulating T lymphocytes had already On successive days the patient received slow infusions
been eliminated from the circulation. of AB89 at doses of 25, 75, and 150 mg; 1 month later a
Interpretation of antisera studies was complicated by 1.5 gm dose was administered. There was a transient
issues of antibody source, purity and specificity. Certain decrease in the lymphocyte count after each treatment,
toxicities were identified, but it was unclear whether but no significant antitumor effect was noted. During
these were due to impurities, hypersensitivity reactions 1980 and 1989 a number of clinical studies were con-
to foreign proteins, or to antitumor effects. Although ducted with Mab directed against antigens found on
side effects were typically attributed to allergic reaction cells from hematologic malignancies [19, 38, 161, 177,
to foreign proteins, it is likely that most of the toxicity 179, 180, 227, 228, 446, 478, 485–488, 518, 569, 600,
observed was actually due to specific interaction with 666] and solid tumor cancers [109, 161, 205, 240, 262,
leukocytes and the release of cytokines [170]. 263, 328, 375, 443, 513, 535, 625, 651–653, 666, 738,
776]. In 1982, the first report of achieving complete
regression of tumor in a patient with B cell lymphoma
Animal Models and Antibody Therapy was published [487]. Most of the principles of antibody
The chapters on antibody therapy in the first three edi- therapy, including immunologic and regulatory mecha-
tions of Principles of Cancer Biotherapy [162], [165], nisms of anti-cancer effects, and the use of immunocon-
[169] summarized some of the important animal work jugates in which antibodies act as carriers were
with antisera and Mab that laid the foundation for the established during these early trials. These early trials
use of such products in man. The demonstration of also established that most toxicities and adverse events
safety, tumor targeting, and anti-tumor effects demon- associated with monoclonal antibody infusions are the
strated in various animal models justified and guided results of interactions with the target antigens, and less
human clinical trials that eventually established Mab as often due to immune interactions with the antibody
an effective anti-cancer treatment modality [816]. The itself. The potential of Mab for the treatment of human
following conclusions can be drawn from the heterolo- malignancy was clearly recognized [163, 533].
gous antisera and studies in animal models. First, anti- In the early 1990s two radiolabeled Mab received regu-
tumor effects can be mediated by Mab in vivo. Second, latory approval for imaging and radioimmunodetection
306 Monoclonal antibody therapy

of tumors. In 1995, Germany granted the first approval of in the sequence of peptides that constitute the hypervariable
a mouse monoclonal antibody for cancer therapy region (Fhv) [418, 643]. The Fhv region constitutes the
(Panorex) for colorectal cancer, although this product “idiotype” of the Ig, and the specific peptide sequences
was never approved in the United States. In the early of the idiotype are termed “idiotopes.” The six hyper-
1990s trials began with murine Mab that had been modi- variable subregions of the variable region are also
fied using molecular biology technologies to produce referred to as the complementarity-determining regions
antibodies that consisted mostly of human amino acid (CDR). “Lock-and-key” is a popular metaphor for the
sequences. In 1997 the United States Food and Drug unique chemical relationship and structural binding
Administration (FDA) approved the first monoclonal between a specific antibody and its antigen target. The
antibody (rituximab) for the treatment of malignancy in affinity of antibody for its antigen is a measure of how
the United States [168]. During the next decade several tightly the antibody binds to the antigen.
unconjugated and conjugated antibody products received The diversity of humeral immunity is made possible by
regulatory approval for commercial use. the various combinations and permutations of arrange-
ments and combinations of Fv regions, heavy chains, and
light chains [419, 643]. More specifically there are vari-
Antibodies and Antigens able (V), diversity (D), and joining (J) genes that code for
When foreign cells are injected into an animal, they sequences on the heavy chain (V,D, and J), and light chain
induce an immune response that results in the produc- (V and J) that provide nearly infinite combinatorial poten-
tion of immunoglobulin (Ig) proteins, called antibodies, tial for rearrangement. Based on their chemical composi-
each of which binds specifically to certain cell surface tion, the light chains of Ig are classified as kappa or
molecules, termed antigens. Collectively these antibod- lambda, and heavy chains as immunoglobulins IgG, IgM,
ies are antisera. The antigens are characterized as IgA, IgD, or IgE. Other differences in heavy chains allow
glycoproteins, glycolipids, polysaccharides, etc. A tumor- subclass characterization such as IgG1 IgG2a, IgG2b,
specific antigen is one that is found only on cancer cells. IgG3, IgG4, etc. Human chromosomes 2, 14, and 22 con-
A tumor-associated antigen is an antigen expressed on tain various loci for light and heavy chains.
tumor cells, but also known to be present on normal The basis for antibody interaction with other compo-
cells. Antibodies consist of sequences of amino acids nents of the immune system, such as complement pro-
that are linked by disulfide bonds that yield two heavy teins and effector cells, resides in carbohydrate residues
chains and two light chains. Each light chain is con- that are present on the Fc portion of the Ig molecule and
nected to a heavy chain by a disulfide bridge, and the Fc receptors (FcR) on effector cells [250, 524, 746]. The
two heavy chains also are connected via disulfide interaction between Fc and FcR is influenced by several
bridges. As shown in Fig. 1, enzymatic cleavage of these factors including the affinity between the antibody iso-
various sites by papain or pepsin yields antibody frag- type and the specific receptor, the composition of the
ments termed Fab, F(ab’)2, and Fc. The basis for speci- sugar side chain on the Fc-portion of the antibody, and
ficity of antibody binding resides in the variable regions inhibitory characteristics of the FcR. Subclasses of
of the heavy and light chains (Fv), and more specifically Fc and receptors have been defined that determine
whether a specific antibody will bind complement
or not. Antibody-dependent cell cytotoxicity (ADCC)
depends in part on the specific N-glycosylation of the Fc
domain of the monoclonal antibody. Cross-linking of
FcR triggers various immune reactions including phago-
cytosis after opsinization, ADCC, and release of various
cytokines that act as inflammatory mediators [117, 701].
FcR that bind human IgG Fc are referred to as FcγR and
these are subclassed based on their binding characteris-
tics and function. From a functional perspective, there
are two main types of FcγR found on the surfaces of
immune effector cells, activating FcγR, and inhibiting
FcγR. Based on genetic polymorphisms the activating
Figure 1. Structure of immunoglobulins and fragments. FcγR include the high-affinity receptor CD64 (FcγRI),
Two heavy chains are connected by a disulfide bond, and each and the low affinity receptors CD32a (FcγRIIa) and
in turn is connected to a light chain by a disulfide bond CD16 (FcγRIIIa). CD64, which is expressed on monocytes,
Robert O. Dillman 307

macrophages, and dendritic cells, is the only FcγR with scale production of Mab [400]. Various laboratories
a high affinity to Mab monomeric IgG. Cells with a utilized this technology to identify large numbers of
CD64 Fc receptor can bind circulating Mab that is not mouse Mab that reacted with tumor-associated antigens
bound to antigen, while the lower affinity Fc bind better [158]. As shown in Fig. 3, in this methodology mice or
to Mab that is bound to antigen. CD32a is expressed on rats are injected with whole tumor cells or purified
these cells, and also on neutrophils, eosinophils, and tumor associated antigens (such as carcinoembryonic
platelets. CD16a is highly expressed on NK cells and
macrophages. There is some evidence that these poly-
morphisms are predictive of the clinical efficacy of cer-
EFFECTOR
tain antibodies. This is leading to specific strategies to C’
CELL
modify the Fc portion of antibodies in order to optimize COMPLEMENT
the anti-tumor effector functions [667, 695].
ACTIVATED
Theoretically antibody therapy is the most tumor spe-
T-CELL
cific approach to systemic cancer treatment that has been
γ - IFN
discovered. As noted earlier, antibodies directed against a
tumor antigen are tumor specific to the degree to which that
antigen is found only on tumor cells. At one time it was TUMOR
IL-2 CELL RICIN A
believed that all cancers were foreign and that tumor-spe- (Toxin)
cific antigens would be readily discovered. Unfortunately,
TNF
phenotypically and genetically, cancer cells are more like α-, β-IFN
normal cells from that host, and as a result, most tumor DNR
(Drug)
antigens are only relatively tumor specific because they are
found in certain normal tissues as well. However, many Y90
tumor-associated antigens are expressed much more heav- (Isotope)
ily only during embryonic development; and, therefore are
relatively tumor specific. Also, certain viruses produce or Figure 2. Schematic of potential applications of monoclonal
antibodies for cancer therapy. Various antigens, represent-
induce antigens that are relatively tumor specific. The idio- ing the heterogeneity of cancer, are depicted on the tumor
type encoded by B cells of a specific B-cell malignancy is cell surface. Bifunctional antibodies are depicted with one
an example of a tumor specific antigen. Fab directed to a tumor antigen and the other to a biologic
response modifier. One antibody has a Fab directed to a CD2
or CD3 receptor on T cells, which results in their activation
Unconjugated Antibodies as Therapeutic and enhanced cytotoxicity. The Fc portion of the antibodies
Agents are attached to various cytotoxic substances including com-
plement, effector cells (granulocytes, monocytes, killer T
As illustrated in Fig. 2 and Table 3, there are many ways cells) of the immune system, and cytotoxic agents such as
in which antibodies may be used in cancer therapy [161, toxins, radioisotopes, and chemotherapy agents. IFN, inter-
feron; TNF, tumor necrosis factor; IL-2, interleukin-2; DNR,
163, 533]. This chapter covers the rationale for the use daunorubicin. Ann Intern Med 1989, 111:592–603. By per-
of unconjugated (or “naked”) antibodies in cancer ther- mission of the American College of Physicians
apy and the current role of such Mab in the treatment of
human malignancy. In subsequent chapters of this book,
the use of conjugated Mab as carriers of cytotoxic sub-
stances including chemotherapy agents, radioisotopes, Table 3. Strategies for in vivo use of monoclonal antibodies
as anti-cancer therapy
and natural toxins are reviewed.
Antibody alone
Complement mediated cytotoxicity (CMC)
Antibody dependent cell-mediated cytotoxicity (ADCC)
Manufacturing of Monoclonal Regulatory interactions (ligands, receptors)
Anti-idiotype vaccines
Antibodies Immunoconjugates
Radiolabeled antibodies
Hybridoma Technology Immunotoxins
Chemotherapy-antibody conjugates
In 1975 Kohler and Milstein, published a landmark Cytokine immunoconjugates
paper in which they described the “hybridoma technol- Cellular immunoconjugates
ogy,” which opened the doors to reproducible and large- Multi-targeted immunoconjugates
308 Monoclonal antibody therapy

HPRT+ Nonsecretory
HPRT −
Immunization spleen cells
B-cell
culture

Fusion in PEG

Selection in HAT medium

Screening of supernatants for antibody


and selection of clones

Recloning and subcloning

Supernatant
(contains 10 to 100
mg /mL MOAb)

Purified ascites
(contains 10 to 20
mg/mL MoAb)

Figure 3. Schema for murine monoclonal antibody production. From [176]. By permission of Biomedical Information Corporation

antigen) to induce a polyclonal immune response against single clone, (2) inability to produce and secrete immu-
the foreign human proteins. The animal’s immune sys- noglobulin themselves, and (3) inability to grow in the
tem responds with production of antibodies directed presence or absence of specific medium constituents. In
against the tumor antigens. The blueprints for the anti- the presence of a cell-fusing substance, such as polyeth-
bodies are contained in by B lymphocytes that are found ylene glycol, individual spleen cells with the genetic
in large quantities in the spleen. The spleen of an immu- machinery for making antibody against the tumor fuse
nized animal is removed and minced into a single-cell with cells from the immortal cell line, thus producing
suspension. If placed in tissue cultures, such cells typi- hybrid cells, or hybridomas that retain the ability to
cally die off in a few days and are unable to proliferate. secrete antibodies. By the plating process of limiting
For this reason, the B lymphocytes are combined with dilution, a single hybridoma clone of cells can be iso-
other mouse B cells, typically mouse myeloma cells, lated that retains the ability to secrete the antibody
that have been specifically selected for certain charac- encoded in the DNA of the mouse spleen cells, hence
teristics, including (1) ability to grow in culture as a the term “monoclonal antibody.” The antibodies are isolated
Robert O. Dillman 309

from culture media and screened for specificity against


various cells and tissues. A major limitation of this
approach is that antigens are defined by the mouse
immune system, and tumor antigens are defined by differ-
ential expression on normal as opposed to cancer cells.
The first large-scale production factories for Mab for
clinical trials were the peritoneal cavities of Balb-C and
athymic mice. Injection of the hybridomas into the peri-
toneal cavity resulted in a malignant ascites that was Figure 4. Comparison of different types of monoclonal anti-
bodies mouse antibodies illustrating the component of
rich in the monoclonal antibody. This technology helped murine amino acid sequences in each. Shown are mouse
launch the biotechnology industry, and made possible antibodies (omabs), mouse/human chimeric antibodies
mass production of promising antibodies [14, 94, 157]. (-ximabs) in which the murine variable region is combined
The techniques of mass production included the in vivo with a human constant region, “humanized” antibodies
(-zumabs) in which only the hypervariable peptide sequences
expansion of hybridomas as ascites in mice, and in vitro
are retained from the original mouse antibody, and human
expansion of such hybrid cells in various large tissue antibodies (umabs)
culture vessels and bioreactors.

optimize ADCC. Other genetic engineering modifications


Recombinant Technology have focused on making smaller antibody molecules as
single chain Fv, Fab, and F(ab’)2 [325, 584].
and Engineering Monoclonal Antibodies
Murine hybridomas that produced desirable antibodies
Human Monoclonal Antibodies
were used as a source for genetic material for mass pro-
duction in unicellular factories, and to make chimeric or In terms of therapeutic potential, there are several rea-
humanized Mab. Recombinant DNA technology was sons why human Mab offer advantages over those of
used to introduce antibody-encoding genes into other other species [21, 258, 416]. First, they are less immuno-
cells to facilitate antibody production by cells such as genic and less allergenic than non-human antibodies.
Escherichia coli, yeast, and Chinese Hamster Ovary Second, certain subclasses of human antibodies are more
(CHO) cells [42, 44, 58, 508]. Certain mouse plasmacy- effective than murine antibodies in facilitating comple-
toma lines also glycosylate proteins in a similar manner. ment and/or antibody-mediated cytotoxicity in vitro.
These unicellular organisms are currently the factories Third, by dose-to-dose comparisons, human antibodies
of choice for Mab production because they produce have longer half-lives and prolonged serum levels
human-like N-glycosylation of the proteins, which is because there is less non-specific uptake, destruction and
crucial for proper folding of the Ig protein for the stereo- metabolism. There is evidence that antibodies are taken
chemistry that is needed for optimal binding to antigen. up by FcR receptors, called neonatal or Brambell recep-
Recombinant DNA technology has displaced hybri- tors (FcRn), which bind the Fc portion of antibodies and
doma technology as the method of choice for monoclo- remove them from the circulation [357]; [442]. Human
nal antibody production. IgG1, IgG2, and IgG4 have high affinities for FcRn
Recombinant techniques have made it possible to which transport endocytosed antibodies from the lyso-
engineer a variety of Mab constructs [6, 157, 631]. some back to the serum, while murine antibodies and
Furthermore, recombinant DNA biotechnology has made human IgG3 have a low affinity for FcRn. For antibodies
it possible to modify mouse Mab into partially human- with a low affinity for FcRn, instead of being transported
ized forms. As discussed later in this chapter, this has led back to the cell surface, they are catabolized [527, 808].
to the production of mouse–human chimeric Mab and Several approaches have been used for isolating human
humanized Mab as diagrammed in Fig. 4, as well as anti- Mab [189, 258, 671]. In one approach, it is presumed that
bodies designed to react with more than one antigen as patients with cancer have developed at least a limited
shown in Fig. 2. As of 2007, of the 18 different therapeu- immune response to their tumor. Regional draining lymph
tic antibody products approved for human use in various nodes, which are the first line of regional immunologic
diseases, ten are manufactured in CHO cell lines and defense against tumor, are excised to obtain a source of
eight are made in murine plasmacytoma cells. Additional B lymphocytes, some of which make antibodies against
modifications include engineering the Fc portion to opti- the tumor. Regional lymph nodes that drain known cancer
mize interactions with complement, or binding to FcR to sites are potential sources of B lymphocytes that may
310 Monoclonal antibody therapy

have been programmed to make antibodies against the the entire repertoire of antibodies which can then be
primary cancer. Immunization with tumor cells or anti- screened for their specificity, and the Fv isolated to pro-
gen, followed by excision of draining lymph nodes, espe- duce fully human recombinatorial products [6, 326,
cially sentinel lymph nodes, has also been used to isolate 631]. Using such libraries, fully human Mabs can be
antibody producing B lymphocytes [282, 299]. Another produced by linking Fv from the display library with
source of immune reactive B cells is the peripheral blood, human Ig hinge and constant regions. Human Mab can
and the frequency of such cells may be enhanced by also be produced by immunizing transgeneic mice whose
immunization. For example, researchers have injected immune system has been replaced with a human immune
irradiated autologous tumor cells into the skin with an system engineered to produce human Mab of a specific
immune stimulant such as Bacillus Calmette-Guerin class and subclass. Immunization of these animals
(BCG) or granulocyte macrophage colony stimulating results in human B cells that produce human Mab.
factor (GM-CSF). Rather than harvesting lymph nodes, 2
to 3 weeks after immunization peripheral blood B lym-
phocytes are collected, some of which secrete human Chimeric and Humanized Monoclonal
antitumor antibodies
Once antibody-producing B cells have been isolated,
Antibodies
they can be fused with other human cells of B cell lin- Because of the difficulties in producing and manufac-
eage that have been selected for similar characteristics turing human Mab, the extensive characterization of
as those immortal mouse B-cell lines described above, murine Mab that react against human tumor associ-
to produce antibody secreting clones [51, 123, 129, 256, ated antigens, and the clinical disadvantages of
257, 670]. It is also possible to fuse human lymph node murine Mab, other strategies have been developed to
B cells with cells from immortal mouse B-cell lines, and create mouse/human chimeric Mab and genetically
still get secretion of human antibody, although such engineered “humanized” Mab (Fig. 4) [1, 44, 58, 124,
cross-species hybridomas are usually unstable [67, 635, 325, 386, 440, 508, 510, 788]. One way to improve
802]. Other investigators have isolated B lymphocytes the activity of a Mab is to class-switch the Mab so
from lymph nodes and then infected these cells with that it has an Fc portion that is more suited for inter-
Epstein-Barr virus to immortalize cells producing action with human complement and/or effector cells
human Ig [96, 113, 189]. Using various techniques, [44, 157, 508]. Human IgG1 is usually considered
human Mab have been produced that react with breast most desirable because it effects both CMC and
cancer [67, 395, 635], lung cancer [669], colorectal car- ADCC. IgG3 also facilitate favorable CMC and
cinoma [299], brain tumors [670], stomach cancer [802] ADCC effects, but these proteins tend to aggregate
and melanoma [241, 364, 686]. However, human Mab which is problematic from a production standpoint. It
were more difficult to develop than murine Mab because has generally been taught that IgG4 is the least use-
of the instability of human hybridomas and immuno- ful, especially in terms of CMC, because of a lack of
globulin secreting cells, low secretion rates, and diffi- Fc receptors for this IgG subclass. In the chimeric
culties in mass producing and screening. approach, the variable domains of the light and heavy
These inherent problems in human Mab production chains of a desirable murine Mab, are linked to hinge
have been overcome by recent advances in transfection and constant domains of a human Ig. This is most
technology and molecular immunology that have made often done with recombinant techniques in which the
it feasible to integrate DNA for human Ig into cells that mouse genes for the variable domains of amino acids
can easily produce large quantities of human Mab. are combined with the human domains for the hinge
Newer technologies such as polymerase chain reaction and constant region amino acids. The genes are then
(PCR) [202] and transgenic mice [271] have led to iden- inserted into bacteria or yeast for large-scale produc-
tification of vast libraries of human antibodies that may tion. The bacterial preparations are typically not gly-
be superior in terms of antigen and antibody selection. cosylated while those grown in yeast are glycosylated,
In recent years various laboratories have been able to which is important for interaction via the Fc portion
clone cDNA for VH and VL contained in hybridomas of the antibody. It is also possible to isolate the genes
and B lymphocytes that were producing low levels of specific for the six hypervariable amino acid sequences
desirable antibodies. These were then linked to human on the light and heavy chains (CDR) of a desirable
hinge and constant genes to produce totally human Mab murine antibody, and these can then be inserted into
in more efficient cellular factories. Today one can take the framework of human Ig to create a “humanized”
Ig producing genes from B-lymphocytes and produce Mab via what is called “CDR grafting”. In this
Robert O. Dillman 311

construct the only residual mouse amino acid Table 4. Components that precede “mab” in the nomenclature
sequences remaining are those that give the antibody for naming a monoclonal antibody
its three-dimensional binding specificity. All of the Infix Target Inflx Origin
unconjugated monoclonal antibody products approved Tu (m) Tumor -a- Rat
for cancer treatment are either chimeric (cetuximab, Ci (r) Cardiovascular -o- Mouse
rituximab), humanized (alemtuzumab, trastuzumab, Li (m) Immune -xi- Chimeric
bevacizumab) or human (panitumumab). Because of human/mouse
Co (l) Colon tumor -zu- Humanized
the human Fc portion of the Ig molecule, products,
Me (l) Melanoma -u- Human
they possess superior cytotoxic potential based on Go (v) Ovarian tumor -axo- Rat/murine
interactions with complement and/or effector cells, hybrid
and are much less immunogenic, which is associated Pr (o) Prostate
with a longer bioavailability [662]. The availability
of “chimeric,” “humanized,” and fully human Mabs
has greatly reduced the limitations resulting from the
production of endogenous human anti-Ig antibodies regulatory approval. The suffice “mab” indicates that
such as human anti-mouse antibody (HAMA), human the product is a monoclonal antibody. The syllable
anti-chimera antibody (HACA), and human anti- directly in front of mab indicates the source such as
human antibody (HAHA) in man, and facilitated the mouse, chimeric, humanized, or human. The prefix of
use of repeated treatments with specific Mab for the name has no specific meaning. A second syllable
extended periods of time. may be added if there is something else linked or
attached to the product. The key components that appear
before “mab” for names adopted for the United States
Antibodies with Multiple Specificities are shown in Table 4. Most of the Mab are either mouse
and/or Functions (-omab), chimeric (-ximab), humanized (-zumab), or
human (-umab). Thus, rituximab is a chimeric Mab that
It is also possible to make Mab with specificity to more
targets tumor (as is cetuximab). Trastuzumab is a
than one antigen-binding site, and more than one func-
humanized mab that targets tumor (as is alemtuzumab).
tional capability. Some examples of bispecific and
Bevacizumab is a humanized mab that has a cardiovas-
bifunctional Mab, in which the antibody construct
cular target (vascular endothelial growth factor).
reacts with more than one antigen, are illustrated in
Panitumumab is a human mab that targets tumor.
Fig. 2 [124, 223, 711, 772, 793]. This can be done by
Limilimumab and tremelimumab are human mabs that
chemically linking either two different Fab pairs or
that have an immune target (CTLA4) while daclizumab
F(ab’)2 pairs, or two intact antibodies. It is also possi-
is a humanized mab with an immune target. Tositumomab
ble to make quadromas (a hybridoma of two monoclo-
and ibritumomab are murine mab that target tumor
nal antibody-secreting hybridomas), which secrete
while oregovomab is a mouse mab that targets ovary.
Mab that react with two different antigenic binding
Catumaxomab is a rat/mouse hybrid anti-tumor mab.
characteristics [123]. One can then select the appropri-
ate quadroma that is producing antibodies with two
different antigenic binding characteristics. In recent
years these approaches have been replaced by recom-
binant DNA techniques in which the genes for differ-
Mechanisms of Antibody-
ent murine variable or hypervariable regions of interest Mediated Anti-Tumor Effects
are linked to a humanized constant region and then As outlined in Table 1, there are at least three different
manufactured in cells [223]. rationales for using antibodies alone in vivo as cancer
treatment. The first rationale involves two different
immune mediated effects involving the Fc portion of the
Monoclonal Antibody Nomenclature immunoglobulin and interaction with other components
Because of the tremendous number of Mabs that are of the host immune system. The second rationale encom-
under development, a nomenclature system has been passes concepts of regulating tumor cells or their
developed that provides information about the construct microenvironment. The third concept relates to the anti-
and the target. These names are reserved until the prod- idiotype cascade as a means of immunization to induce
uct is being tested in trials that are considered pivotal for endogenous antibodies.
312 Monoclonal antibody therapy

Complement Mediated Cytotoxicity macrophages, granulocytes and eosinophils have such


receptors and can precipitate tumor cell lysis when
Complement mediated cytotoxicity (CMC) or comple- linked via Ig to a cell. Collectively, such cells are referred
ment dependent cytotoxicity (CDC) involves the fixation to as “effector cells.” The Fc portion of Ig is crucial for
of complement via the Fc portion of Ig and activation of linking antibody to effector cells via their FcR as shown
the complement protein cascade resulting in membrane in Fig. 6; the specific carbohydrates on the Fc are crucial
damage and cell destruction as shown in Fig. 5 [238, for this interaction [117, 524]. The numbers of Fc recep-
768]. Murine antibodies are usually ineffective in fixing tors and affinity for Fc predict the degree of ADCC
human complement in vitro. Exceptions to this are IgM activity [447, 796]. Antibody may first bind to its cellu-
and certain IgG3 murine antibodies [109, 794]. In man, lar target followed by effector cell binding via the Fc, or
studies of hemolytic anemia have confirmed that human alternatively, effector cells may first bind to the Fc of the
IgM is the most efficient human Ig class for complement antibody, and then be carried to the tumor target. Ex vivo
fixation followed by IgG1, IgG3, and IgG2. Because incubation of Mab and effector cells followed by in vivo
complement fixation occurs at the Fc portion of antibody, delivery has produced anti-tumor responses in animal
this approach requires whole intact Ig or an Fc segment, models [308]. Circulating target cells coated with antibody
rather than antibody subunits. As noted earlier in this are removed in the liver and spleen, presumably because
chapter, in the era of chimeric, humanized, and human of Fc receptors on tissue macrophages in these sites. For
Mab, human Fc constructs can be selected to optimize human Mab, the ability to bind specifically to macrophages
for complement binding if that is considered desirable. of the reticuloendothelial (RE) system is restricted to
IgG1 and IgG3 antibodies and is absent from IgM and
Antibody Dependent Cell-Mediated other IgG subclasses. There is evidence that mouse Mab
differ in their subclass binding to human effector cells.
Cytotoxicity
Various studies have confirmed that among murine
The second immunological cytotoxic mechanism is Mab, the hierarchy for effecting ADCC is IgG2a, followed
ADCC [304, 305]. Many lymphocytes, monocytes, by IgG3, apparently because of enhanced Fc receptor

COMPLEMENT-DEPENDENT ANTIBODY MEDIATED


TUMOR CELL LYSIS

Na+
C
K+
H2O
IgG

Figure 5. Diagram of complement


mediated cytotoxicty (CMC) featuring
antibody binding, fixation of complement, TUMOR CELL MEMBRANE TUMOR CELL
and cell death & ANTIBODY DAMAGE LYSIS

ANTIBODY-DEPENDENT CELL - MEDIATED


CYTOTOXICITY (ADCC)

Fc RECEPTOR

Figure 6. Diagram of antibody


dependent cell-mediated cytotoxicity LYMPHOCYTE
featuring antibody binding,
attachment of effector cells via the
Fc receptor to the Fc tail of the LYSIS
antibody molecule, and cell death NOT C’ DEPENDENT
Robert O. Dillman 313

binding [308, 340]. In addition, different antibodies of advantages and for preventing or delaying apoptosis that
the same subclass and isotype, directed against the might be targeted for antibody therapy [269, 480, 568,
same antigen, exhibit different degrees of Fc binding 731]. Advances in understanding of cellular physiology
[179, 660]. Murine IgG3 anti-disialoganglioside anti- have led to identification of a number of ligand–recep-
bodies have been cytotoxic with human effector cells in tor–signal transduction systems in which a ligand binds
vitro [108, 305, 720]. Among commercially available to a receptor which initiates a series of enzymatic reac-
chimeric and humanized antibodies, most have been tions that signals the nucleus of the cell to activate genes,
engineered with a human IgG1 Fc portion that can effect translation, and transcription to produce proteins that
ADCC [691]. are important in cell function, differentiation, prolifera-
For both ADCC and CMC the in vivo anti-tumor tion, and apoptosis. Epithelial growth factor (EGF) and
effect is also dependent on the ability of the host vascular endothelial growth factor (VEGF) and their
immune system to provide sufficient complement receptors are some of the best known examples of such
proteins or effector cells to produce cytotoxicity. The interactions. Increased levels of these receptors have
use of various biological response modifiers may be been found in increased quantities in cancer cells and/or
useful in augmenting such an immune response. To other rapidly proliferating cells.
maximize efficiency of effector cell-antibody bind- Conceptually, Mab directed against growth factor
ing, some investigators have utilized leukopheresis receptors may either block or down-regulate the
to harvest leukocytes, and then incubated these with number of receptors on the cell surface, and thus
antibodies to enrich the antibody-effector cell popu- impair a cell’s ability to differentiate and/or divide,
lation. Various lymphokines, such as interleukins-2, -4, ultimately resulting in cell death or apoptosis. Such
-12, and -15 may be useful to enhance the cytotoxic antigens may also serve as targets for Mab that can
activity of the effector cells. It has been demon- induce CMC and/or ADCC, or as targets for cytotoxic
strated that cytotoxicity, either by ADCC or CMC, immunoconjugates. Many receptors internalize after
involves a threshold of antigen binding sites. their ligand or an antibody bind to them resulting in
Lymphokines, such as alpha and gamma interferon, what is sometimes referred to as “antigen modula-
can enhance target antigen expression in some tis- ton.” Rapid internalization limits the potential for
sues [180, 274, 275, 432, 519]. Gamma interferon CMC or ADCC, but may be an advantage for the
increases Fc receptors on effector cells and thereby internalization of conjugated cytotoxic chemotherapy
can enhance ADCC [181, 519, 767, 775]. There are agents or toxins.
good examples from animal models suggesting that
various biologicals may be used in combination to
maximize an anti-tumor effect. The agents most
Idiotype Network
commonly used for this purpose in human trials are One of the first targets proposed for a regulatory
interferon alpha, IL-2 and GM-CSF. These other bio- approach was the Ig idiotype of B cell lymphoid malig-
logicals might be used systemically, or targeted specifi- nancies based on the cascade of anti-idiotype antibodies
cally by other antibodies. involved in regulating B cell proliferation [65, 246, 247,
351]. Cells from a malignant B-cell clone express and
sometimes secrete a specific antibody with a unique
Regulatory Approaches binding ability. The peptide sequences in the hypervari-
Malignancy may result from the overproliferation of able region of the light and heavy chains of the antibody
cells, the failure of cells to undergo programmed cell are the idiotopes that collectively constitute the idiotype
death (apoptosis), or a combination of these. Antibody of that Ig, and account for its specificity. Under normal
modification of these events constitutes a second mech- conditions memory T cells signal B cells to produce Ig
anism of antibody therapy, and is encompassed by the via surface idiotype receptors. Other B cells may pro-
term “regulatory,” in contrast to “immunologic,” to duce anti-idiotype antibodies that may be important in
characterize this potential mechanism [163]. Such negative feedback control of a given B-cell clone that
receptor targets can also be associated with cytotoxic has been activated. A detailed description of the “net-
effects mediated by complement and/or effector cells. work theory” of anti-idiotype regulation is very com-
In contrast, ligand or microenvironment targets may plex, and beyond the scope of this chapter. In its simplest
produce anti-tumor effects purely by regulatory biologic construct, infusion of an antibody directed against a
effects. It is known that tumor cells have a variety of B-cell lymphoma idiotype might suppress (growth reg-
receptors that are important for growth or proliferative ulate) that clone back to its baseline state by inducing
314 Monoclonal antibody therapy

Antigenic
stimulation

B-Cell proliferation
and differentiation Ig

Antigenic
stimulatioin

Anti-idiotype

Figure 7. Diagram of anti-idiotype feedback inhibition of B-cell clonal proliferation, based on the specificity of the hypervariable
region of the antibodies produced by that clone of cells

apoptosis and/or limiting proliferation. How this might such products are discussed in more detail in this book
relate to B-cell malignancy is depicted simplistically in in the chapter on vaccines.
Fig. 7. It is also important to note that the idiotype of a
given B-cell tumor is perhaps the most tumor specific Epidermal Growth Factor and its Receptors
antigen that has been identified.
Idiotype network theory has led to a concept of taking
(EGF and EGFR)
advantage of idiotype targets as a form of passive immu- Another heavily studied regulatory system is the human
nization [404]. According to the “idiotype network” epidermal growth factor receptor (EGFR) family of four
theory, the idiotype of an Ig is very immunogenic and transmembrane receptors abbreviated as Her1, Her2,
important in negative-feedback self-regulation. Injection Her3, and Her 4 or Erb-B1, Erb-B2, Erb-B3, and Erb-
of a mouse antibody (AB1) directed against a patient’s B4 respectively), which are involved in regulation of
tumor antigen will lead to production of many different cell proliferation and survival. EGFR are overexpressed
antibodies, but some will be anti-idiotype antibodies on the tumor cells of subsets of patients with most epi-
(AB2) directed against the hypervariable determinants thelial solid tumors [608, 611]. The prototype for this
of the AB1. Some of these AB2 may present the same family of receptors is Her1 (a.k.a. EGFR or EGFR1)
three-dimensional antigenic structure as the original which binds to a variety of ligand growth factors includ-
tumor antigen. Subsequently, such AB2 anti-idiotype ing epidermal growth factor (EGF), transforming growth
antibodies may induce anti-anti-idiotype antibody (AB3), factor beta (TGF-β), epiregulin, betacellulin, and amphi-
which because of the “lock-and-key” structural relation- regulin, which activate the cytoplasmic tyrosine kinase
ship between antigen and antibody, will have the same domain of the receptor, resulting in downstream activa-
binding specificity as the original mouse antibody tion of various signal transduction intermediates that
(AB1), except that it will be a human antibody (AB3) result in cell proliferation and other functions associated
[316, 781]. A more direct approach using this same with malignancy [471, 480]. This is the target of the
principle involves simply taking a well-characterized commercially available antibodies cetuximab (Erbitux)
anti-anti-idiotype antibody (AB2) and directly injecting and panitumumab (Vectibix). Her2 (a.k.a. Her-2 neu)
it as an immunogen to try to induce a human AB3 that originally attracted interest when it was found to have
will react with the tumor antigen. This theoretical oncogenic properties and to be over expressed on tumor
approach has been demonstrated in animals and may cells from about 25% of breast cancer patients. [632,
have application in both cancer and autoimmune dis- 673]. Her2 is unique among the EGFR family in that it
eases [314, 378]. Early clinical trials testing this has no known ligand, but instead can bind with each
approach have been associated with successful produc- of the other EGFR (a process called “dimerization”)
tion of sufficient antibodies to produce immunologic which also induces signal transduction. When over
and anti-tumor effect in man. [39, 104, 229, 230, 493, expressed, Her2 can dimerize with itself to induce the
494]. This concept and results of clinical trials testing tyrosine kinase activity that leads to signal transduction.
Robert O. Dillman 315

Heregulin is a known ligand for Her3, which unlike the Table 5. Issues for clinical application of monoclonal antibodies
other heregulin receptors, has no tyrosine kinase activ- Affinity and avidity of antibody for tumor antigen
ity. Neuroregulins 1, 3, and 4 and epiregulin are ligands Ability to effect complement and/or antibody-dependent cell
for Her4. mediated lysis
Biological significance of the antigen to the malignant cell
Biological behavior of the antigen (secreted, internalized, rate
Vascular Endothelial Growth Factor and its of production)
Density of antigen expression
Receptors Dose and schedule of administration
Expression of antigen on normal tissues
Judah Folkman hypothesized that angiogenesis was Human anti-antibody response (HAMA, HACA, HATA, etc.)
critical for growth and metastasis of tumors. [226]. Heterogeneity of antigen expression
The angiogenesis-inducing ligand vascular endothe- Location, size, and vascularity of tumor mass
lial growth factor (VEGF) and its cell membrane Therapeutic index: efficacy vs. toxicity
receptor, vascular endothelial growth factor receptor Total body burden of tumor and antigen
(VEGFR) were predicted to be good targets for anti-
cancer therapy [218, 634]. The commercially avail-
able monoclonal antibody bevacizumab (Avastin)
binds VEGF. VEGF (a.k.a. VEGF-A) is one of a fam-
ily of related proteins that induce angiogenesis and/or
Antitumor Effects
lymphoangiogenesis via binding one of several related In Vivo Binding to Malignant Cells
receptors.
VEGF is the central mediator of tumor angiogenesis, In association with intravenous (i.v.) infusions of anti-
which is crucial for proliferation, invasion, and metasta- bodies, several studies used fluorescein conjugated anti-
sis. It is produced by cells from many tissues and mes- mouse antibodies to show that many of these products,
senger RNA levels of the VEGF gene are over expressed especially those resulting from immunizing animals
in most tumors. Elevated levels of VEGF are predictive with malignant hematologic cells, did bind to circulat-
of poor prognosis. A variety of experiments suggested ing blood tumor cells and cells in the bone marrow in
that this ligand would be a useful target for monother- man [19, 38, 161, 177, 488, 518, 600]. Mab binding to
apy, or in combination with other therapeutic modalities tumor cells located in lymph nodes, tumor masses, and
[12, 55, 248, 420, 681]. skin infiltrates were directly demonstrated using immu-
nofluorescence and immunoperoxidase techniques [179,
328, 375, 513, 535], and indirectly with low doses of
Considerations for Clinical use Mab conjugated to technetium, indium or iodine, as
radioactive tracers [92, 183, 286–289, 417, 427, 451,
of Monoclonal Antibodies 496, 514, 515, 719]. The relative specificity of this
uptake was established by failure to image any “false
as Cancer Therapy positive” lymph node sites, and the successful imaging
As summarized in Table 5, there are a number of factors of nonpalpable nodes that subsequently were proven to
that must be considered, when choosing an antibody for contain cancer by surgical excision and histopathology
clinical development, and there are a number of specific evaluation [183, 719]. Further proof of binding specific-
issues associated with therapeutic application. This sec- ity was shown in a patient with T-cell lymphoma, in
tion focuses on the general principles of antibody ther- whom a radiolabeled anti-CD5 murine Mab was con-
apy that were, for the most part, established during centrated in lymph nodes, but injection of a radiolabeled
clinical trials conducted between 1980 and 1989 with murine anti-melanoma Mab of the same subclass was
Mab directed against antigens found on cells from not associated with lymph node uptake [92]. Various
hematologic malignancies [19, 38, 161, 177, 179, 180, studies showed that antibody binding was more easily
227, 328, 446, 478, 485–488, 518, 569, 600], and solid demonstrated in cutaneous tumors than in solid tumors
tumor cancers [109, 161, 205, 240, 262, 263, 375, 439, located in viscera or in lymph nodes [535]. This may
513, 535, 625, 651–653, 666, 738, 776]. Many of the relate to cutaneous blood supply, but it could also relate
studies used products that reacted with CD5 [38, 177, to the relatively small size at which cutaneous tumors
179, 227, 228, 488, 486, 488], or anti-idiotype antibod- can be recognized. Some investigators have suggested
ies [446, 478, 487], or the antibody 17-1A against ade- that direct infusion into the lymphatic system may be
nocarcinomas [109, 240, 439, 651–653, 738]. superior to i.v. infusions in certain disease settings
316 Monoclonal antibody therapy

[719], but this is not a practical option because of the In patients in whom tumor regressions have been
technical difficulties associated with such an approach. seen, the precise mechanism of antibody-mediated
In general, after an i.v. infusion of a large quantity of tumor regression is difficult to ascertain. Anti-leukemic
Mab, the hierarchy of binding to antigen-bearing cells is effects are dependent on the Mab used, the rate and dose
circulating blood cells > bone marrow > skin > lymph of Mab infused, the density of antigen expression, and
nodes > small tumor nodules > large tumor masses. whether there is circulating antigen or endogenous anti-
There has been great interest in finding ways to over- bodies that bind to the therapeutic antibody. In earlier
come the interstitial pressure effects found in larger trials of mouse Mab, given in limited quantities, in most
tumor lesions, which impair penetration of antibodies instances the anti-leukemic responses were relatively
into the core areas of such tumors [52, 345]. transient, but administration of chimeric and humanized
Mab in large quantities sufficient to sustain a large phar-
macokinetic “area-under-the curve” for prolonged peri-
Clinical Efficacy and Mechanisms ods of time, have been associated with meaningful
Significant antitumor responses were relatively uncom- durable clinical response. In the absence of sustained
mon in early trials with mouse Mab, but most of these levels of Mab, antibody-coated target cells are removed
involved relatively limited dosing in pilot, phase I, or from the circulation, but are rapidly replaced by cells
limited phase I/II trials, and were not definitive tests of from other organs such as bone marrow, lymph nodes,
Mab as cancer treatment. Nevertheless, it was encour- and possibly spleen. Some cells may survive by traffick-
aging that tumor responses following murine Mab ther- ing to other sites, and then return to the circulation. For
apy were reported for both hematologic malignancies non-modulating antigens, the leukemia cell count
and solid tumor cancers. Some of the best responses in remains depressed as long as Mab levels persist in the
these early trials took place in patients with follicular circulation. However, for modulating (internalizing)
(nodular) lymphoma and malignant melanoma, two antigens, the cell count begins to recover in association
diseases in which the frequency of spontaneous regres- with entry into the circulation of cells with a lower den-
sions had suggested an existing antitumor immune sity of antigens. Such modulated leukemia cells subse-
response [699]. quently re-express the target antigen in vitro or in vivo
As a general observation, when Mab bind in suffi- once the antibody concentration has dropped to a negli-
cient number or density to cells in the peripheral blood, gible level [639, 663]. Antigen modulation (cycling,
those targeted cells are removed in the reticuloendothe- internalizing) has been a limiting factor for many anti-
lial system. Human trials showed that infusions of 51Cr bodies directed against hematopoietic malignancies, but
or 111I-labeled autologous tumor cells resulted in marked is not an issue for Mab that react with stable membrane
uptake of the isotope label first in the lung, then in the antigens, especially when the target antigen is important
liver and spleen [161, 488, 518]. Investigators rarely as a receptor for cell proliferation, or helps a cell delay
could detect associated decreases in complement levels, or avoid programmed cell death.
although Ritz et al. [600] found deposits of C3 on mono- The precise explanation for the responses seen in
clonal antibody-coated cells in one ALL patient, and B-cell lymphoma patients treated with anti-idiotype
complement deposition was demonstrated in tissues in Mab has not been clearly defined. Idiotype is a modulat-
colon cancer patients receiving KS1/4 [205], and in ing target, which led investigators to adopt therapeutic
melanoma patients receiving the R24 antibody [738]. strategies of repeated low dose therapy so that target
Changes in the viability of circulating target cells have cells would have a sufficient density of antigen to effect
been observed following monoclonal antibody binding, CMC or ADCC. Investigators postulated either a direct
although this is difficult to demonstrate because of tim- cytotoxic effect or a regulatory effect via the idiotype
ing and technical reasons [19, 518]. These studies sug- network [65, 246, 247, 351]. The most responsive group
gest that binding of Mab to peripheral blood cells may was follicular lymphoma. Analyses suggested that those
actually damage the cell membrane and that such cells follicular lymphoma patients who responded had a
are then removed in the reticuloendothelial system greater infiltration with T cells prior to therapy [478].
rather than lysing in the intravascular compartment. The Responses were more readily achieved in patients with
best evidence of cell destruction, rather than sequestra- very low levels of circulating idiotype, which is proba-
tion, comes from studies following radiolabeled autolo- bly indicative of dose threshold related to antigen bur-
gous tumor cells [177, 488, 518], and the finding of an den. As discussed later in this chapter, even a decade
increased lactate dehydrogenase (LDH) associated with after its approval, it is still not clear whether immu-
decreased AML cells [19]. nolgic and/or regulatory effects are more important for
Robert O. Dillman 317

the clinical effects observed with the anti-CD20 chimeric right axillary recurrence. There was uptake of 111Indium
Mab rituximab. anti-p240 in the right axilla but there was also uptake
Although the best responses in patients with solid in the left axilla, which was clinically negative. Sub-
tumors have been achieved with Mab that effect both sequently, a small left axillary node was palpated but
ADCC and CMC in vitro, regulatory mechanisms spontaneously regressed. No other lymphadenopathy
related to the function of the target antigen (receptor) was visualized by imaging and no other lymphade-
may be more important. For example, the mechanism nopathy was palpated by examination. Both axillae
for the anti-tumor effects that leads to the excellent clin- were explored with confirmation of tumor in the right
ical results seen with antibodies to human EGFR are axilla, but no tumor was found on the left.
probably related to the importance of those receptors in There are at least four possible explanations for these
sustaining proliferation. Secondary immunologic effects intriguing observations of what clearly were antigen-spe-
may also be important. Sequential biopsies from mela- cific interactions. First, there may have been a local anti-
noma patients who exhibited a clinical response to the tumor effect against microscopic tumor cells. Second,
R24 mouse antibody revealed increasing infiltration there may have been antibody binding to residual antigen
with CD3+, CD8+, Ia+ T cells in the presence of degranu- retained in draining regional lymph nodes. Third, there
lated tissue mast cells [779]. Complement deposition may have been antibody reaction with regional B lym-
was also noted. Even though responses were seen within phocytes that were expressing anti-idiotypic antibody to
2 weeks of beginning therapy, tumors continued to endogenous anti-p240 or into p97 antibodies. A fourth
recede well beyond the treatment period despite the possibility is that the uptake was in macrophages or den-
presence of HAMA. These responses may involve a dritic cells that had been loaded with tumor antigen.
more complex interaction with the host immune system, The mechanism for the beneficial clinical effects of
perhaps triggering a cascade of inflammatory events anti-VEGF antibodies is also more complex than origi-
including activation of T lymphocytes that persist for a nally thought. It was hypothesized that blocking VEGF,
long period of time [318], or they could be inducing so that it could not bind to VEGFR, would interfere with
anti-apopotic and/or anti-proliferative effects. It has tumor vascular supply and starve the tumor of oxygen,
been suggested that mast cells not only have local stimulating factors, and other nutrients. There is now
inflammatory effects, but also may function as immu- some evidence that changes in afferent and efferent blood
noregulatory cells in a an immune response [243]. vessels themselves may be important more important in
Unfortunately, most advanced cancer patients do not concentrating chemotherapy at the tumor site than anti-
have tumors that are as easy to analyze sequentially as tumor effects from altered tumor blood supply.
melanoma. Selection of patients with primarily lymph
node or soft tissue disease facilitate prospective studies,
but they also result in unintentional bias because soft
Adverse Events: Toxicity and side Effects
tissue and lymph node metastases tend to be more The various adverse events observed with commercially
responsive to any intervention than bone, liver, brain, or available anti-cancer Mab are shown in Table 6. These
other visceral metastases. can be characterized as infusion reactions, acute or
Other interesting observations raise questions regard- delayed hypersensitivity reactions, or non-immunologic
ing the possible mechanisms of antitumor affects against biologic effects. It should be emphasized that the major
microscopic disease [183, 287]. A young patient under- adverse events associated with Mab are secondary to the
went a left radical neck dissection for regional spread of biologic effects induced by antibody–antigen binding
melanoma. Subsequently an anti-p97 antibody labeled rather than immune reactions to the antibody itself [170,
with 111Indium illuminated three apparent lesions in the 175, 184]. This explains why most of the toxic effects
right neck, although repeated physical examinations and described for murine Mab are also seen with the chimeric,
a computerized tomographic scan of the region were humanized, and human forms of Mab directed to the
negative for tumor. No other lesions or lymph nodes same antigen. Table 6 clearly shows that alemtuzumab,
were visualized, but 3 weeks later he developed three is associated with the most toxicity, even though it is a
palpable lymph nodes in the right neck in sites consis- humanized antibody, because it reacts with circulating T
tent with the scan. A neck resection was recommended cells and other mononuclear cells that release large
but he declined surgery, and the neck lesions subse- amounts of cytokines in response to the Mab binding to
quently resolved. He was known to remain free of dis- CD52 and the destruction of the cells by CMC and/or
ease for over 3 years. A second male patient had had a ADCC. Rituximab reacts with circulating B cells that
melanoma resected from his upper back and later had a are not as rich in inflammatory cytokines.
318 Monoclonal antibody therapy

Table 6. Percent of patients experiencing specific acute toxicities in clinical trials of monotherapy with a potpourri of murine and
human antibodies (177 patients with 20 different malignancies), rituximab, trastuzumab, alemtuzumab, bevacizumab, cetuximab and
panitumumab
19 Mo 3 Hum Ritux Tras Alem Beva Cetux Panitu
Antibody Mo/Hu Ximab Zumab Zumab Zumab Ximab Humab
Target Many CD20 EGFR-2 CD52 VEGF EGFR1 EGFR1
# of Patients 177 356 352 149 234 420 229
Tumor target Several Lymph Breast CLL Colo Colo Colo
Abdominal Pain <5 14 22 11 8a 26 25
Angioedema – 11 – – – – –
Arthralgia – 10 6 – – – –
Asthenia/malaise – 26 42 13 5a 48 –
Bronchospasm – 8 – 9 – – –
Chills/rigors 13 33 32 86 – 21 <5
Cough – 13 26 25 – 11 14
Diaphoresis 10 15 – 19 – 21 <5
Diarrhea – – – 2a 25 21
Dizziness – 7 13 12 – – –
Dyspnea – 7 22 26 – 17 <5
Fever 15 53 36 85 – 27 <5
Headache – 19 26 24 – 26 <5
Hypertension – – – – 8a – –
Hypotension – 10 – 32 – – <5
Myalgia – 10 – 11 – – –
Nausea 7 23 33 54 6a 29 23
Edema – 8 10 13 – 10 12
Pruritus 12 14 – 24 – 11 57
Rash 12 15 18 40 – 90 90
Rhinitis – 12 14 7 – – –
Throat irritation – 6 12 12 – – –
Urticaria 12 8 – 30 – – –
Vomiting 6 10 23 41 6a 25 19
Ritux = rituximab, Tras = trastuzumab, Alem = alemtuzumab, Bev = bevacizumab, Cetux = cetuximab,
Panitu = panitumumab
a
Bevacizumab data in this table reflects only grade 3 to 5 toxicity.

Because of the individual regulatory development of murine plasmacytoma cells has a higher rate of infusion-
these products, it was not always clear which adverse associated flu-like symptoms, such as fever, headache,
events were product-related as opposed to target-related, chills, and dyspnea, compared to the human antibody
and this has led physicians to attribute toxicity to the that is manufactured in CHO cells.
Mabs themselves, when, in fact, the effects were predict-
able because of the significance of the antigen target, and
the cascade of events that follow CMC and ADCC, many
Infusion Reactions
of which are due to the subsequent release of cytokines, These include a constellation of symptoms that predict-
as opposed to a hypersensitivity reaction to the antibody. ably occur during or shortly after infusion of a Mab.
However, there are allergic or hypersensitive differences The typical symptom complex includes fever, chills,
as well. For instance, cetuximab and panitumumab both sweats, prostration, nausea, and sometimes dyspnea and
target EGFR1, and have certain toxicities in common hypotension, which occur within a matter of minutes to
that relate to biologic effects associated with blocking hours after an i.v. antibody infusion is initiated. They
EGFR, such as acneiform skin rash and gastrointestinal are due to direct effects on tumor and non-tumor cells
symptoms including abdominal pain, nausea, vomiting which express the antigen, and indirect effects mediated
and diarrhea. However, the chimeric Mab that is made in by the secondary release of various cytokines as a result
Robert O. Dillman 319

of antibody binding to the target antigen, or formation and bioavailability of the murine Mab which limited
of immune complexes with circulating soluble antigen. binding to the target antigen. The observation that some
Most infusion reactions associated with Mab are non- F(ab’)2 can cause the same reaction suggests that anti-
allergic in nature, and rather are due to the interaction of body binding to antigen may be sufficient to induce
the antibody with circulating cells via antigen or Fc cytokine release even without interaction of the Fc por-
receptors, and perhaps endothelial cells. However, tion of the antibody in an ADCC reaction [721].
because of the effects of cytokines, it can be clinically Antibodies that react with circulating T cells, which
difficult to distinguish between allergic and non-allergic are more numerous and contain more cytokines than B
infusion reactions. For many years it has been clear that lymphocytes are associated with more severe infusion
most infusion-related adverse events are non-allergic in reactions. Binding to granulocytes also triggers severe
nature, and are directly related to antibody-antigen infusion reactions. As described earlier in this chapter,
interactions and the interaction with complement and studies with radiolabeled cells showed that once anti-
effector cells rather than being IgE, mast cell or eosino- body binds to circulating cells, they are removed in the
phil mediated reactions [170, 175, 184]. The antigen– reticuloendothelial system including the lung, liver, and
antibody binding to circulating cells in immunocompetent spleen [177, 485, 518]. When large numbers of cells are
patients, and subsequent interaction with complement removed in the lungs, this may be associated with dysp-
or effector cells, is predictably followed by the release nea and hypotension. For this reason, when it is known
of cytokines and/or the removal of those circulating that an antibody will react with circulating cells, the ini-
cells via macrophages in the reticuloendothelial system tial infusion rate needs to be slow, and high-dose bolus
[19, 38, 79, 177–179, 180, 227, 228, 435, 446, 478, administration is avoided. Once circulating target cells
485–488, 518, 569, 600, 787]. The onset of these reac- are no longer present, subsequent infusions may be
tions is typically within 15–30 min after initiating an safely given at more rapid rates. For instance, the author
antibody infusion, but depends on the dose and rate of has administered full dose rituximab over 30 min during
infusion, and the distribution of the target antigen. subsequent weekly treatments once B lymphocytes have
Typical adverse events associated with binding to cir- been cleared from the circulation, and others have con-
culating cells are the same as those seen with high doses firmed the safety of this approach in formal trials with
of various cytokines, such as interferon, interleukin-2, delivery over 45 to 90 min [82, 655].
or high doses of GM-CSF. These include fever, rigors/ For patients who are initiating therapy with a mono-
chills, sweats, maculopapular erythematous skin rash, clonal antibody that reacts with circulating cells,
urticaria, pruritus, edema, hypotension, tachycardia, premedication with steroids, acetamenophen and/or
headache, nausea, vomiting, diarrhea, fatigue, elevated diphenhydramine decrease the severity of symptoms
hepatic transaminases, throat tightness, pain, thrombo- associated with an infusion reaction, but does not typi-
cytopenia, dyspnea, bronchospasm, anaphylactic shock, cally prevent such a reaction [175]. Once the target cells
and even death [435]. The severity of infusion reactions are gone, there is no need to give such pre-medication.
vary greatly depending on the nature of the antibody It is possible that combined prophylactic therapy with
(mouse or human), the distribution and density of the histamine blockers and steroids starting 24 h before
target antigen on circulating cells, and the immunocom- treatment would reduce many of these toxicities, much
petence of the patient. Nearly all patients who have as they reduce the immune reactions associated with
received antilymphocyte and/or antigranulocyte anti- infusion of paclitaxel chemotherapy.
bodies have experienced such side effects if they had
levels of circulating target cells at the time of infusion.
The reactions are predictable whether a patient is receiv-
Tumor lysis Syndrome
ing murine, chimeric, or humanized antibodies, suggest- Tumor lysis syndrome is a constellation of symptoms
ing that the antigenic target is the most important and complications associated with the rapid cytotoxic
variable, although differences related to the Fc portion death of rapidly proliferating tumor cells [817, 818]. It
of the antibody are also important. The only time these has been well described in association with systemic
reactions were not seen was in patients repeatedly chemotherapy for rapidly proliferating malignancies
treated with murine Mab who had high titers of endog- such as acute leukemia, and patients with bulky tumor
enous antimouse antibodies that apparently neutralized loads of Hodgkin’s disease, large B cell lymphoma, and
the infused antibody [179]. Thus, the presence of endog- testicular cancer in which highly effective systemic
enous human antimouse antibodies actually prevented chemotherapy destroys large number of proliferating
many of these side effects by altering the pharmacology cells in a short time period. This is followed by increases
320 Monoclonal antibody therapy

in serum potassium, uric acid, and phosphates released such complications were rare, and are even less common
by the destroyed cells, with subsequent hypotension, in association with chimeric and humanized antibodies.
renal failure, and other medical complications that can Patients with a known history of allergic reaction to
be life-threatening or lethal. For single-agent unconju- rodents or their by products, were typically excluded
gated monoclonal antibody therapy, true “tumor lysis from trials of murine antibodies. Acute allergic reactions
syndrome” occurs when a patient is treated with an anti- have included anaphylactic shock, less severe anaphy-
body that targets large numbers of proliferating tumor lactoid reactions such as bronchospasm, and generalized
cells in the blood stream, but does not happen when pruritus and urticaria [170, 175, 184]. The more severe
non-replicating cells are removed from the circulation reactions can be successfully managed with epinephrine.
[170, 175, 184]. This is of particular concern in the set- Patients who experience full blown anaphylaxis during
ting of acute leukemia, lympholeukemias of mantle cell or shortly following an antibody infusion should not be
and large B cell lymphoma, and prolymphocyte-pre- retreated with the agent, but it is important that a predict-
dominate chronic lymphocytic leukemia (CLL) in which able infusion reaction not be confused with such a severe
large numbers of antigen-positive tumor cells are pres- allergic reaction. Dermatologic effects including pruritis,
ent in the circulation, where the antibody can opsonize diffuse or focal maculopapular rashes and urticaria may
large numbers of cells resulting in their lysis in the cir- be seen alone, or as part of a full anaphylactic reaction
culation and in the reticuloendothelial system [79, 179]. with laryngeal edema, hypotension, and bronchospasm.
Many of the descriptions of so-called “tumor lysis syn- Pruritus and urticaria alone typically resolve without
drome” in the literature were really predictable antigen– treatment, but may be responsive to diphenhydramine or
antibody infusion related reactions rather than true epinephrine.
tumor lysis syndrome with the complications that result
from lysis of large numbers or proliferating cells and Side Effects Secondary to Binding
release of intracellular products including high levels of
nucleic acids that lead to hyperuricemia [79, 787, 798].
to Non-Malignant Tissues
Antibody-induced tumor lysis syndrome has only been Adverse events may also occur as a consequence of
observed in the setting of large numbers of rapidly pro- direct effects on noncancerous tissue that also express
liferating circulating target cells; it has not been observed the target antigen. This has been especially true for
in the absence of a large leukemic component of dis- antibodies that cross-react with adenocarcinomas and
ease, probably because of the slower penetration of anti- cells of the gastrointestinal tract. Such antibodies have
bodies into tumor tumor masses [345]. Unless the been associated with a high frequency of diarrhea, nau-
patient is truly at high risk for tumor lysis syndrome sea and vomiting, abdominal pain, and even large and/
there is no need for prophylactic measures such as pre- or small bowel mucosal damage in some patients [109,
hydration, sodium bicarbonate to alkalinize the urine, 240, 593]. Other antibodies that are known to cross-
and anti-uric acid agents such as allopurinol or rasbu- react with antigens on neural tissue have been associ-
ricase. For example, in classical CLL, there are primar- ated with specific pain syndromes in some patients
ily mature lymphocytes in the circulation whose [109, 205, 651, 652]. Cross-reactivity with normal tis-
excessive number is more the result of being long-lived sue antigens is particularly a concern when antibodies
rather than hyperproliferation, and true tumor lysis syn- are conjugated to cytotoxic substances such as radio-
drome does not occur in that setting, even with the isotopes, chemotherapy agents, or natural toxins. For
removal of hundreds of thousands of cells in just a few instance, the incidence of gastrointestinal toxicity was
minutes, although, infusion reactions are common, and much greater when a frequently tested adenocarcinoma
can be life threatening in a patient who is medically antibody was given conjugated to a vinca chemother-
compromised by cardiopulmonary, hepatic, or renal dis- apy analog, or methotrexate, as compared to adminis-
ease. However, a CLL patient with a large number of tration of the naked antibody [205, 636]. Another
prolymphocytes is at risk for tumor lysis syndrome. adenocarcinoma antibody that cross-reacted with anti-
gens on neural sheaths, produced unacceptable neuro-
Acute Hypersensitivity Reactions toxicity when conjugated to the A chain of the natural
toxin, ricin [541]. Dermatologic toxicities are predict-
and Anaphylaxis able complications of agents that interfere with EGFR
Because the first Mab were mouse proteins, there was signal transduction, such as cetuximab and panitu-
great concern that infusion of these products would be mumab [343, 551, 650] and the anti-Her2 Mab trastu-
associated with acute anaphylactoid reactions. Fortunately, zumab is associated with cardiac toxicity [374, 658, 678].
Robert O. Dillman 321

Bevacizumab has been associated with hemorrhage, Antibody Issues


vascular thrombosis, hypertension, and proteinuria,
presumably all because of effects on blood vessels. Immunoglobulin Class and Subclass
[211, 538, 698]. The complications associated with
In lymphoma, murine anti-idiotype Mab of various sub-
these three commercially available antibodies are dis-
classes produced clinical responses [446, 478, 487]. In
cussed in more detail later in this chapter.
melanoma, only murine IgG3 Mab and humanized
anti-GD2 or anti-GD3 antibodies produced clear-cut
Immune complexes responses [109, 328, 738]. The responses seen with
murine anti-CD5 IgG2a Mab in T-cell lymphoma and
In some instances, Mab are given to patients who are CLL were limited [38, 177, 179, 180, 227, 228, 485,
known to have free or soluble circulating antigen. 486, 825]. Even though it appears that mouse IgG3, and
Examples include the circulating idiotype in lym- perhaps some mouse IgG2a Mab, have greater potential
phoma, and carcinoembryonic antigen (CEA), or to effect ADCC than other subclasses of murine Ig [308,
prostate specific antigen (PSA). Immune complexes 340, 511, 691], the preferred strategy is to use chimeric,
may also be formed because of HAMA, HACA, and humanized, or human Ig because of the advantages
HAHA. Only rarely have symptoms been noted in the associated with the human Fc. Some complement-fixing
presence of the immune complexes formed by the IgM Mab would be useful, but their size limits tumor
binding of antibody to circulating antigen, probably penetration in solid tumors, and administration of large
because of the small size of such complexes since the doses could lead to hyperviscosity. The issues related to
monoclonal antibody binds to only one determinant murine Mab are of decreasing importance since most
on the circulating antigen. For this reason, the pres- investigators and companies have converted their prom-
ence of circulating antigen is not a contraindication to ising murine Mab into chimeric or humanized Mab.
antibody treatment, although the binding to soluble Most of these conversions have been to a human IgG1
antigen greatly alters the pharmacokinetics of the subclass because these are predictably associated with
antibody. However, acute arthralgias, myalgias, nerve efficient CDC and ADCC [660]. Class and subclass
palsies, fever, and skin rashes have occasionally been switching of antibodies has enabled us to establish more
seen in this setting and attributed to the acute immune directly the importance of the binding of antibody to Fc
complex formation [170, 175, 184]. During trials of receptors on effector cells.
murine Mab, the presence of HAMA predictably
decreased the half-life of the antibody, but was rarely
associated with complications that could be attributed Human Anti-Immunoglobulin Response
to immune complexes.
Because they are foreign proteins, it was not surprising
that murine Mab produce human antimouse antibodies
Delayed Allergic Reactions and Serum (HAMA) responses in immunocompetent patients,
especially when large quantities are administered. Trials
Sickness involving single exposure and multiple exposure to Mab
Because of the anticipated production of HAMA, there confirmed that this is a substantial obstacle to repetitive
was concern that delayed reactions such as serum sick- therapy, especially in patients with solid tumors, who
ness would be a significant problem following infusion typically are less immunosuppressed that patients suf-
of murine Mab. Fortunately, immune complex compli- fering from hematologic malignancies. [131, 164, 185,
cations related to HAMA have been uncommon, but 382, 640, 664]. Using radioimmunoassay (RIA) and
they can occur [170, 175, 184]. Classic serum sickness enzyme-linked immunosorbent assays (ELISA), inves-
has been seen 2 to 3 weeks following exposure to mod- tigators detected HAMA in virtually all patients who
erate and high doses of murine Mab, and rarely after have received mouse antibodies except for those with
chimeric Mab. A typical symptom complex includes CLL, probably because of the immunodeficiency asso-
fever, malaise, arthralgias/arthritis, myalgias, maculo- ciated with that disorder. It also appears that HAMA is
papular erythematous skin rash, and fatigue. Proteinuria reduced in many B-cell lymphoma patients, and perhaps
has been rarely observed in these few patients and renal the anti-B cell antibodies contribute to this. However,
insufficiency is extremely rare. Serum sickness can be recent studies involving radiolabeled anti-CD20 murine
managed with non-steroidal, anti-inflammatory agents, Mab as therapy for previously untreated B-cell lym-
and corticosteroids in more severe cases. phoma patients resulted in 63% of patients exhibiting
322 Monoclonal antibody therapy

HAMA within a median of 3.3 weeks of being treated HAMA or HACA, or HAHA. Today there is no need to
[362]. This suggests that prior chemotherapy and/or try to obtain approval of a therapeutic mouse monoclonal
greater mutations of recurrent and progressive disease antibody unless one desires short pharmacokinetics such
induce a relative immunodeficiency that lowers the rate as in radioimmunotherapy to reduce the amount of total
of HAMA in more heavily treated patients. Interestingly, body irradiation [174]. These preparations are a signifi-
many CTCL patients who developed HAMA had previ- cant improvement over mouse antibodies, and even when
ously experienced a clinical response to treatment, per- HACA or HAHA occur, they tend to appear later and
haps because they are the more immunocompetent often at lower titers so that repeated intermittent therapy
patients, or perhaps because their disease was less resis- is possible over several months. Based on the large clini-
tant because of less exposure to chemotherapy. cal experiences with current chimeric and humanized
Once HAMA is produced, they effectively neutralize antibodies, it appears that the risk of HACA or HAMA is
most clinical effects of therapy, although targeting of only on the order of 1–2% of all patients treated, and this
tumor cells can still be demonstrated. It appears that a rarely interferes with treatment.
small percentage of antimouse antibodies react specifi- There is a clinical caveat associated with HAMA
cally with the idiotype of the mouse protein rather than because exposure to such antibodies may sensitize
only with specific murine Ig isotype determinants [404, patients who subsequently will have limited therapeutic
664]. As discussed earlier in this chapter, it has been sug- benefit and/or be at risk for allergic reaction if further
gested that this anti-idiotype cascade may contribute to antibody doses are given for diagnostic or therapeutic
the anti-tumor effect as an indirect vaccine [313, 404]. purposes. The HAMA may also confound the interpre-
Efforts to block the antimouse immune response tation of serum diagnostic tests such as those for pros-
with chemotherapy, radiotherapy, corticosteroids, and tate specific antigen (PSA) or carcinoma embryonic
cyclosporine, have not been successful. Some investiga- antigen (CEA) etc. that are measured using momabs.
tors suggested that infusions of high doses of antibody For instance, we have seen a patient develop significant
would eliminate the antimouse response, but this was HAMA that resulted in chronic T cell lymphopenia and
not borne out in other trials. When patients receive high aberrant PSA assays results secondary to exposure to
doses of antibody, they must be evaluated for a pro- the OKT3 mouse monoclonal antibody associated with
longed period of time because, depending on the assay an adoptive cell therapy in which cells were incubated
used, the excess of mouse antibody may block any evi- in vitro with the OKT3.
dence of antimouse antibodies during the early phase of
the HAMA, HACA, or HAHA response. Some investi-
gators have suggested that attachment of polyethylene Antigen issues
glycol may decrease the immune response [784], but
this not been confirmed in humans. In the presence of
Antigenic Modulation
HAMA, some investigators have combined plasma- Antigen modulation is a dynamic process in which sur-
pheresis with administration of higher doses of antibod- face antigen is decreased in the presence of excess anti-
ies, but this was of limited benefit. body. Electron microscopy with autoradiography and
125
Even with modern commercially available antibodies, I-labeled monoclonal antibody studies showed that
there is still the potential for human (HAMA) and/or modulation is the result of internalization of the antigen
human anti-chimeric antibodies (HACA) following and the bound antibody [599, 639, 663]. This is pre-
treatment with a chimeric antibody, and even human ceded by “capping” of the antibody–antigen complex, a
anti-human antibodies (HAHA) directed against allo- process during which the complex appears to localize to
typic human antigens on human Mab. Human immune one region of the cell surface. Mab directed against
responses that result in the production of HAMA, HACA, many hematopoietic surface antigens and growth factor
and HAHA are readily detected by immunoassays that receptors on solid tumors induce modulation in vitro
utilize the therapeutic antibodies in the assays used in the and in vivo. Modulation may in reality be an example of
treatment of patients. As expected, most of the antibod- antibody acting as a surrogate ligand for a receptor that
ies associated with chimeric antibodies are HAMA normally internalizes after binding to a ligand.
directed to the residual murine determinants. Most of the Modulation occurs within minutes of exposure to Mab,
antibodies in HACA or HAHA are directed to allotypic but is reversible once the monoclonal antibody is
epitopes and carbohydrates on the Fc portion. Fortunately removed from the system because of the ongoing pro-
the various chimeric and humanized Mab that have duction or recycling of antigen/receptor. This has been
entered the clinic have been associated with minimal demonstrated both in vitro and in vivo.
Robert O. Dillman 323

Antigenic modulation must be differentiated from Free Antigen


immunoselection that might result from the elimination
of antigen-positive cells, thereby leaving only cells that Many tumor antigens are either shed in small quantities,
express no or only low levels of the target antigen. The or may be released during apoptosis. Some tumor anti-
conditions for modulation, or antibody-mediated down gens are shed in large quantities, constituting potential
regulation of a receptor in vivo include presence of immu- blocking factors to monoclonal antibody target cell
noreactive Mab in the serum, and persistent existence of binding. Such immune complexes might also produce
tumor cells that exhibit absent or reduced expression of damage to normal tissues and organs, as well. Most of
the targeted antigen, but continued expression of another the hematopoietic antigens detected by Mab are gener-
phenotypic marker for the cell in question. For example, ally not shed to an extent that interferes with measure-
the C22 antigen on malignant and normal B cells modu- ment of monoclonal antibody serum levels. However,
lates after binding of antibody, such that CD22 becomes excess circulating antigen has been a practical problem
undetectable, but CD19 and CD20 are other markers of B for anti-idiotype antibodies in the treatment of lym-
cells that can be measured. In the absence of antibody, the phoma. Immediately following treatment, blood antigen
antigen must be re-expressed to prove there is no immu- levels decrease precipitously as they complex with anti-
noselection of cells. Thus, if the cells that now appear to body, but the remainder of the Mab have access to tumor
be CD22 negative because of modulation are placed in antigen. Thus, the obstacle of circulating antigen can be
cell culture, re-expression of CD22 proves that only mod- overcome with higher monoclonal antibody doses.
ulation has taken place rather than immunoselection of However, immune complex-mediated disease may
for a CD22 negative subclone of cells. occasionally be a complication of this approach.
Modulation is a dynamic process that consists of anti-
gen expression, antibody binding, antibody–antigen
internalization, re-expression of surface antigen, addi-
Antigen Heterogeneity
tional antibody binding, etc.; such that at any point in If a given tumor cell does not express the antigen
time there is always at least some amount of residual detected by a given monoclonal antibody, there is no
surface antigen expression, although this may be diffi- basis for monoclonal immune-mediated cytolysis or
cult to prove depending on the sensitivity of the assay receptor inhibition for that cell. For many years there
being used. This phenomenon has important implica- was great debate as to whether any single Mab could be
tions for passive monoclonal antibody therapy because clinically efficacious because of the tremendous hetero-
during modulation, there are insufficient quantities of geneity in human cancer cell phenotypes, especially for
Mab on the cell surface to effect target cell elimination solid tumors [220, 307, 522, 637]. Fortunately, in recent
by complement or effector cells. For circulating cells, years single antibody products have proven effective
there is a threshold of monoclonal antibody-binding that enough as single agents to gain regulatory approval.
is required before cells are eliminated. In fact, in some With our expanded knowledge of tumor phenotype and
cases, cells with a high density of target antigen are rap- tumor cell antigens, in the future it may be possible to
idly eliminated while cells lower in antigen persist and/ employ rationale combinations of Mab to overcome this
or enter the circulation so that total target cell count is problem [433, 534]. Ideally, such a combination or
relatively unaffected [177, 179, 180]. “cocktail” would include Mab in quantities directly
Modulation, or down regulation of receptors may be related to specific antigen expression on an individual
desirable for antibodies that act through a regulatory patient’s cancer cells rather than fixed proportions being
mechanism, perhaps by altering signal transduction. administered to each patient with a given disease.
Continued presence of antibody is needed to sustain this However, just as with chemotherapy, a combination
effect, which has important implications for duration of may only be effective if its individual components have
therapy. Rapid internalization greatly limits the therapeu- some antitumor effects of their own.
tic potential of antibodies that effect complement and/or
cell-mediated cytotoxicity, but may be useful or a neces-
sary condition for drug or toxin antibody immunoconju- Dose, Schedules, and Pharmacokinetics
gates. As a general rule, modulation is much more common
for hematopoietic cell antigens than solid tumor antigens,
Antibody Dose
but it appears that some of the best targets for treatment of There is a dose/response relationship at low monoclonal
solid tumors are modulating/internalizing antigens that antibody doses (<10 mg) because of the volume of distri-
function as growth factor receptors for tumor cells. bution, nonspecific uptake and metabolism, and the
324 Monoclonal antibody therapy

importance of number of antibody molecules on cell sur- be given over a few minutes with no untoward effects.
faces for complement, reticuloendothelial, or effector Prolonged continuous infusions or repeated bolus infu-
cell-mediated effects. At higher dose levels that provide sions are probably necessary for Mab that work via regu-
antibody excess, these dose–response relationships are latory mechanisms in order to keep receptors
no longer important, but duration of sustained serum lev- down-regulated or blocked. This may also be desirable in
els (area under the curve) is important for tumors with order to establish a gradient effect in treating solid tumors
non-modulating antigen targets. In the case of antigens because of tumor penetration problems. Bolus infusions
targeted for a regulatory affect (whether modulating or are well tolerated by most patients if the monoclonal anti-
not), sustained serum levels are needed to continuously body preparation is free of aggregates and there is no
block or down regulate receptors that are continually cross-reactivity with circulating cells. Intermittent bolus
being produced by differentiating cells. The cells may infusions may be preferred if the down-regulated receptor
emerge from stem cells or progenitor cells that do not needs to be re-expressed in order for the antibody/antigen
express the target antigen, and therefore not directly (receptor/ligand) interaction to produce an antitumor
affected by antibody therapy. Extrapolation from animal effect via CMC or ADCC. In the past, because HAMA
studies, and from studies of biopsies from patients was anticipated, some investigators adopted a strategy to
receiving mouse antibodies, suggested that gram quanti- deliver the maximum dose of murine Mab within 2 weeks,
ties of antibodies would be needed for a therapeutic because HAMA is generally detectable within 2 weeks of
effect because of the importance of tumor penetration initiating treatment. Modern chimeric and humanized
and saturation. Several investigators noted that direct in antibodies are able to be effectively delivered over many
vivo tissue binding (as opposed to binding to circulating weeks to months because HAMA, HACA, and HAHA
cells) could only be demonstrated at doses of 30 to 50 mg have rarely interfered with serum levels. So far there are
or higher [179, 180, 328, 535]. Some radiolabeled mono- no studies that have rigorously addressed the correlation
clonal antibody studies showed that more tumors were between dose, infusion rate, or schedule, even for the
imaged with doses of 10 mg or greater [183, 287, 514]. antibodies that are now in widespread clinical use.
The clinical successes of recently approved Mab have
been associated with delivery of high doses repeated at
relatively short intervals, such as every 1 to 2 weeks. At
Serum Levels and Pharmacokinetics
these doses and schedules, continually detectable serum Because of known antibody–antigen interactions, it has
levels of antibody are assured in most patients. It seems been easy to devise various enzyme-linked immunosor-
likely that there are critical thresholds for dose and/or bent assays (ELISA) and radioimmunoassays (RIA) to
area under the curve of sustained antibody levels to pro- measure serum levels of Mab during and following infu-
duce an anti-tumor effect. Whether the dose/response sions [25, 34, 205, 443, 665, 706]. Serum Mab levels have
relationship is important beyond those threshold levels is been easily detected except during low infusion rates, or in
unclear, but sustained serum levels is likely to be related the presence of high levels of circulating antigens, high
to duration of response in some settings. circulating tumor burden, or in the presence of HAMA. In
the leukemias, monoclonal antibody levels tend to fall
rapidly following an infusion because of continuing
Infusion Rates/Schedules absorption by circulating cells and entry of additional cells
Rapid infusions of Mab that bind to antigen on circulat- into the circulation. However, with some antibodies, with
ing blood cells and/or trigger endogenous immune or without the presence of antigenic modulation, serum
responses leading to cytokine release and cell activation Mab levels are sustained for many days to weeks depend-
via interactions with Fc receptors, can be life threatening. ing on the dose given. Using 24- to 48-h infusions, peak
[79, 170, 175, 178, 179, 184, 435, 787]. When antibodies Mab concentrations of several μg/ml have persisted for up
react with circulating cells, infusion rates greater than to 2 weeks. The significance of serum levels at any time
25 μg/ml are typically associated with infusion reactions. point must be viewed in the light of the variables listed
Impure preparations that contain microaggregates, pyro- above. In addition to tumor burden, other important vari-
gen, or other contaminants may also be more toxic if ables include circulating antigen, antigenic modulation,
infused rapidly, but this should not be a problem with and the production of antimouse antibodies. With the anti-
products approved for clinical use. Most investigators CD20 chimeric antibody rituximab, there is a clear asso-
have been satisfied with 2 to 6-h infusions for delivery of ciation between the area under the curve for the antibody
higher doses of antibody, but pure preparations of anti- and tumor response, and return of normal and/or normal
bodies which do not react with circulating blood cells can B-lymphocytes does not occur until serum levels of ritux-
Robert O. Dillman 325

imab are negligible [34]. The dose and schedule for the B-lymphocytes. In the following decade, eight addi-
antibodies in clinical use today are all given at high doses tional monoclonal antibody products obtained regula-
that sustain levels for several weeks to months. Chimeric, tory approval for the treatment of various malignancies.
humanized, and human Mab consistently demonstrate The rest of this chapter will focus on the six unconju-
superior pharmacokinetics, including higher peak levels gated Mab that have been approved by the United States
and prolonged sustained blood levels, compared to their Food and Drug Administration (US FDA) for the treat-
mouse counterparts. This is especially true following ment of malignant disease; radiolabeled antibodies and
repeated administration because of the negative effects of immunotoxins are covered elsewhere in this book.
HAMA on mouse antibody pharmacokinetics. Recent
studies suggest that recycling of IgG after interaction with
Fc gamma receptors may also play a role in sustaining Rituximab (Rituxan®, Biogen-Idec
serum levels [357, 422, 442]. Pharmaceuticals, San Diego, CA and
Genentech, South San Francisco, CA)
Commercially Available Rituximab and CD20
Unconjugated Mab for Treatment The anti-CD20 monoclonal antibody rituximab (Rituxan)
became the first commercially available monoclonal
of Human Malignancy antibody for the treatment of a human malignancy when
Although many were disheartened by the early observa- it was approved by the US FDA in November 1997, with
tions with murine Mab, the experience with these murine a marketing indication for B-cell lymphoma (Table 7)
products, and humanization of antibodies through [168]. The anti-CD20 murine Mab that originally had
molecular engineering, suggested that Mab would still been named 2B8, and later ibritumomab, was converted
live up to their potential [161, 163, 166, 167, 172, 173, to a mouse/human chimeric antibody and named ritux-
428, 533]. “Magic bullets” for cancer therapy became a imab [782]. Rituximab includes murine variable regions
reality in November 1997 when the anti-CD20 mono- from the anti-CD20 murine Mab ibritumomab and human
clonal antibody rituximab (Rituxan) became the first IgG1 kappa constant regions that have been combined
monoclonal antibody product approved by the US Food using recombinant DNA technology. The genetically
and Drug Administration for the treatment of a malig- engineered chimeric monoclonal antibody is manufac-
nant disease, namely B-cell lymphoma [168]. Ironically, tured in CHO cells. In vitro it is cytolytic against CD20-
this product was produced by the company IDEC positive cells in the presence of human complement and
Pharmaceuticals (San Diego, CA), which was originally human effector cells [260, 441]. Rituxmab’s binding to
founded to develop anti-idiotype antibodies, but instead CD20 also appears to have a regulatory effect on B cells
gained success on the basis of a highly effective anti- by promoting apoptosis [473, 661], and it appears to be
body that reacts with most normal and malignant more effective in this regard than other anti-CD20 antibodies

Table 7. Commercially available monoclonal antibodies for the treatment of human malignancies
Year FDA
Generic name Commercial name Antigen Antibody type Fc/Fv Ig type Cell production approved
Rituximab Rituxan CD20 Chimeric IgG1 CHO 1997
Trastuzumab Herceptin EGFR-2 Humanized IgG1 CHO 1998
Daclizumab Zenapax CD25 Humanized IgG1 CHO 1998
Alemtuzumab Campath CD52 Humanized IgG1 CHO 2001
Bevacizumab Avastin VEGF Humanized IgG1 CHO 2004
Cetuximab Erbitux EGFR-1 Chimeric IgG1 MP 2004
Panitumumab Vectibix EGFR-1 Human IgG2 CHO 2006
CD = cluster designation
EGFR = epidermal growth factor receptor
VEGF = vascular endothelial growth factor
CHO = Chinese Hamster Ovary
MP = murine plasmacytoma
326 Monoclonal antibody therapy

such as IF5 and anti-B1 [661]. The regulatory conse- rash associated with the binding of antibody to circulat-
quences of rituximab binding to CD20 may be more ing B lymphocytes. There was very limited toxicity dur-
important in vivo than the immunologic-based anti-tumor ing subsequent infusions. There was no evidence of
effects. In vitro the presence of rituximab was associated hypogammaglobulinemia or infections as a consequence
with enhanced sensitivity to chemotherapy which pro- of suppression of humeral immunity.
vided a strong rationale for combined modality treatment Retreatment with rituximab at a dose of 375 mg/m2
[156]. Rituximab is used alone or in combination with weekly for 4 weeks resulted in a response rate of 41%
chemotherapy in indolent B cell malignancies, and is among 40 patients who previously enjoyed a response to
used in combination with chemotherapy in aggressive B rituximab that had lasted 6 months or longer [149]. The
cell malignancies. 18-month median duration of response was even longer
CD20 is a 35 kD antigen with multiple transmem- than the 13 months noted in the pivotal trial, probably
brane domains that is expressed on normal and malig- because most of the patients had a smaller tumor burden
nant B cells, but not on non-hematopoietic tissue, B-cell and more favorable pharmacokinetics than the original
progenitors, plasma cells, T-lymphocytes, monocytes, population of patients treated with rituximab. There was
dendritic cells, or stem cells [782]. CD20 is neither no clear explanation as to why the remaining patients
internalized nor shed in significant amounts. No ligand failed to respond to retreatment, but resistance was not
for CD20 has been identified, but its over-expression due to loss of CD20 expression or HAMA or HACA
appears to be important for resistance to apoptosis of B against rituximab. There was no emergence of a CD20-
lymphocytes [473, 661]. negative subclone as a consequence of a treatment-related
immunoselection process. While occasional patients do
recur with CD20 negative tumors [145], this has proved
Clinical Trials with Rituximab to be an exception, and most patients retain CD20
Early Clinical Trials with Single-Agent positivity throughout the course of their disease. A study
of 13 patients in a similar trial conducted in Japan had a
Rituximab response rate of 38% with rituximab re-treatment but
Maloney et al. conducted the early trials with C2B8 the median duration of response was only 5.1 months in
(rituximab) [459–461]. In a dose escalation phase I trial this study [339].
of single i.v. doses from 10 to 500 mg/m,2 two partial
responses and four minor responses were observed
among 15 patients [459]. In a subsequent phase II trial,
Rituximab in Follicular Lymphoma
multiple doses of 375–500 mg/m2 were given i.v. weekly Rituximab is highly active as a single agent in the treat-
for 4 weeks [461]. There were four complete responses ment of previously treated and previously untreated fol-
and 16 partial responses among 34 evaluable patients licular lymphoma, as shown in Table 8 [15, 56, 97, 125,
for an overall objective response rate of 47%. Most of 147, 231, 251, 277, 460, 461, 474, 562, 790, 791].
the partial responders remained in remission for 4–12 In previously treated follicular lymphoma patients,
months following treatment. objective response rates have ranged from 46% to 63%
The pivotal trial for regulatory approval of rituximab [15, 97, 145, 231, 461, 474, 562, 790]. In previously
was a multi-institutional study involving 166 patients untreated follicular lymphoma patients response rates
with low-grade or indolent lymphoma, mostly follicular, are generally higher, ranging from 67% to 78% [125,
who had relapsed after prior chemotherapy [474]. This 251, 277, 791]. Most of these trials utilized the standard
trial yielded an objective response rate of 50% with a 4-week course of treatment at 375 mg/m2, but Piro et al.
median duration of response of more than a year; 90% of used an 8-week treatment schedule [562], Aviles et al.
responses lasted more than 6 months. Patients in these and Bremer used a 6-week treatment schedule [15, 56],
trials had already failed a median of four prior chemo- and Hainsworth et al. utilized additional 4 weeks of
therapy regimens. The response rate in patients who had therapy every 6 months for 2 years [277]. Pooling data
failed high-dose chemotherapy and stem cell transplant from similar trials shows a response rate of 29/37 (78%)
was a remarkable 73% (18/23). None of the responding for untreated patients who received more than 4 weeks
patients relapsed in less than 4 months, and 10% remained of therapy, 100/143 (70%) for untreated patients who
in continual remission for 2–5 years. One third of patients received the standard 4 weeks of therapy, 59/89 (66%)
had their initial infusion interrupted because of the clas- for relapsed patients who received more than 4 weeks of
sical infusion reaction complex of fever and chills and therapy, and 238/458 (52%) for relapsed patients who
occasional shortness of breath, hypotension, and skin received the standard 4 weeks of therapy.
Robert O. Dillman 327

Table 8. Rituximab as a single agent in follicular lymphoma Table 9. Rituximab as a single agent in small lymphocytic
lymphoma
Proportion
Citation Clinical setting responding % Response Proportion
Citation Clinical setting responding % Response
[277] Untreated 29/37 78%
[15] Relapsed/ 13/17 76% [277] Initial therapy 15/23 65%
refractory [231] Relapsed/ 4/29 14%
[125] Untreated, low 36/49 73% refractory
tumor burden [562] Relapsed/ 1/7 14%
[791] Untreated 26/37 72% refractory
Grade 1
[474] Relapsed/ 4/32 13%
[562] Relapsed/ 21/30 69%
refractory
refractory
[251] Untreated 38/57 67%
[97] Relapsed/ 24/38 63%
refractory
[56] Relapsed/ 25/42 59%
refractory Table 10. Rituximab as a single agent in marginal zone
[474] Relapsed/ 75/130 58% lymphoma
refractory Proportion
[790] Relapsed/ 36/70 56% Citation Clinical setting responding % Response
refractory
[145, 147] Relapsed/ 12/22 55% [127] No prior 21/24 87%
refractory > chemotherapy
10 cm [469] Relapsed/refractory 20/26 77%
[231] Relapsed/ 32/70 46% gastric
refractory [127] Prior 5/11 45%
[251] Relapsed/ 59/128 46% chemotherapy
refractory

McLaughlin and Foran trials utilized the standard


4-week course of treatment at 375 mg/m2 [231, 474]
Long term follow up of the French trial was reported while the Piro trial used an 8-week course of rituximab
by Solal-Celigny et al. for 49 untreated follicular lym- [562], and the Hainsworth trial prescribed the standard
phoma patients with a low tumor burden who were therapy followed by planned repeat 4-week courses of
treated with the standard dose and schedule of ritux- therapy every 6 months for 2 years [277]. Patients in the
imab, 375 mg/m2 IV weekly × 4 [683]. Based on the relapsed/refractory setting experienced a relatively brief
recent Follicular Lymphoma Internal Prognostic Index duration of response, while those who received more
(FLIPI), 45% of these patients would be considered low than the standard 4 weeks as initial treatment for SLL
risk, 41% intermediate risk, and 14% poor risk. The enjoyed a much longer progression free survival.
eventual response rate was actually 80% with 49%
achieving a CR. Median PFS was 18 months and 34%
had an unmaintained remission that last more than 5 Rituximab in Marginal Zone Lymphoma
years, and 94% were still living at 5 years. Results of rituximab in mantle zone lymphoma are sum-
marized in Table 10. These indolent lymphomas can
Rituximab in Small Lymphocytic Lymphoma occur anywhere, but when they occur in the gastrointes-
tinal tract, they are referred to as mucosa-associated
Clinical results for rituximab in small lymphocytic lym- lymphoma tissue (MALT). In the Concani study the pri-
phoma (SLL), another of the indolent lymphomas, are mary lymphoma was located in the stomach in 15
summarized in Table 9. Objective response rates in the patients and was extragastric in 20 [127]. At study entry
setting of relapsed or refractory SLL were much lower 12 patients had Ann Arbor stage IE, 3 had stage IIE, and
than relapsed or refractory follicular lymphoma, but 20 had stage IV disease. The Martinelli trial included 26
high response rates were observed when rituximab was evaluable patients presenting with gastric MALT at any
used as initial therapy for previously untreated patients. stage, relapsed/refractory to initial treatment or not suit-
These patients are clinically distinguishable from CLL able for eradication with antibiotics for anti-Helico-
because they had less than 5,000 lymphocytes/μl. The bacter pylori therapy [469]. The median survival of 36
328 Monoclonal antibody therapy

months reflects the inclusion of previously untreated course of treatment 3 months later in patients who had
patients and exclusion of patients with marginal zone not progressed [187]. With standard dosing it often took
lymphoma from other sites. In contrast, in the Conconi several months before a response could be declared, and
trial, the progression free survival was 22 months for maximum responses were noted more than 12 months
those who had not received prior therapy compared to after starting treatment. In the trials that utilized repeat
12 months for patients who had relapsed after previous dosing, the median time to best response was about 18
anti-lymphoma therapy [127]. months. Pooled data showed that 31/56 (55%) had an
objective response when two cycles of treatment were
used compared to 34/116 (29%) for patients who
Rituximab in Lymphoplasmacytic received one standard 4-week course of rituximab.
Lymphoma
These tumors are characterized by secretion of IgM Rituximab in Large B Cell Lymphoma
with a range of clinical presentations that include a very Results of trials for single-agent rituximab in aggressive
high levels of IgM paraproteinenemia with minimal lymphomas are summarized in Table 12. A French trial of
lymphadenopathy (Waldenströms macroglobulenemia), 54 patients with aggressive lymphoma included 30 with
a chronic lymphoid leukemia, or an indolent lymphoma. large B cell lymphoma [121]. This was designed as a ran-
These lymphoplasmacytoid lymphomas previously were domized phase II trial in which all patients initially
a small subset of patients in the A group of the Working received 8 weeks of rituximab therapy instead of 4, but
Group Formulation. Unlike the cells in CLL and SLL one half were randomized to receive 375 mg/m2 weekly ×
and plasma cells, the B-lymphocytes associated with 8 weeks, and the other group received 500 mg/m2 on
this disease group have a high-expression of CD20. weeks 2–8 after an initial standard dose. There was no dif-
Furthermore, although this entity usually does have ference in response rate between the two groups; so, the
increased numbers of circulating B-lymphocytes of the response data was grouped together. The overall response
malignant clone, lymphocyte numbers typically are not rate was 31% with a median response duration of greater
as high as in CLL. Published reports of rituximab in the than 8 months. The response rate was 37% in large cell. In
treatment of lymphoplasmacytic lymphomas are shown another European trial a response rate of 25% was
in Table 11. Response rates from 16% to 65% have been observed in 28 patients with relapsed intermediate grade
reported with progression-free survival rates ranging lymphomas who were treated with the standard course of
from 16 to 30 months [187, 231, 249, 726, 728]. In an treatment with 375 mg/m2 weekly for 4 weeks [231]. The
earlier trial Treon used the standard 4-week dose and median duration of response was about 1 year.
schedule of rituximab treatment [726], but subsequently
used two 4-week courses of treatment during the first
and third months for all patients. Gertz used a single Rituximab in Mantle Cell Lymphoma
standard 4-week course of treatment at 375 mg/m2 [249] The randomized phase II French trial described above
as did Foran [231]. Dimopoulos gave a repeat 4-week included a 30% response rate for 12 patients with mantle

Table 11. Rituximab as a single agent in lymphoplasmacytic Table 12. Rituximab in large B-cell lymphoma (LBCL)and mantle
lymphoma cell lymphoma
Clinical Proportion Proportion
Citation setting responding Response rate Citation Clinical setting responding % Response
[728] Untreated and 19/29 65% [121] Relapsed large 11/30 37%
relapsed B cell
[726] Relapsed/ 11/22 50% [723] Relapsed large 21/57 37%
refractory B cell
[187] Relapsed/ 6/12 50% [231] Relapsed/ 13/40 33%
refractory refractory
[187] Untreated 6/15 40% mantle cell
[249] Untreated 12/34 35% [121] Relapsed/ 3/10 30%
[249] Relapsed/ 7/35 20% refractory
refractory mantle cell
[231] Relapsed/ 4/25 16% [231] Untreated 10/34 29%
refractory mantle cell
Robert O. Dillman 329

cell lymphoma who were to receive a planned treatment those treated at 1.0 to 1.5 g/m2, and 75% for those treated
course of 8 weeks [121]. Other investigators reported a at the highest dose of 2.25 g/m2, but patients who were
29% response rate in 34 patients with newly diagnosed treated at the higher doses were more often relapsed after,
mantle cell lymphoma, and a 33% response rate in 40 rather than refractory to, fludarabine. Huhn et al. reported
patients with mantle cell lymphoma that was either a 25% response rate using the standard dose and schedule
refractory to or had progressed after chemotherapy of four weekly doses of 375 mg/m2, but they restricted the
[231]. These patients were treated with the standard tumor cell load, as measured by the number of circulating
4-week course of treatment at 375 mg/m2. The median lymphocytes and the spleen size, in the first two cohorts
duration of response was about 1 year. of patients included in the study [333]. Median duration
of response was only 5 months.
A more practical and less expensive strategy in the
Rituximab in Chronic Lymphocytic setting of relapsed CLL is to use Rituximab in combina-
tion with chemotherapy or after a response has been
Leukemia
achieved with chemotherapy. Those strategies reduce
CLL and SLL are morphologically and phenotypically the risk of more intense infusion reactions that are asso-
the same with co-expression of CD19/20 and CD5, but ciated with higher numbers of CD20+ circulating cells,
clinically CLL is characterized by a peripheral blood and would not necessitate repeated or dose of rituximab.
lymphocytes count of greater than 5,000/μl. The density Response rates were much higher in previously untreated
of CD20 expression varies greatly among different CLL who also received more than four weekly doses of
patients. Theoretically higher or repeated doses would be rituximab. Thomas et al. treated for 8 weeks [716], while
needed to optimize the efficacy of rituximab in CLL Hainsworth et al. utilized planned retreatment with
because of the large sink of antigen positive circulating additional 4-week courses of treatment every 6 months
cells that would have to be eliminated before targeting for 2 years [277, 278].
disease in lymph nodes and other tissues. As summarized
in Table 13, in patients with relapsed CLL, standard and
high doses of rituximab have been associated with
Rituximab in Hairy Cell Leukemia
responses of 25–45%, but the duration of response only The circulating lymphocytes of hairy cell leukemia
lasted 5 to 10 months with standard dose rituximab [80, (HCL) typically express very high levels of CD20.
333, 529]. Byrd et al. reported a 45% response rate among Rituximab has been used to treat relapsed HCL using
33 patients who received thrice weekly therapy escalating both the 4 and 8 week treatment schedules. The 4-week
from 100 mg/m2 initially to minimize infusion reactions, schedule produced responses in 5/10 patients in one
up to the standard 375 mg/m2 three times a week [80]. All study [418], and in 6/24 patients who had all relapsed
but one of the responses was partial and the median dura- after cladbribine [819]. The 8-week schedule produced
tion of response was 10 months. O’Brien et al. observed responses in 8/15 relapsed patients [717].
36% response rate among 40 patients, with a median
duration of response of 8 months, in a dose escalation
study in which they administered up to 2.25 g/m2 weekly
Rituximab in Multiple Myeloma
for 4 weeks [529]. Response was correlated with dose: CD20 is rarely expressed on plasma cells, but has been
22% for patients treated at 500 to 825 mg/m2, 43% for reported to be present on malignant cells from up to
20% of patients with multiple myeloma [727]. Rituximab
Table 13. Rituximab as a single agent in chronic lymphocytic has rarely produced objective responses in multiple
leukemia myeloma even in patients whose plasma cells were felt
Clinical Proportion to overexpress CD20. Using the standard dosing of
Citation setting responding Response rate 375 mg/m2, Treon et al. saw objective responses in only
[716] Untreated, low 17/19 90% 1/19 patients [727], and Hussein et al. reported responses
burden only 2/21 patients prior to starting chemotherapy 5
[277] Untreated 18/26 70% weeks later [338]. There has been a much greater unpub-
[278] Untreated 25/43 58% lished experience in the community practice of medi-
[80] Relapsed 15/33 45% cine with anecdotal reports of occasional excellent
[529] Relapsed 14/40 36% responses in refractory myeloma patients whose plasma
[342] Relapsed 8/23 35%
cell overexpressed CD20. Another strategy is to use
[333] Relapsed 7/28 25%
rituximab as an adjuvant treatment after an initial
330 Monoclonal antibody therapy

response to chemotherapy, in an effort to suppress the B specific interactions with the CD20 antigen, the term
cell clone that leads to the malignant plasma cells. Based “maintenance” should be reserved for the use of addi-
on this same rationale, rituximab may be more active tional doses of rituximab to maintain serum levels of
against smoldering myeloma than myeloma with a high rituximab such that cells that express CD20 do not emerge
proliferative index. from pre-CD20-positive B cell clones. Theoretically
this should keep CD20+ clones from appearing, but also
keeps normal B-cell immunity suppressed since normal
Rituximab in Hodgkin’s Disease B cells also express CD20. No clinical trial has ade-
Hodgkin’s disease is a malignancy of B-lymphocytes quately addressed the issue of “maintenance” rituximab
[687]. Lymphocyte-predominant Hodgkin disease in a pure sense.
(LPHD) is characterized by indolent nodal disease that As noted earlier in this section, retreatment with ritux-
tends to relapse after standard radiotherapy or chemo- imab is often still effective at the time of recurrence in
therapy and the malignant cells of LPHD are CD20+. patients who had previously had an objective response to
As shown in Table 14, rituximab alone has been associ- rituximab [148, 339] which suggests that tumor reap-
ated with very high response rates in LPHD, but the peared only because levels of rituximab were insufficient
median progression free survival has usually been no to keep it suppressed. If this is correct, then, continued
more than a year [203, 586, 803]. Rituximab is also treatment with rituximab should prolong progression-
being explored in other Hodgkin’s histologies. In the free survival, but not necessarily overall survival. The
Younes trial patients had to have received at least two real question is, what strategy is best for preventing
prior treatment regimens, and received 6 rather than 4 becoming rituximab-refractory? True refractoriness
weeks of therapy [803]. would be defined by disease progression during treat-
ment with rituximab. For clinical trials purposes it also
has been defined not only as disease progression during
Extended therapy and Maintenance therapy, but also as progression of disease within 6
Rituximab months of treatment in a patient who had stable disease
The term “maintenance” has been used to describe the or a response, which has very different implications. The
use of rituximab therapy after induction therapy for mechanisms of resistance to rituximab appear to relate to
the treatment of B-cell lymphoma. However, much of (1) loss of cellular dependence on CD20 expression in
this usage actually represents adjunctive, sequential terms of regulation of cellular resistance to apoptosis
therapy, or consolidation therapy following an initial or (probable) (2) existence or production of CD20 at a rate
induction therapy that that did not include rituximab. that rapidly consumes standard doses of rituximab (prob-
Since the mechanism of action of rituximab relates to its able), (3) loss of CD20 expression (uncommon), or (4)
induction of neutralizing anti-rituximab antibodies (very
rare), (5) lack of complement (unlikely), or (6) insuffi-
Table 14. Rituximab as a single agent in Hodgkin’s disease cient or inadequate effector cells (unlikely).
Median Several trials suggest that patients who have not expe-
progres- rienced progressive disease while taking single-agent
Clinical Proportion % sion-free rituximab benefit from receiving more than the standard
Citation setting responding Response survival 4 weeks of therapy as summarized in the accompanying
[586] Relapsed, 12/14 86% >12 Table 15. Such approaches have been associated with
LPHD > months higher response rates and longer durations of progres-
30%
sion free survival than were reported with standard ther-
CD20+
[203] Untreated 12/12 100% 10
apy. However, the 95% confidence intervals for response
LPHD months rates overlap with those observed with the 4-week sched-
[203] Relapsed 10/10 100% 10 ule, thus, these results are not clearly better than those
LPHD months seen with 4 weeks of treatment. Randomized trials
[803] Relapsed 5/22 22% 8 months involving patients with follicular lymphoma show that
NSHD, 2 giving additional dose of rituximab after standard dosing
prior
treatments
does prolong PFS [251, 280]. In the Swiss trial the addi-
tion of one additional 375 mg/m2 dose every 2 months
LPH = lymphocyte predominate Hodgkin’s disease for 4 months was associated with a superior progression
NSHD = nodular sclerosing Hodgkin’s disease
free survival of 23 vs. 12 months (p = 0.02) and was even
Robert O. Dillman 331

Table 15. Rituximab treatment schedules utilizing more than the standard 375 mg/m2 weekly × 4 weeks in patients with indolent or
follicular lymphoma (no chemotherapy)
Schedule after 4 weeks of standard
Citation Clinical setting rituximab therapy Patients Response rate PFS
[389] Relapsed indolent 4 doses months 3 if PR or SD 25 76% –
[15] Relapsed follicular 2 more weeks 17 76% >34 months
[277] Relapsed indolent 4 doses months 6,12,18,24 60 74% 34 months
[280] Relapsed indolent 4 doses months 6,12,18,24 35 52% 31 months
[251] Initial & relapsed follicular 1 dose months 3,5,7,9 75 66% 23 months
[264] Relapsed indolent 1 dose monthly if <25 μg/ml 22 63% >25 months
[562] Relapsed follicular 4 more weeks 37 60% >19 months
[56] Relapsed indolent 2 more weeks 68 59% 14 months
PFS = progression free survival

more striking in patients who had not received chemo- group trial (RESORT, ECOG 4402) that also attempts to
therapy before (36 vs. 19 months, p = 0.009) [251]. address this issue in previously untreated low-risk indo-
To date the only trial that has tried to address the issue of lent lymphoma patients whose initial therapy is stan-
time to rituximab-refractoriness is the phase II random- dard-dose rituximab alone. Patients are then randomized
ized trial by Hainsworth et al. [280], in which patients to “maintenance” utilizing one 375 mg/m2 dose every 3
with relapsed low-grade lymphoma were treated with the months until relapse, instead of four doses every 6
standard 375 mg/m2 weekly × four dosing with ritux- months for 2 years, or are randomized to retreatment
imab, and then those who did not have progressive dis- with standard-dose rituximab at that time of relapse.
ease were randomized to either planned retreatment with The clinical trial definition of refractoriness is the key
rituximab every 6 months for 2 years, using the same endpoint, but this trial also attempts to address whether
dose and schedule, or to delayed treatment until disease prolonged inhibition of CD20-positive B cells beyond 2
progression occurred, then re-instituted rituximab ther- years impairs humoral immunity.
apy. This design of “maintenance” is not really “ratio- In terms of optimizing progression free survival, the
nale” since 6 months is too long to sustain serum levels optimal way to give “maintenance” rituximab would be
in some patients, and 4 weeks of therapy is excessive in based on sustaining serum levels in an effort to prevent
many patients in terms of rates of cellular catabolism and the emergence of CD20+ cells. However, a commercial
hepatic metabolism vs. interference with appearance of assay for serum CD20 levels is not currently available.
the CD20 molecule on the cell surface. For these reasons The only trial performed to date that utilized serum CD20
it is not surprising that there was no difference in the levels to guide re-treatment enrolled only 31 patients, and
time to becoming refractory to rituximab between the rituximab levels were only sustained for 1 year; so it did
retreatment every 6 months compared to retreatment not adequately address this issue [264]. In this trial ritux-
relapse. However, the additional rituximab therapy was imab serum levels were measured monthly, and if the
associated with a higher response rate 52% vs. 35%, level dropped below 25 μg/ml, an additional 375 mg/m2
complete response rate (27% vs. 4%). There was a strik- dose was administered. For the 22 patients who were
ing difference in the time to disease progression, medi- monitored as planned, half of the patients required addi-
ans of 31 months vs. 8 months (p = 0.007), but no tional dosing by 5 months. The response rate for 29 eval-
difference in time to become “rituximab refractory” uable patients was 59%, and was 63% for 22 patients
which was about 30 months in both arms, nor in overall with indolent histology. If a commercial assay for mea-
survival at a median follow up of 42 months. suring serum levels of rituximab became available, it
However, this was a small trial that was stopped early could become the major determinant for optimizing ritux-
because it was originally designed for patients with imab dosing for individual patients [585].
indolent lymphoma who had relapsed after chemotherapy In practice most physicians have adopted one or more
alone, at which time they were treated with rituximab. of the published schedules of maintenance therapy for
Once rituximab became a standard part of initial therapy their patients with indolent lymphomas, based on the
for patients with indolent lymphomas, there were no benefits in terms of response rates and progression free
longer any patients available that met eligibility require- survival, which is a highly valued endpoint by both
ments for the study. There is an ongoing cooperative patients and practicing physicians. However, most
332 Monoclonal antibody therapy

patients are not initially treated with rituximab alone, cyclophosphamide (PC) [186, 306, 370, 413], pentostatin
but rather with rituximab in combination with chemo- and mitoxantrone (PM) [160], cladribine with or without
therapy. The issue of maintenance rituximab therapy in cyclophosphamide [601], chlorambucil [464], chloram-
that setting for both indolent and aggressive lymphomas bucil, mitoxantrone, and prednisone (CMP) [317], infu-
is discussed latter in this chapter. sional etoposide, platinum, vincristine, cyclophosphamide,
and doxorubicin (EPOCH) [350, 786], cyclophosph-
amide, vincristine, doxorubicin, dexamethasone, metho-
Rituximab with Chemotherapy trexate, and cytarabine (hyperCVAD-MA) [605, 718]
In vitro studies showed additive and/or synergistic benefit ifosfamide, cisplatin, etoposide (ICE) [379], irinotecan,
from combining rituximab with chemotherapy. [156]. mitoxantrone, dexamethasone (IMD) [523]. As a gener-
Some studies suggest that the presence of rituximab helps alization, the addition of rituximab does not increase tox-
overcome resistance, perhaps because of altered efflux of icity in any significant way, although in randomized trials
chemotherapy drugs, or decreased resistance to apopto- there has been a slightly higher rate of neutropenia, but
sis. In the treatment of lymphoma, rituximab has been not febrile neutropenia. There also appear to be no advan-
combined with virtually every chemotherapy regimen in tages to sequencing chemotherapy and rituximab as
common use including cyclophosphamide, vincristine, opposed to giving the agents together, since they do not
and prednisone (CVP) [279, 465], cyclophosphamide, have any overlapping toxicities. Several trials have been
doxorubicin, vincristine, and prednisone (CHOP) [98, conducted that confirm the safety and efficacy of admin-
122, 139, 279, 319, 329, 423, 532, 729, 757], cyclophos- istering Rituximab in combination with chemotherapy in
phamide, doxorubicin, vincristine, prednisone and etopo- various B cell lymphoproliferative disorders.
side (CHOEP) [555], fludarabine alone [82, 141],
fludarabine and cyclophosphamide (FC) [373], fludara-
bine and mitoxantrone (FM) [476, 813], cyclophosph-
Indolent and Follicular Lymphoma
amide, mitoxantrone, vincristine, and prednisone (CMOP) Combinations of rituximab and a variety of chemothera-
[197], and fludarabine, chlorambucil, and mitoxantrone pies have produced high response rates with durable
(FCM) [233], pentostatin alone [193], pentostatin and progression free survival (Table 16). The combinations

Table 16. rials of chemotherapy with concurrent rituximab in patients with indolent lymphoma, mostly patients with follicular lymphoma
Citation Clinical Setting Chemo Regimen Proportion Responding Response Rate
[476] Untreated FMD 76/76 100%
[139,140] Relapsed/untreated CHOP 38/38 100%
[319] Untreated follicular CHOP 214/223 96%
[233] Recurrent follicular FCM 35/37 95%
[532] Untreated CHOP 32/34 94%
[141] Relapsed/Untreated Fld 35/39 90%
[197] Untreated CMOP 38/42 90%
[468] Untreated/relapsed Chl 24/27 89%
[729] Untreated/Relapsed LPCL CHOP 11/13 85%
[160] Untreated PM 20/24 83%
[465] Untreated indolent CVP 131/162 81%
[618] Recurrent follicular FC 37/54 81%
[725] Recurrent follicular FC 60/75 80%
[193] Relapsed Pento 44/57 77%
[306] Untreated/relapsed LPCL PC 10/13 77%
FMD = fludarabine, mitoxantrone, dexamethasone
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
FCM = fludarabine, chlorambucil, mitoxantrone
CMOP = cyclophosphamide, mitoxantrone, vincristine, prednisone
PM = pentostatin, mitoxantrone
CVP = cyclophosphamide, vincristine, prednisone
FC = fludarabine, cyclophosphamide
Clb = chlorambucil
Fld = fludarabine
Pento = pentostatin
C = cyclophosphamide
Robert O. Dillman 333

have not produced additive toxicity, but because of con- As summarized in Table 18, randomized trials have
cerns about the potential for such toxicity, other investi- confirmed a higher response rate and longer progression
gators have utilized rituximab after chemotherapy for free survival when rituximab is added to chemotherapy
patients who were already had a response to therapy. compared to results for the same chemotherapy alone.
This approach of chemotherapy followed by sequential Some studies also suggested there may be an improved
therapy or consolidation rituximab therapy also has overall survival as well. This improved survival has been
been associated with very high response rates and dura- accomplished without any clinically important increase
ble remissions (Table 17). in toxicity. Because of this rituximab with or without che-
Two randomized phase II trials have compared the motherapy has become the treatment of choice for patients
concurrent and sequential approaches but were not with indolent lymphomas who require therapy.
definitive in showing differences [471, 532]. The larger
of the two trials did show a slightly higher response rate
and longer progression free survival for the combined
Chemotherapy and Rituximab in Chronic
approach [471]. Since there has been no evidence of Lymphocytic Leukemia
additive toxicity for the combination regimens, and Although rituximab does not have an FDA regulatory
there is no evidence that the sequential approach offers marketing indication for CLL, numerous phase II trials
any therapeutic advantages, most physicians use the have shown that the combination of chemotherapy plus
concurrent approach. standard doses of rituximab has substantial activity in
Because of the prevalence of follicular lymphoma, the disease as shown in Table 19. This has included
more trials have been conducted with chemotherapy and purine analogs plus rituximab [82, 192, 601, 641], and
rituximab in this B cell malignancy than any other tumor combinations of alkylating agents, purine analogs, plus
type. Longer follow up has established that survival for rituximab [186, 370, 373, 413, 601, 783]. In one ran-
such patients has been improved since the introduction domized phase II trial, concurrent fludarabine plus
of rituximab in the salvage setting, and as part of initial rituximab was compared to the sequence of fludarabine
therapy of patients with follicular lymphoma [141, 327, followed by rituximab in 104 patients [82]. The response
619]. In a trial of 38 patients with indolent lymphomas, rate was higher for concurrent therapy (90% vs. 77%) as
80% of whom had not been previously treated, RCHOP was the complete response rate (47% vs. 28%). A retro-
was associated with a response rate of 100%, a median spective comparison of these patients to a similar popu-
progression-free survival of nearly 7 years, and after 9 lation of patients treated with fludarabine alone at the
years of median follow-up, median survival had not same dose in an earlier randomized trial suggested that
been reached [141]. patients treated with the combination of fludarabine plus
rituximab had much better outcomes, including a higher
response rate (84% vs. 63%), higher complete response
Table 17. Trials of chemotherapy followed by rituximab rate (38% vs. 20%) better progression free survival after
(sequential therapy) in patients with indolent B cell lym- 2 years (67% vs. 45%), and better overall survival at 2
phoma, mostly follicular lymphoma
years (93% vs. 81%) [83].
Citation Clinical Chemo Proportion Response
setting regimen responding rate
[813] Untreated CHOP 67/68 98% Large B Cell Lymphoma
FCL
[532] Untreated CHOP 34/35 97%
The three large randomized trials summarized in Table
[813] Untreated FM 69/782 96% 20 have demonstrated the benefits of combining ritux-
FCL imab with chemotherapy in the treatment of large B cell
[470] Untreated FMD 69/73 95% lymphoma. In France Coiffier et al. compared RCHOP
[754] Untreated FMD 34/36 95% to CHOP in 399 previously untreated patients, ages
[119] Untreated FC 29/33 88%
FCL 60–80 with large B-cell lymphoma, using an eight-
[273] Untreated FM 26/30 87% course schedule that included antibody and chemother-
[462] Untreated CHOP 71/84 84% apy all being given on day one of each course,
FCL prednisone, then Rituximab, then the cyclophosph-
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone amide, doxorubicin, and vincristine [122]. After 1 year
FM = fludarabine, mitoxantrone of follow up and 126 events (disease progression or
FMD = fludarabine, mitoxantrone, dexamethasone death), the CHOP plus Rituximab arm was superior in
FC = fludarabine, cyclophosphamide response rate (76% vs. 60%, p = 0.004), 1-year event-free
334 Monoclonal antibody therapy

Table 18. Randomized trials of chemotherapy ± rituximab in indolent lymphomas


Citation Clinical setting Treatment Regimen Proportion Responding PR Rate CR Rate PFS
[319] Untreated follicular RCHOP 214/223 96%
CHOP 184/205 90%
p = 0.011
[745] Relapsed follicular RCHOP 199/234 85% 30% 33 months
CHOP 166/231 72% 16% 20 months
p < 0.001 p < 0.001 p < 0.001
[465] Untreated follicular RCVP 131/162 81% 41% 32 months
CVP 91/159 57% 10% 15 months
p < 0.0001
[317] Untreated follicular RMCP 166/181 92% 50% >47 months
MCP 133/177 75% 25% 29 months
p = 0.0009 p = 0.004 p < 0.0001
[476] Untreated indolent RFMD 80 100% 88% 76% 3-year
FMD 80 96% 85% 60% 3-year
[233] Relapsed RFCM 52/66 79% 34% –
FCL & Mantle Cell FCM 36/62 58% 12% –
p = 0.01
[75] Untreated RCHOP 36 98%
Lympho- plasmacytoid
CHOP 36 69%
p = 0.01
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
RCVP = rituximab, cyclophosphamide, vincristine, prednisone
CVP = cyclophosphamide, vincristine, prednisone
RMCP = rituximab, mitoxantrone, chlorambucil, prednisolone
MCP = mitoxantrone, chlorambucil, prednisolone
RFMD = rituximab, fludarabine, mitoxantrone, dexamethasone
FMD = fludarabine, mitoxantrone, dexamethasone
RFCM = rituximab, fludarabine, chlorambucil, mitoxantrone
FCM = fludarabine, chlorambucil, mitoxantrone
PR = partial response
PFS = progression free survival
OS = overall survival

Table 19. Phase II trials of chemotherapy plus rituximab in CLL survival (69% vs. 49%, p < 0.0005) and overall survival
% (83% vs. 68%, p < 0.01). Longer follow up at a median
Clinical Median Proportion Resp- of 5-years confirmed the tremendous advantage of
Citation setting age Regimen responding onse RCHOP in terms of PFS and OS, without identification
[373] Untreated 58 years FCR 213/224 95% of any long-term toxicities [219].
[370] Untreated 63 years PCR 58/64 91% A similar trial was conducted by Habermann et al. in
[82] Untreated 63 years FR 46/51 90% a U.S. intergroup trial that enrolled 632 patients over the
[641] Both 59 years FR 27/31 87%
age of 60, except that a different schedule of RCHOP
[186] Untreated 72 years PCR 7/9 78%
[783] Relapsed 59 years FCR 138/179 77% was used, and for the 415 responding patients, there was
[413] Untreated 62 years PCR 26/34 75% a secondary randomization to maintenance rituximab
[601] Relapsed 55 years CCR 14/26 54% vs. observation for 2 years [276]. In this trial the RCHOP
[192] Relapsed 60 years PR 19/59 33% regimen was two cycles of rituximab followed by eight
[186] Relapsed 70 years PCR 2/12 17%
cycles of CHOP with three more cycles of rituximab
FCR = fludarabine, cyclophosphamide, rituximab given before the third, fifth, and seventh doses. Patients
FR = fludarabine, rituximab
PCR = pentostatin, cyclophosphamide, rituximab who had achieved a CR or PR by the end of eight cycles
CCR = cladribine, cyclophosphamide, rituximab were randomized to either observation or to four 4-week
PR = pentostatin, rituximab courses of rituximab at 375 mg/m2 per dose, given every
Robert O. Dillman 335

Table 20. Randomized trials of chemotherapy ± rituximab in large B cell lymphomas


Treatment Patients
Citation Clinical setting regimen enrolled PR rate CR rate PFS OS
[122] Untreated elderly RCHOP 399 86% 76% 53% 3-year 62%
[219] CHOP 71% 63% 35% 3-year 51%
p = 0.007 p = 0.005 p = 0.0008 p = 0.008
[276] Untreated elderly RCHOP 632 or 77% 35% 52% 3-year 67%
a
CHOP 425 76% 35% 39% 3-year 58%
p = 0.003 p = 0.05
[555] Untreated low-risk RCHOP 824 81% 76% 2-year 94%
young
CHOP – 67% 60% 2-year 87%
p = 0.0001 p < 0.0001 p = 0.001
[556] Elderly RCHOP-14 1222 70% 2-year
CHOP-14 57% 2-year
p < 0.0001
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
PR = partial response
CR = complete response
PFS = progression free survival
OS = overall survival
a
Response data are based on all 632 patients, but survival data is limited to 425 patients who did not receive “maintenance” rituximab
therapy, four weekly doses every 6 months for 2 years.

6 months for 2 years. For comparability with the French in Table 21. One of the earliest was conducted by Vose
study, an analysis was performed that excluded the 207 et al. in which rituximab was given on day 1 and CHOP
patients who received maintenance rituximab, which on day 3 of each of six treatment cycles to 33 patients
demonstrated a superiority of RCHOP in terms of PFS and with intermediate grade lymphomas, about two third
OS, although there was no difference in response rate. with large B-cell lymphoma (Working Group
Both of the above trials dealt with elderly patients. Formulation G) and the remainder with large cell fol-
Pfreundshuh et al. conducted a trial of rituximab and licular (Working Group Formulation D) [757, 758].
CHOP-like regimens compared to the same chemotherapy The trial by Case et al, in which patients received six
alone in 824 patients under the age of 60 years who had to eight cycles of RCHOP administered on the same
was considered low-risk large B cell lymphoma [556]. day, followed by 4 cycles of maintenance rituximab
Low-risk was defined as International Prognostic Index for responders, confirmed the activity of concurrent
0–1, stage II–IV disease, or bulky stage I disease. RCHOP therapy in the community setting [98]. The
CHOP-like regimens included the addition of etoposide Wohrer trial appears to confirm that RCHOP is highly
(CHOEP) or addition of methotrexate (MACOP). Any active in gastric large B cell lymphoma [792]. The
response less than a CR was considered a treatment fail- encouraging results from the REPOCH regimen have
ure. This trial also confirmed a substantial advantage for led to randomized trials comparing it to RCHOP [350,
patients who received rituximab with the chemotherapy. 786]. The encouraging results with 14-day RCHOP
In elderly patients Pfreundschuh et al. have conducted regimens, which are administered with prophylactic
a four-arm trial (RICOVER-60) comparing six and eight filgastrim or pegfilgastrim, has led to phase III trials
cycles of RCHOP-14 and CHOP-14 [557]. RCHOP-14 using this approach compared to 21-day schedules of
was clearly superior to CHOP-14, and six cycles of RCHOP [62, 283, 596]. Rituximab plus chemotherapy is
RCHOP-14 showed a survival advantage over eight also active in the salvage setting as evidenced by trials
cycles of RCHOP-14. What remains to be done is to with REPOCH, RICE and RIMD [350, 379, 523].
compare RCHOP-14 to RCHOP-21.
Many phase II trials have been conducted with Mantle Cell lymphoma
RCHOP and other rituximab plus chemotherapy regi- Table 22 summarizes some of the trials that have been
mens which also demonstrated excellent response rates conducted with concurrent rituximab and chemotherapy
as initial treatment or at the time of relapse, as shown in patients with mantle cell lymphoma. Howard et al.
336 Monoclonal antibody therapy

Table 21. Phase II trials of chemotherapy with concurrent rituximab in patients with large B cell lymphoma
Citation Clinical Setting Chemo Regimen # Patients Response Rate PFS OS
[757,758] 76% LBCL CHOP 33 94% 82% 5-years 88%-5-years
[792] Gastric LBCL CHOP 15 100%
[98] LBCL CHOP 101 91% 76% 2-years
[283] LBCL CHOP-14 49 >90%? 80% 2-years 90% 2-years
[596] LBCL CHOP-14 26 100% 70% 2-years 79% 2-years
[62] LBCL CHOP-14 50 >90% 68% 2-years 72% 2-years
[786] Initial, aggressive EPOCH 61 92% 82% 5-years 88% 5-years
[350] Relapsed or refractory EPOCH 50 68% 30% 2-years 40% 2-years
[379] Relapsed or refractory ICE 36 78%
[523] Relapsed elderly IMD 30 74% 37% 45%
LBCL = diffuse large B cell lymphoma
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
EPOCH = etoposide, platinum, vincristine, cyclophosphamide, doxorubicin
ICE = ifosfamide, cyclophosphamide, etoposide
IMD = irinotecan, mitoxantrone, dexamethasone
PFS = progression free survival
OS = overall survival

Table 22. Clinical trials of rituximab plus chemotherapy in mantle cell lymphoma
Treatment Proportion
Citation Clinical setting regimen responding PR rate CR rate PFS OS
[424] Untreated RCHOP 58/62 94% 34% 21 months NSD
CHOP 45/60 75% 7% 14 months
p = 0.005 p = 0.0002 p = 0.013
[329] Untreated RCHOP 38/40 96% 48% 17 months
[605] Untreated RHyper- 94/97 97% 87% 64% 82%
CVAD-MA 3-year 3-year
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
RHyper-CVAD-MA = rituximab, cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate,
cytarabine
PR = partial response
CR = complete response
PFS = progression free survival
OS = overall survival

showed that RCHOP was very active in mantle cell not only been used for mantle cell lymphoma, but also
lymphoma, but the PFS was not better than what had for the treatment of adult acute lymphoblastic leukemia
been seen historically with CHOP alone [329]. However, and Burkitt-type lymphomas. Thomas et al. treated 28
when Lenz et al. conducted a randomized trial, RCHOP evaluable patients with this regimen as initial therapy
was superior to CHOP in terms of response rate, CR [718]. Rituximab was given on days 1 and 11 of the
rate, and PFS, but not OS [424]. In a single institution hyper-CVAD courses and days 1 and 8 of methotrexate
trial, Romaguera et al. found their R-hyperCVAD-MA and cytarabine courses. The median age was 46 years,
regimen to be highly active [605]. Both RCHOP and but 29% were over the age of 60 years. The response
R-hyperCVAD-MA are used as initial therapy in rate was 27/28 with 86% complete responders. The
patients with mantle cell lymphoma, and most medi- 3-year progression free and overall survival was 88%.
cally fit patients go on to consolidation with high dose
chemotherapy and autologous stem cell rescue. HIV Associated Lymphoma
As summarized in Table 23, the addition of rituximab to
Burkitt’s Lymphoma and ALL chemotherapy has not been shown to improved outcome
The combination of rituximab and hyper-CVAD with in HIV-associated, non-Hodgkin’s lymphoma (NHL).
methotrexate and cytarabine (R-hyperCVAD-MA) has These relatively small trials do suggest some survival
Robert O. Dillman 337

Table 23. Trials of rituximab and chemotherapy in HIV positive patients with B cell lymphoma
Treatment Infectious
Citation Clinical setting regimen # Patients CR rate PFS OS death
[823] Untreated R-CDE 74 70% 59% 2-years 64% 2-years 8%
[53] Untreated R-CHOP 52 77% 75% 2-years 4%
[365] Untreated R-CHOP 75 58% 10.4 months 32 months 14%
CHOP 75 47% 8.6 months 25 months 2%
p = 0.147 p = 0.035
R-CDE = rituximab, cyclophosphamide, dexamethasone, etoposide
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
CHOP = cyclophosphamide, doxorubicin, vincristine, prednisone
CR = complete response
PFS = progression free survival
OS = overall survival

benefit, but also raise issues regarding an increased risk ten patients including a complete remission in four.
from infection that was not evident in non-HIV lymphoma Rituximab has also been used in the treatment of primary
patient populations. Spina et al. used 96-h infusion R-CDE CNS B-cell lymphoma. Limited benefits were observed
(cyclophosphamide 187–200 mg/m2/day, doxorubicin in patients with recurrent disease, and rituximab is now
12.5 mg/m2 per day, and etoposide 60 mg/m2 per day) in being combined with high dose methotrexate as part of
74 patients with HIV-associated NHL, most of whom initial treatment.
were receiving concurrent highly active antiretroviral
therapy (HAART) [823]. Ten patients were diagnosed Maintenance Rituximab following Chemo-
with opportunistic infections during or within 3 months of
the end of R-CDE, and 17 patients developed non-oppor-
therapy or Rituximab Plus Chemotherapy
tunistic infections for an infection rate of 37%. Two There has been interest in using maintenance rituximab
patients died of bacterial sepsis and four of opportunistic after an initial response to a combination of rituximab
infections. In contrast Boue et al. observed a 17% rate of and chemotherapy. As discussed earlier there also have
infection, but they did not address the risk of infection in been treatment strategies of chemotherapy followed by
the months after completion of therapy [53]. The one ran- rituximab, which is really a consolidation or sequential
domized trial that has been conducted showed a trend treatment approach rather than maintenance. Randomized
toward higher response rate and survival with the addition trials have failed to show an advantage for the sequential
of rituximab, but a higher rate of infectious death in the strategy compared to the combinations of rituximab with
group that received rituximab [802]. the same chemotherapy [276, 476, 532]. Compared to
observation, randomized trials have shown that adding
Central Nervous System Lymphoma rituximab after an initial response to CVP, FCM, or
In the absence of central nervous system (CNS) disease, CHOP is associated with longer PFS in indolent lym-
negligible levels of rituximab are detected following i.v. phoma [233, 322, 745], and adding rituximab after an
infusions, presumably because of the blood–brain bar- initial response to CHOP prolongs PFS in large B cell
rier. However, significant levels can be achieved in the lymphoma [276]. However, chemotherapy alone is no
presence of parenchymal disease or other inflammatory longer considered an appropriate initial option since
processes that alter the blood–brain barrier, or after rituximab plus chemotherapy is consistently superior to
direct injection into the CSF. Rubenstein et al. treated chemotherapy alone in terms of response rates and pro-
ten patients with CNS relapse of LBCL with a planned gression free survival for indolent and aggressive lym-
nine injections of rituximab at 10 mg, 25 mg, or 50 mg phomas, and overall survival for large B cell lymphoma.
doses via Ommaya reservoir over 5 weeks [613]. The Table 24 shows the results of randomized trials that have
maximum tolerated dose was determined to be 25 mg evaluated the effect of maintenance rituximab after ritux-
based on patients who received the 50 mg dose experi- imab plus chemotherapy, but many were trials in which this
encing dose-limiting hypertension. These patients also was a secondary randomization limited to patients who
had systemic symptoms including abdominal cramps or had responded to either chemotherapy alone, or chemo-
nausea and vomiting. Responses were observed in all therapy plus rituximab after the primary randomization.
338 Monoclonal antibody therapy

Table 24. Randomized trials of rituximab + chemotherapy ± “maintenance” rituximab in B cell lymphoproliferative disorders
Citation Clinical setting Induction regimen Maintenance rituximab # randomized PFS OS
[276] Elderly untreated RCHOP 4 weekly doses q 6 80 77% 2-years NSD
months × 2 years vs. observation
93 75% 2-years
LBCL p = 0.81
[745] Relapsed RCHOP 1 dose q 3 months × 2 years vs. observation 91 52 months NSD
FCL 98 23 months
p = 0.004
[234] Relapsed RFCM 4 doses at 3 months & 9 months vs. 41 36 months NSD
observation
FCL 40 26 months
p = 0.035
LBCL = large B cell lymphoma
FCL = follicular lymphoma
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
RFCM = rituximab, fludarabine, chlorambucil, mitoxantrone
PFS = progression free survival
OS = overall survival

The accompanying table excludes patients who had Table 25. Rituximab in combination with biologicals other
received only chemotherapy as their induction treatment, than antibodies
since this is not longer considered optimal therapy. Clinical Biological Number of Response
Citation setting agent patients rate
[617] Relapsed IFN-α 64 70%
Rituximab with other Biologics indolent
Rituximab with Biologicals other than Monoclonal [149] Relapsed IFN-α 38 45%
indolent
Antibodies [239] Relapsed IL-2 20 55%
To the extent that rituximab’s clinical benefits are due to indolent
CDCC and ADCC, combining rituximab with other bio- [259] Relapsed IL-2 34 26%
logicals may enhance its clinical benefit. Table 25 sum- indolent
marizes several studies that explored such combinations [380] Relapsed IL-2 54 10%
indolent
[8, 9, 148, 239, 259, 368, 380, 475, 609, 617, 743]. [475] Relapsed GM-CSF 12 83%
Thalidomide was given orally and all of the cytokines indolent
were given s.c. Ranges of response were 45–75% for [609] Relapsed GM-CSF 35 60%
interferon-α, 26–55% for IL-2, 60–83% for GM-CSF, indolent
[8] Relapsed IL-12 43 69%
37–69% for IL-12. The large variance in response rates
indolent
for specific combinations is almost certainly due to the [9] Relapsed IL-12 30 37%
specific characteristics of the patient populations in each indolent
small trial. Only randomized trials can establish the [743] Relapsed G-CSF 19 42%
superiority of these responses compared to rituximab indolent
[368] Relapsed Thalidomide 16 81%
alone, which produced response rates from 45% to 85% Mantle Cell
as a single agent in indolent lymphomas. Whether such
IFN-α = interferon alpha
trials will be conducted is questionable since rituximab IL-2 = interleukin-2
plus chemotherapy is considered standard induction GM-CSF = granulocyte macrophage colony stimulating factor
therapy for these malignancies. IL-12 = interleukin-12
G-CSF = granulocyte colony stimulating factor
Rituximab with other monoclonal antibodies
Rituximab has been combined with the anti-CD52 anti-
body alemtuzumab, and the anti-CD22 antibody epratu- et al. and Leonard et al. gave epratuzumab at 360 mg/m2
zumab as shown in Table 26. Faderl et al. gave rituximab i.v.ly over 60 min followed by rituximab at 375 mg/m2,
and alemtuzumab at their standard doses [214]. Strauss weekly for 4 weeks [425, 697].
Robert O. Dillman 339

Table 26. Rituximab in combination with other monoclonal activation of complement, B-lymphocyte cytolysis, and
antibodies TNF-alpha release [40, 787]. The severity of the reaction
Clinical Biological Number of Response depends on the rate of rituximab infusion, the number of
Citation setting agent patients rate CD20+ cells, and the immune competence of the patient.
[214] Relapsed Alemtuzumab 25/48 52% In patients who have high numbers of rapidly proliferat-
CLL ing CD20-positive cells, classic tumor lysis syndrome
[697] Relapsed Epratuzumab 21/33 64% can occur. The rate of penetration and binding of ritux-
FCL
imab to CD20-positive cells located in tumor masses is
[426] Relapsed Epratuzumab 10/15 67%
FCL much slower; therefore in the absence of large numbers
[697] Relapsed Epratuzumab 2/16 13% of circulating tumor cells, tumor lysis syndrome is only
NHL encountered when rituximab is given chemotherapy.
[697] Relapsed Epratuzumab 7/15 47% Once circulating B cells have been eliminated, ritux-
LBCL
[426] Relapsed Epratuzumab 4/6 67%
imab can be infused rapidly with many using an infusion
LBCL time of 45 min for subsequent weekly infusions [82,
CLL = chronic lymphocytic leukemia 170, 175, 655]. If rituximab therapy is being re-started
FCL = follicular lymphoma after being discontinued for several months, and B lym-
NHL = non-Hodgkin’s lymphoma phocytes are again present in the blood stream, then the
LBCL = diffuse large B cell lymphoma slow infusion rate should be used. Once circulating target
cells are no longer present, subsequent infusions may be
safely given at more rapid rates. For instance, the author
Toxicity and side Effects administers full dose rituximab over 30 min during sub-
The toxicities associated with rituximab were summa- sequent weekly treatments once B lymphocytes have
rized earlier in this chapter in Table 6. Rituximab is gen- been cleared from the circulation.
erally well tolerated. The most common toxicities Despite its excellent safety record, rituximab does
observed are the “flu-like” symptom complex associ- carry several “Black Box Warnings” from the US FDA.
ated with i.v. infusion followed by the binding of ritux- These include fatal infusion reactions, tumor lysis syn-
imab to circulating CD20 positive B cells and their drome, severe mucocutaneous reactions, and progres-
subsequent elimination in the reticuloendothelial sys- sive multifocal leukoencepalopathy. As noted above,
tem [170, 175, 184]. In the pivotal trial for rituximab, depending on the clinical setting and rate of rituximab
33% of patients had their first infusion interrupted infusion, patients may experience hypoxia and dyspnea
because of side effects, but toxicities were rarely seen that can be associated with pulmonary infiltrates and
with subsequent infusions after circulating B lympho- acute respiratory distress syndrome, cardiac arrhythmias
cytes were cleared from the circulation [474]. Because including ventricular fibrillation and cardiac shock, and
of the predictably of the initial infusion reaction, cau- myocardial infarction. It is noted that 80% of fatal infu-
tion should be taken with patients who have underlying sion reactions occurred with the first infusion. Tumor
cardiac or respiratory problems who may not tolerate lysis syndrome with renal failure and death can occur
the stress of the cytokines released, and the brief with rituximab alone in the presence of high numbers of
decrease in oxygen saturation associated with the initial rapidly proliferating CD20-positive cells in the circula-
congestion of antibody coated cells in the lungs. Care tion, or when rituximab is given with chemotherapy in
should also be taken when treating patients with high patients who have a high tumor burden of CD20+ cells
circulating B lymphocyte counts, especially if those anywhere in the body. Rare cases of severe mucocutane-
cells strongly express CD20, although most patients ous reactions including Stephens-Johnson syndrome
will tolerate rituximab therapy [79], [363]. and bullous pemphigoid have been reported. Progressive
When rituximab is administered i.v., antibody mole- multifocal leukoencephalopathy associated with the
cules immediately contact CD20+ B cells in the circulation. Creutzfeld-Jacob virus has been rarely observed.
After binding, these are removed in the reticuloendothe-
lial system including the lung, liver, and spleen. Some
Summary
may also be lysed as part of CDC. Both the binding to
CD20 and elimination by CDC and ADCC are associ- The approval and resounding success of rituximab opened
ated with the release of cytokines which produce fever, the doors to other unconjugated or “naked” Mab. In 1998,
chills, sweats, dyspnea, tachycardia, and sometimes the year in which the drug was released, it became the
hypotension. Rituximab infusion is rapidly followed by most successful cancer drug ever launched, surpassing
340 Monoclonal antibody therapy

the achievement of the chemotherapeutic paclitaxel construct, Burroughs-Wellcome decided to discontinue


(Taxol), even though the latter had proven useful in mul- trials with this agent [199]. Subsequently, Ilex Oncology
tiple solid tumors including lung, breast and ovarian obtained the product and elected to proceed with develop-
cancer. After introduction as an active therapy for relapsed ment, and manufactured the humanized derivative in CHO
patients with indolent lymphoma, rituximab quickly cells. Alemtuzumab reacts with the CD52 antigen, which
become part of the standard treatment of virtually every is present on most lymphomas and chronic leukemias, dis-
B-cell malignancy. Rituximab has proven to be an out- eases, and is expressed on many cell types including
standing agent producing high rates of durable responses malignant and normal B and T lymphocytes, natural killer
in a broad range of B cell lymphoproliferative disorders, cells, monocytes, macrophages, dendritic cells, some neu-
especially when combined with chemotherapy. Rituximab trophils [195]. CD52 is also expressed on tissues of the
is appropriate for use either alone or in combination with male reproductive system including epididymis, seminal
chemotherapy as the initial treatment of indolent lympho- vesicles, and mature sperm. CD52 is not internalized after
mas. It is appropriate to combine rituximab with chemo- antibody binding, and it does not appear to be secreted.
therapy for the treatment of newly diagnosed aggressive
lymphomas including large B cell and mantle cell. It is
appropriate to combine rituximab with a purine analog
Clinical Trials with Alemtuzumab
with or without an alkylating agent in the treatment of Early clinical trials of with anti-CD52 Mab
newly diagnosed CLL. Initial trials with anti-CD52 were conducted in lym-
Because of the phenomenal impact and success of phoma. There were no significant clinical responses among
rituximab, there are numerous other anti-CD20 antibody nine patients who received the rat IgG2b monoclonal
products being developed in an effort to improve on the antibody [195]. In contrast to these results, two of three
existing agent. Most of the rationale for such efforts patients did respond to a rat/human IgG1 chimeric con-
relate to the potential to improved on binding to the struct [284]. These two patients had lymphadenopathy,
CD20 target, or enhancement of immune function splenomegaly, and bone marrow involvement that
because of alterations in the Fc portion of anti-CD20 responded to doses of only 1–20 mg. Tang et al. reported
antibodies based on several reports suggesting that the partial responses in three out of seven patients with recur-
Fc polymorphisms may predict clinical efficacy [95, rent indolent lymphoma, who received thrice weekly
385]. However, others have failed to confirm this obser- treatment with 30 mg of the humanized antibody [708]. In
vation. [81, 216]. early trials, the rat version of the antibody was given to
five CLL patients [195, 196]. Three had sustained
decreases in circulating leukemic cells, but only one of
Alemtuzumab (Campath®, Bayer the three had resolution of marrow infiltrating by leuke-
Healthcare, Tarrytown, NY) mic cells. A sixth CLL patient received a rat/human chi-
Alemtuzumab and CD52 meric IgM CAMPATH antibody and had no response.
Burroughs-Wellcome sponsored clinical trials of the
The second Mab approved by the U.S. FDA for a hemato- human IgG1 construct and clinical responses were seen in
logic malignancy was alemtuzumab (Campath), an anti- patients who had failed fludarabine chemotherapy. One
CD52 monoclonal antibody that was approved in May report described responses in 6/16 patients who received
2001 based on data submitted for the treatment of patients 30 mg thrice weekly for 16 weeks [347]. However, the
with CLL that had recurred or been refractory to the purine company was disappointed in the associated toxicity, and
analog fludarabine. Development of this product and a elected to discontinue the trials [199]. Subsequently ILEX
humanized version actually started several years before Pharmaceuticals obtained the rights to the antibody and
rituximab. The original rat antibody Campath-1G was proceeded with additional registration trials in CLL patients
genetically modified to create Campath1-H, later called who had relapsed or been refractory to fludarabine.
alemtuzumab, by joining the hypervariable region of the
rodent antibody with a human IgG1 kappa variable frame- Alemtuzumab in CLL
work and constant regions [196]. Rat versions of this anti- Table 27 summarizes the results of various clinical trials
body showed limited clinical activity [195], but in vitro of single agent alemtuzumab in patients with CLL [128,
cytotoxicity in the presence of human complement and/or 222, 320, 372, 449, 503, 539, 577, 578]. The trials with
effector cells was greatly enhanced with the human IgG1 30 mg i.v. over 2 h thrice weekly in patients who had
construct, and also showed activity in vivo [284]. Despite relapsed after or progressed during fludarabine treatment
some encouraging clinical results with the humanized were used to support the FDA approval of alemtuzumab
Robert O. Dillman 341

Table 27. Alemtuzumab as a single agent in chronic lymphocytic leukemia


Citation Clinical setting I.V. dose and schedule Number of patients Response rate
[449] Untreated 30 mg sc thrice weekly for 18 weeks 38 87%
[320] Untreated 30 mg iv thrice weekly for 12 weeks 149 83%
[503] Relapsed post chemotherapy 30 mg iv thrice weekly for 12 weeks 91 55%
[128] Relapsed post chemotherapy 10 mg sc thrice weekly for 18 weeks 16 50%
[577] Relapsed post fludarabine 30 mg iv thrice weekly for 12 weeks 136 40%
[539] Previously treated 30 mg iv thrice weekly for 12 weeks 29 38%
[372] Relapsed post fludarabine 30 mg iv thrice weekly for 12 weeks 93 33%
[578] Relapsed post fludarabine 30 mg iv thrice weekly for 12 weeks 24 33%
[222] Median 3 prior chemotherapies 30 mg sc or iv thrice weekly for 12 weeks 115 23%

for the treatment fludarabine-resistant CLL [372]. These chlorambucil 40 mg/m2 monthly for up to 1 year as initial
trials demonstrated response rates of about 33%, and therapy, with response rates of 63% vs. 37% and PFS of
durations of response from 7 to 11 months. A high per- 20 vs. 14 months [576].
centage of patients also had stable disease. Because of Alemtuzumab has also been explored in other CD52-
severe infusion reactions associated with the reactivity positive leukemias. In a small study a response rate of
of alemtuzumab with B and T lymphocytes, granulo- 73% was observed among 15 patients with T-cell pro-
cytes, and monocytes, in these trials the dose of alemtu- lymphocytic leukemia [545]. In a larger trial a response
zumab was gradually increased from three to ten and rate of 51% and CR rate of 13% was seen in 75 patients
then to 30 mg thrice weekly. with relapsed T cell prolymphocytic leukemia who were
A more rationale schedule than escalating boluses is treated with alemtuzumab at a dose of 30 mg i.v. thrice
continuous i.v. or subcutaneous (s.c). The s.c. strategy has weekly [371]. In a small study that included nine patients
worked well with a decrease in the severity of infusion with acute myeloid leukemia and six with acute lymphoid
reactions with similar efficacy, but local skin reactions leukemia, a 20% response rate was observed, but 87%
occur in almost all patients and can be severe. Based on of patients experienced an infection [722].
an analysis of serum samples from 30 patients who were Because of the efficacy of purine-analog based chemo-
treated with i.v. alemtuzumab, and 20 patients treated by therapy regimens with or without rituximab, many recent
the sc route, blood concentrations were similar and cumu- alemtuzumab trials have focused on its use to eradicate
lative doses slightly higher using the sc route, but the sc minimal residual disease in responding CLL patients.
route appeared to be associated with appearance of anti- Thrice weekly doses of 10 to 30 mg have been adminis-
alemtuzumab antibodies in some patients [285]. Other tered i.v. or s.c. for 4 to 12 weeks as a consolidative therapy
investigators have prolonged treatment from 12 weeks to [499, 530, 780]. These studies have shown that consolida-
18 to 30 weeks with some increase in response rates and tion with alemtuzumab does increase clinical, phenotypic,
CR rates with low rates of opportunistic infections in and molecular complete response rates, but there is an
patients who had received extensive prior chemotherapy increased risk of opportunistic infections, even if anti-viral,
[128, 449, 714]. In a study of 36 CLL patients who were anti-bacterial, and anti-Pneumocystis prophylaxis is used.
treated with the standard i.v. dose and schedule, there
was no correlation between response to alemtuzumab and Alemtuzumab in lymphoma
the high-affinity, FcgammaR receptor polymorphisms Alemtuzumab has activity in both B and T cell lymphop-
FcgammaR3A and FcgammaR2A [437]. roliferative disorders including lymphomas. As shown in
In a European registration trial 297 previously Table 28, response rates were highest in patients in the
untreated CLL patients were randomized to oral mycosis fungoides phase of cutaneous T cell lymphoma
chlorambucil 40 mg/m2 monthly vs. standard i.v. alem- (CTCL), especially in patients who were previously
tuzumab [320]. The Mab produced a superior response untreated. Infusional complications from cytokine
rate, 83% vs. 55% (p < 0.0001) and PFS which has release were common, especially with the first infusion
resulted in a marketing indication as initial therapy, even [37, 377, 383, 448, 450, 736, 814]. Cytopenias were
though in the U.S. purine-analog based therapy has been common and associated with high rates of infection,
considered the treatment of choice ever since random- including some deaths, which resulted in early termination
ized trials showed that fludarabine was superior to oral of several trials.
342 Monoclonal antibody therapy

Table 28. Alemtuzumab as a single agent in lymphoma and other lymphoproliferative disorders other than B cell chronic lym-
phocytic leukemia
Citation Clinical setting I.V. dose and schedule Proportion responding Response rate Percent infections
[736] 16 indolent, 2 aggressive NHL 30 mg iv thrice weekly 8/18 44% 67%
[383] Relapsed indolent NHL 30 mg iv thrice weekly 3/18 17% 72%
[448] Relapsed Indolent NHL 30 mg iv over thrice weekly 6/50 14% 32%
[209] PTCL 30 mg iv thrice weekly 5/14 36% 57%
[814] CTCL & PTCL 10 mg iv thrice weekly 6/10 60% 10%
[37] CTCL Mycosis fungoides 10 mg sc thrice weekly 12/14 87% 29%
[450] CTCL Mycosis fungoides 30 mg iv thrice weekly 12/22 55% 50%
[377] CTCL Mycosis fungoides 30 mg iv thrice weekly 3/8 38% >50%
NHL = non-Hodgkin’s lymphoma
CTCL = cutaneous T cell lymphoma
MF = mycosisi fungoides
PTCL = peripheral T cell lymphoma

Alemtuzumab with Chemotherapy in CLL warnings for Alemtuzumab include severe pancytope-
nias after higher cumulative doses, infusion reactions,
Alemtuzumab has been combined with fludarabine in and infections, including opportunistic infections.
the treatment of CLL. Kennedy et al. reported obtaining Because it reacts with both B and T cells in the circu-
complete response in six patients treated with both lation, infusion reactions associated with administration
agents concurrently, who were felt to be refractory to of alemtuzumab are common and often severe. The high
each drug as single agents [377]. Elter et al. reported an percentage of significant infusion reactions limits the
83% response rate among 36 patients who had previ- ability to deliver high doses i.v. initially, and in trials
ously received a mean of 2.6 chemotherapy regimens about 5% of patients had to discontinue treatment
[207]. Two patients developed fungal pneumonias, one because of infusion-associated reactions. For this reason,
patient died of Escherichia coli sepsis, and two patients in the registration trials doses were initially escalated
had reactivation of subclinical cytomegalovirus. from 3 to 10 mg to 30 mg during the first week, and then
administered as 2-h infusions of 30 mg thrice weekly for
Alemtuzumab with other Biologicals in CLL an additional 11 weeks. Because of the systemic toxicity,
approximately one log lower mg doses of alemtuzumab
Alemtuzumab has been combined with rituximab in the are used compared to rituximab. These low doses are
treatment of patients with CLL. In one trial of 41 CLL unable to sustain serum levels and promote good pene-
patients the response rate for the combination was more tration into large tumor masses, which is why the best
than 50% and median PFS was 6 months, but infections results with this schedule of therapy have been achieved
occurred in more than 50% of patients [214]. In a sec- in the blood and bone marrow rather than large lymph
ond combination trial, patients were divided into three nodes. This problem is being overcome by giving s.c.
cohorts that received alemtuzumab at doses of 3, 10, or doses and continuing treatment for longer duration, but
30 mg during therapy, 1/12 patients had a response this also prolongs immunosuppression.
[517]. G-CSF was combined with alemtuzumab in the A major limitation for clinical use of alemtuzumab is
treatment of 14 CLL patients who had been heavily pre- the immunosuppression that accompanies prolonged
treated (median 3.5 chemotherapy regimens, in an effort T-lymphopenia and neutropenia, which is associated
to reduce infections complications and perhaps increase with an increased risk of bacterial infections, and oppor-
efficacy [436] G-CSF was administered at a dose of tunistic infections including Herpes virus, cytomegalo-
5 μg/kg daily 5 days before and throughout standard i.v. virus infections, fungal infections, Pneumocystis carinii,
alemtuzumab therapy. Five patients responded, but nine and mycobacterial infections. The risk of opportunistic
patients developed neutropenia and six patients had infections can be reduced, but not eliminated, by admin-
reactivation of cytomegalovirus. istering prophylactic antibiotics and antiviral agents.

Toxicity and Side Effects Summary


The toxicities associated with Alemtuzumab were sum- Alemtuzumab is active in the treatment of both B and T
marized earlier in this chapter in Table 6. Black box cell malignancies, and it widely used as a salvage and
Robert O. Dillman 343

consolidation agent in CLL. It might have had much Indirect support for this comes from a clinical trial in
wider use against B cell lymphoid malignancies had which breast cancer patients, whose tumor cells had
rituximab not been approved and widely adopted before high expression of Her2 by immunohistochemistry
alemtuzumab became available. Thus, even though (IHC), were treated with docetaxel and trastuzumab;
alemtuzumab has a marketing indication in CLL, and tumor samples contained significantly increased num-
rituximab does not, the latter is more widely used in bers of natural killer cells and increased expression of
CLL. Enthusiasm for alemtuzumab has also been muted Granzyme B and TiA1 in lymphocytes compared with
because of its toxicity, especially immunosuppression, controls [11]. Based on data submitted to the US FDA,
which has greatly limited efforts to combine the agent the recommended initial loading dose is 4 mg/kg admin-
with chemotherapy. istered as a 90-min infusion followed by a weekly main-
tenance dose of 2 mg/kg administered as a 30-min
infusion. A 3 week dosing schedule has also been vali-
Trastuzumab (Herceptin®, Genentech, dated [25, 431].
South San Francisco, CA) Her2 is overexpressed in only about 25% of patients
with breast cancer; so, testing for high expression of
Trastuzumab and Her2
the antigen (receptor) by immunohistochemistry
In September 1998 the U.S. FDA approved trastuzumab (IHC), or for overexpression of the Her2-neu gene by
(Herceptin), a humanized Mab that reacts with the sec- fluorescence in situ hybridization (FISH), is critical
ond component of human epidermal growth factor for appropriate patient selection. Large trials have
receptor (EGFR), known as Her2, for the treatment of confirmed that there is poor reproducibility in perfor-
metastatic breast cancer, making it the first Mab mance and interpretation of IHC assays for HER2 in
approved for the treatment of a solid tumor [171]. laboratories that do not process and analyze large
Trastuzumab consists of the idiotopes from the hyper- numbers of samples. Even in the best of hands, there
variable region of murine antibody 4D5 united with a is not complete concordance between IHC and FISH.
human IgG1 kappa antibody that reacts with the Most centers who rely on IHC perform FISH for
p185HER2/neu receptor [93]. The Mab was humanized patients who are 2+ by IHC because many trials have
by inserting the complementary-determining regions demonstrated lower response rates in IHC 2+ com-
(CDRs) of the murine 4D5 into the constant and vari- pared to 3+, while benefit is almost always seen in
able framework of a consensus human IgG1 to produce patients who are FISH positive. The Dako Herceptest
rHuMAb 4D5, which was more active in ADCC assays for IHC assay of HER2 was used to determine eligi-
than its mouse counterpart. Trastuzumab is produced bility in the pivotal trial and is considered the most
using recombinant DNA technology with CHO cells reliable IHC test available. However, many questions
serving as the manufacturing factory. have been raised regarding the reliability and repro-
Her2 is a 185 kD transmembrane receptor that is a ducibility of this test, especially on paraffin-fixed
member of the EGFR tyrosine kinase family of recep- rather than fresh tissues [191, 544, 550, 733]. Many
tors and is involved with the autophosphorylation of samples were felt to be falsely positive for Her2 based
specific tyrosine residues after being activated by bind- on negative FISH tests [733]. Comparative studies
ing of the EGF ligand to the receptor. Early analysis confirmed wide variation in interobserver reproduc-
suggested that a subset of about 25% of breast cancer ibility, and confirmed that the two extremes, 0 and 3+
patients had tumors that overexpressed this receptor were more reproducible than 1+ and 2+, and there was
[673]. Overexpression of the erbB-2 proto-oncogene poor discrimination between 2+ and 3+ which resulted
results in overexpression of the HER-2 receptor on the in a high rate of false positive tests [544].
cell surface and increased cell proliferation [632]. Dowsett et al. compared results for 426 breast carci-
Binding of trastuzumab to the HER2 receptor results in nomas from 37 different hospitals for patients that were
internalization (modulation, down regulation) of the being considered for trastuzumab therapy by perform-
receptor and competitive inhibition to binding of EGF ing IHC and FISH in three reference centers [191]. Only
ligands to the receptor [675]. Such inhibition interferes 2/270 (0.7%) of IHC 0/1+ tumors were FISH positive
with phosphorylation and the subsequent signal trans- and only 6/102 (5.9%) of IHC 3+ tumors were FISH
duction that facilitates cell proliferation. The human negative, which suggested that FISH testing was most
IgG1 constant regions on the Fc of the humanized anti- valuable for validating that IHC 2+ tumors were Her2
body mediate ADCC in vitro; so trastuzumab may also negative. In a large U.S. adjuvant therapy trial, reference
produce anti-tumor effects via the immune system. laboratories confirmed Her2 positivity for 86% of 2,535
344 Monoclonal antibody therapy

registered patients with only 82% concordance between produces objective response rates of 12–33% depending
local and high-volume central laboratories for IHC, and on the specific patient population [23, 25, 68, 118, 755].
88% for FISH when the same methodologies were used In a phase II trial in 46 patients with metastatic breast
[550]. For the discordant cases, the central and refer- cancer in whom at least 25% of tumor cells expressed
ence laboratories had 94% agreement for IHC 0, 1+, and Her2 by IHC, a schedule of 250 mg i.v. followed by
2+ with 95% agreement that these were FISH negative. weekly doses of 100 mg for 9 additional weeks yielded
Compared to the central laboratories, rates of discor- five responses among 43 evaluable patients [23]. Low
dance were 24% for 75 laboratories that interpreted less grade fever was seen in 15% and was believed to be
than 100 HER2 samples per month and only 3% for 29 related to immune complexes formed with shed antigen.
laboratories that interpreted 100 or more HER2 samples The largest trial of trastuzumab as a single-agent,
per month [542]. which was one of the two pivotal trials that led to regu-
latory approval of the agent, was conducted in 222
patients with Her2-positive metastatic breast cancer that
Clinical Trials with Trastuzumab in Breast had recurred after chemotherapy [118]. Trastuzumab
Cancer alone was given at an initial dose of 4 mg/kg followed
Various clinical trials have confirmed the efficacy of by weekly doses of 2 mg/kg. There were eight complete
trastuzumab, especially in combination with chemo- and 26 partial responses with a median duration of
therapy, for the treatment of Her-2 positive breast cancer response of 13 months. In the 222 patients who had
in patients with recurrent metastatic breast cancer, and failed chemotherapy, there were four complete and 21
as the initial treatment for metastatic breast cancer, and partial responses for an overall objective response rate
as adjuvant therapy [331]. At this time most physicians of 12% by intent to treat analysis. The median duration
continue trastuzumab maintenance indefinitely or until of response among the 25 responders was 9 months, and
disease progression since Her2 receptor is continually their median survival was 13 months, which are compa-
being produced by some malignant cells. rable to chemotherapy. The patient population included
66% who had received prior adjuvant chemotherapy;
Trastuzmab Alone 68% had received two or more chemotherapy regimens
Clinical results from various trials with trastuzumab as a for metastatic cancer and 55 patients (25%) had relapsed
single agent in the treatment of metastatic breast cancer after high-dose chemotherapy and autologous stem cell
are summarized in Table 29. In patients with Her2-positive rescue prior to receiving antibody therapy. The response
metastatic breast cancer that has relapsed after chemother- rate was 14% among patients whose tumors were Her2
apy, eHer2available data suggests that trastuzumab alone 2+ or 3+ by IHC, but 20% for patients whose tumors
were FISH+ vs. 0% for FISH−. Treatment was gener-
ally well tolerated with 40% of patients experiencing
Table 29. Clinical trials of trastuzumab alone in Her2-positive
metastatic breast cancer fever during the first infusion. Cardiac dysfunction was
noted in 5% which was usually reversible when treat-
Clinical I.V. dose and Number of Response ment was discontinued.
Citation setting schedule patients rate
Burris et al. gave trastuzumab at twice the standard
[68] Initial 8 mg/kg, then 52 33% doses by the weekly schedule as initial treatment for 8
therapy 4 mg/kg
weekly for 8 weeks for patients with Her2-positive metastatic breast
weeks cancer who had never received chemotherapy [68]. They
[755] Initial 4 or 8 mg/kg, 114 26% reported a response rate of 33% among 52 evaluable
therapy then 2 or patients in a trial that enrolled 61 patients. This response
4 mg/kg rate is the highest reported for single-agent trastuzumab.
weekly
[26] Initial 8 mg/kg, then 105 19% However, in a randomized phase II trial conducted by
therapy 6 mg/kg q 3 Vogel et al, there was no suggestion that this higher dose
weeks produced a higher response rate [755]. In this trial there
[118] Relapsed 4 mg/kg then 222 15% was no significant difference in results for an 8 mg/kg
2 mg/kg
loading dose and 4 mg/kg per week maintenance vs. the
weekly
[26] Relapsed 250 mg, then 43 12% standard 4 mg/kg loading dose and 2 mg/kg per week
100 mg maintenance. The failure to demonstrate any difference
weekly × 10 between these doses is not surprising in view of the sus-
weeks tained serum levels of trastuzumab that were achieved at
Robert O. Dillman 345

the lower dose. In the Vogel trial, trastuzumab alone as these trials were the taxanes (paclitaxel or docetaxel), or
initial treatment for metastatic breast cancer resulted in gemcitabine. The various agents were associated with
seven complete and 23 partial responses, but the response response rates between 25% and 75% with median PFS
rates were 35% for patients whose tumors were IHC 3+ of 6 to 12 months. Response rates of 50% or greater were
compared to 0% for tumors that were IHC 2+, and 34% reported in 17 of the 20 trials which appeared to be much
among patients whose tumors were FISH+ compared to higher than observed with chemotherapy alone. Many of
7% for tumors that were FISH−. The most common these reports noted higher response rates in the subsets
adverse events attributed to trastuzumab were chills of patients who had 3+ IHC Her2 expression by IHC and/
(25%), asthenia (23%), fever (22%), pain (18%), and or overexpression by FISH, as opposed to 2+ IHC Her2
nausea (14%) and two patients (2%) with histories of car- or negative FISH. The addition of trastuzumab was not
diac disease experienced reversible cardiac dysfunction. associated with any additive toxicities, other than cardiac
It would be more convenient for patients if trastu- disease in association with anthracyclines.
zumab were given at higher doses and less frequently
than weekly. Baselga et al. tested an every 3-week Combination Chemotherapy and Trastusumab
schedule of trastuzumab delivery as initial therapy for Table 31 summarizes published phase II clinical trials
105 patients with Her2-positive metastatic breast cancer that used combination chemotherapy plus trastuzumab
[25]. They observed response rate was 19%, but was for treatment of patients with Her2-positive breast cancer
34% for patients whose tumors were IHC 3+ and/or [235, 483, 500, 547, 549, 604, 690, 750]. Most of these
FISH+ for Her2. As with other trastuzumab schedule, trials utilized chemotherapy combinations of taxanes
the most common treatment-related adverse events were and platinum agents. Combination chemotherapy has
rigors, fever, headache, nausea, and fatigue. consistently produced higher response rates and longer
progression free survival in clinical trials in patients
Trastuzumab Plus Single-Agent Chemotherapy with metastatic breast cancer, although survival advan-
In vitro and animal studies showed that trastuzumab aug- tages have been harder to establish. As can be seen in
mented the anti-tumor cytotoxicity of various chemother- the accompanying table, phase II trials of combination
apy agents [24], [547]. Table 30 summarizes published chemotherapy plus trastuzumab appear to produce
phase II clinical trials, grouped by the single-agent chemo- slightly higher response rates (40–92%) than were seen
therapy that was combined with trastuzumab for treatment with single agents plus trastuzumab, and the PFS was
of patients with Her2-positive breast cancer [11, 22, 70, 71, somewhat longer (range 6–16 months).
103, 112, 155, 212, 235, 267, 344, 431, 498, 537, 543, In a randomized trial, trastuzumab and paclitaxel with or
546, 604, 630, 657, 710]. Most of the single agents used in without carboplatin were compared as first-line therapy

Table 30. Phase II clinical trials of trastuzumab plus concurrent single-agent chemotherapy in metastatic breast cancer
Citation Clinical setting Chemotherapy Number of patients Response rate
[546] Multiple prior treatments Cisplatin 37 24%
[498] 92% prior chemo Docetaxel q 3 weeks 25 70%
[212] 1st or 2nd chemo Docetaxel weekly 30 63%
[11] Neoadjuvant Docetaxel 23 61%
[710] 1st or 2nd chemo Docetaxel weekly 26 50%
[630] Measurable metastatic Docetaxel q 3 weeks 40 65%
[537] 95% prior taxane & anthracycline Gemcitabine 61 38%
[112] Measurable metastatic Liposomal Doxorubicin 29 52%
[235] No prior therapy Paclitaxel weekly 33 62%
[657] 0–3 prior chemo Paclitaxel weekly 88 61%
[431] No prior chemo Paclitaxel q 3 weeks 32 59%
[267] Prior taxane and anthracycline Paclitaxel weekly 25 56%
[604] No prior chemo Paclitaxel q 3 weeks 88 36%
[344] No prior chemo Vinorelbine 37 78%
[69] 82% prior chemo Vinorelbine 40 75%
[71] Initial chemo Vinorelbine 54 68%
[155] 0 or 1 prior chemo for mets Vinorelbine 40 50%
[22] No prior chemo Vinorelbine oral 30 68%
[103] No prior chemo Vinorelbine 62 63%
[543] Prior chemo Vinorelbine 35 51%
346 Monoclonal antibody therapy

for 196 women with HER-2-overexpressing (2+ or 3+ addition of carboplatin based on response rates of 52%
by IHC) metastatic breast cancer [604]. Treatment con- vs. 36% (p = 0.04), and median PFS of 11 vs. 7 months
sisted of six cycles of trastuzumab 4 mg/kg loading dose (hazard ratio 0.66, p = 0.03). In consecutive phase II tri-
and 2 mg/kg weekly thereafter with paclitaxel 175 mg/ als, weekly paclitaxel and carboplatin plus trastuzumab
m2 every 3 weeks, with or without the addition of carbo- appeared superior to an every 3-week schedule of the
platin (AUC = 6) every 3 weeks. Endpoints favored the same agents with response rates of 81% vs. 65%, median
PFS 14 vs. 10 months, and median OS 3.2 vs. 2.3 years,
and was associated with less hematologic toxicity
Table 31. Combination chemotherapy and trastuzumab for [549].
metastatic breast cancer Similar to what has been seen in other trials with
Clinical Number of Response
anthracyclines, the combination of epirubicin plus car-
Citation setting Chemotherapy patients rate boplatin was associated with a high rate of significant
cardiotoxicity (10/45) [750]. The majority of cardiac
[547] 15% prior Docetaxel + 59 58%
adjuvant Carboplatin events occurred late during therapy with trastuzumab
taxane alone. Half of the patients were asymptomatic and all
[547] No prior Docetaxel + 62 79% cases of CHF were resolved using cardiac therapy.
taxane Cisplatin
[750] Initial Docetaxel + 45 67%
therapy Epirubicin
Randomized Trials of Chemotherapy
[549] Initial Paclitaxel + 48 81% With or Without Trastuzumab in Breast Cancer
therapy Carboplatin Randomized trials comparing chemotherapy plus trastu-
weekly zumab to chemotherapy alone are shown in Table 32.
[549] Initial Paclitaxel + 43 65% One of the pivotal trials that led to regulatory approval of
therapy Carboplatin
q 3 weeks trastuzumab compared trastuzumab plus chemotherapy
[604] No prior Paclitaxel + 88 52% to chemotherapy alone in 469 patients with metastatic
chemo- Carboplatin disease who had not received prior chemotherapy for
therapy q 3 weeks metastatic disease [674]. All patients had tumors that
[236] No prior Paclitaxel + 40 52%
overexpressed Her2, which was defined as 2+ or 3+ by
chemo- Gemcitabine
therapy IHC on a scale of 0–3. An initial 4 mg/kg trastuzumab
[483] No prior Paclitaxel + 13 92% dose was followed by 2 mg/kg weekly. The first dose of
chemo Gemcitabine Mab was infused i.v. over 90 min, but in the absence of
[690] 60% Cisplatin + 20 40% significant infusion related toxicity, subsequent doses
taxane, Gemcitabine
90%
were infused i.v. over 30 min. Patients who had not
anthra- received an anthracycline previously were randomized
cylcine to receive trastuzumab alone or with cyclophosphamide
[500] 2nd line Gemcitabine + 30 50% 600 mg/m2 i.v. and doxorubicin 60 mg/m2 or epirubicin
therapy Vinorelbine (AC) i.v. every 3 weeks for six cycles. Patients who had

Table 32. Randomized trials of chemotherapy ± trastuzumab in breast cancer


Citation Clinical setting Chemotherapy Proportion responding Response rate Median PFS
[674] Initial therapy metastatic breast, prior dox Paclitaxel + T 38/92 41% 7 months
Paclitaxel 16/96 17% 3 months
p < 0.001 p < 0.001 p < 0.0001
[674] Initial therapy metastatic breast, no prior dox AC + T 80/143 56% 8 months
AC 58/138 42% 6 months
p = 0.02 p = 0.10 p = 0.002
[470] Initial chemo Docetaxel + T 57/93 61% 12 months
32/93 34% 6 months
Docetaxel p = 0.002 p = 0.0001
T = trastuzumab
AC = doxorubicin or epirubicin plus cyclophosphamide
PFS = progression free survival
Robert O. Dillman 347

received adjuvant chemotherapy that included an anthra- trastuzumab arm was associated with more grade 3 and
cycline were randomized to receive paclitaxel 175 mg/m2 4 neutropenia (32% vs. 22%), more febrile neutropenia
i.v. over 3 h alone every 3 weeks, or with trastuzumab. (23% vs. 17%), and one patient (1%) in the Mab-
The combination of chemotherapy plus trastuzumab was containing arm had symptomatic heart failure.
superior for key end points including: response rate (50%
vs. 32%, p < 0.0001), duration of response (9.1 vs. 6.1 Adjuvant Treatment of Breast Cancer
months), progression-free survival (median 7.4 vs. 4.6 Based on the positive benefits of trastuzumab in mea-
months, p < 0.001), and overall survival (death at 1 year, surable metastatic breast cancer, it was widely assumed
22% vs. 33%, p = 0.008), and median survival 25.1 vs. that a similar benefit would be conveyed in the adjuvant
20.3 months, p = 0.046) with a 20% risk reduction of setting against microscopic metastatic disease. Three large
death. As shown in Table 32, the differences were most randomized trials have confirmed the expected advan-
striking for paclitaxel plus trastuzumab vs. paclitaxel tage of this approach as shown in Table 33. Because of
alone. The median OS was 22 months for paclitaxel plus the cardiotoxicity associated with trastuzumab, patients
trastuzumab vs. 18 months for paclitaxel alone (p = 0.17) with active cardiac disease were excluded from these
and 27 months for AC plus trastuzumab vs. 21 months trials. Based on Her2 biology, it is probable that there is
for AC alone (p = 0.16). more of an advantage to give trastuzumab during, rather
This trial clearly demonstrated that cardiotoxicity is than only after chemotherapy, but this has not yet been
associated with trastuzumab. Cardiac dysfunction was established in trials. It is also probable that there is an
noted in 11/91 (12%) patients who received paclitaxel advantage for giving trastuzumab indefinitely, even if it
plus trastuzumab alone compared to only 1/95 (1%) has already been given with chemotherapy, but this also
who received paclitaxel alone, and in 28/143 (20%) who has not been proven. Finally, completed trials have
received AC plus trastuzumab compared to 7/135 (3%) attempted to address whether it is better to give adjuvant
who received AC alone. Even though trastuzumab does trastuzumab for 2 years or 1 year, but it is probable that
enhance the efficacy of anthracycline containing regi- it is better to give trastuzumab indefinitely if there is
mens, the increased cardiac toxicity that is seen with reason to believe that the breast cancer has not been
such combinations has made this an inappropriate treat- completely eliminated.
ment strategy. Three large randomized trials have been reported, at
Another randomized trial compared docetaxel plus least in preliminary form [353, 560, 606, 677]. Two U.S.
trastuzumab to docetaxel alone using in patients with cooperative group trials were combined for one prelimi-
metastatic breast cancer who had received no prior che- nary analysis [606]. The National Surgical Adjuvant
motherapy. Docetaxel was given every 3 weeks and Breast and Bowel Project (NSABP) trial B-31 compared
trastuzumab was given weekly. This trial confirmed a doxorubicin and cyclophosphamide followed by pacli-
similar relative advantage for the addition of trastu- taxel every 3 weeks with the same regimen plus 52
zumab with roughly a doubling of response rate and weeks of trastuzumab beginning with the first dose of
PFS, and better OS 31 vs. 23 months (p = 0.032). The paclitaxel. The North Central Cancer Treatment Group

Table 33. Randomized trials of trastuzumab in the adjuvant treatment of high-risk breast cancer
Citation Clinical setting Treatment arms Number of patients DFS OS
[606] Node + or high risk node Trastuzumab no trastuzumab 1,672 87% 3-years 94% 3-years
1,679 75% 3-year 92%-3 years
0.48 HR p = 0.015
p < 0.0001
[560] Node + or high risk node Trastuzumab observation 1,703 86% 2-years
1,698 78% 2-years
[677] 0.54 HR 0.66 HR
p < 0.0001 p < 0.0001
[353] Node + or high risk node Trastuzumab observation 116 91% 3-years
116 86% 3-years
0.58 HR p = 0.15
p = 0.005
DFS = disease free survival
OS = overall survival
HR = hazard ratio
348 Monoclonal antibody therapy

(NCCTG) trial N9831 compared three regimens: doxo- Neo-Adjuvant Treatment of Breast Cancer
rubicin and cyclophosphamide followed by weekly As summarized in Table 34 trastuzumab is also being
paclitaxel, the same regimen plus 52 weeks of trastu- combined with chemotherapy as part of neo-adjuvant
zumab initiated concomitantly with paclitaxel, and the therapies with pathologic complete response rates rang-
same chemotherapy regimen followed by 52 weeks of ing from 13% to 65% [70, 77, 78, 130, 334, 495]. After 3
trastuzumab after paclitaxel. For an early analysis at a weeks of neoadjuvant treatment with trastuzumab alone,
median follow up of 2 years, the two similar arms from 23% of patients had a PR before going on to receive 12
each trial were combined and compared: doxorubicin more weeks of trastuzumab combined with paclitaxel
plus cyclophosphamide followed by paclitaxel with or before surgery [495]. One randomized trial, which was
without trastuzumab concurrent with paclitaxel [606]. designed to accrue 164 patients was closed early because
The international, multicenter trial (HERA) random- of the marked superiority of the trastuzumab-containing
ized more than 5,000 women with HER2-positive node arm [77]. Chemotherapy consisted of four cycles of pacli-
positive or high-risk node negative breast cancer, com- taxel followed by four cycles of 5FU, epirubicin, and
pared 1 or 2 years of trastuzumab given every 3 weeks cyclophosphamide, with weekly trastuzumab for all 24
with observation after completion of locoregional ther- weeks. After 3 years of follow up, there have been no
apy and at least four cycles of neoadjuvant or adjuvant recurrences in the patients who received neoadjuvant
chemotherapy [560, 677]. Initial reports have compared therapy, and they have a better PFS (p = 0.041) [78].
the observation and 1-year trastuzumab treatment arms
in terms of interim disease free survival [560], and over- Trastuzumab and Hormonal Therapy for Breast
all survival analyses [677]. In the European trial, 1,010 Cancer
women with node-positive or high risk node-negative About half of Her2-positive patients are hormone recep-
breast cancer were randomized to adjuvant therapy that tor positive which has led to interest in the interaction
consisted of three cycles of either docetaxel or vinorel- “cross-talk” between Her2 and hormone receptors which
bine, followed by three cycles of fluorouracil, epirubicin, appears to increase resistance to hormonal therapies.
and cyclophosphamide in both groups, with a secondary There is a recent report of a 26% response rate for 31
post-chemotherapy randomization for the 232 patients evaluable patients who were treated with the aromatase
whose tumors were Her2-positive to either observation inhibitor letrozole in combination with weekly trastu-
or trastuzumab for 9 weeks [353]. Because of the use of zumab [464]. Her2 positivity was confirmed for 82%
anthracyclines in these trial designs, patients have been (IHC3+ and/or FISH+) and 82% had previously been
closely monitored for cardiac toxicity, which has been treated with tamoxifen. The median PFS was 6 months.
higher in the trastuzumab arms. In the NSABP B-31 trial
16% of women discontinued trastuzumab therapy due to Trastuzumab in Other Tumor Types
clinical evidence of myocardial dysfunction or signifi- Her2 is over expressed at a low frequency in many other
cant decline in left ventricular function. As summarized adenocarcinoma,; so trastuzumab is also being tested
in Table 33, all three of these trials have shown a benefit in other tumors in the subsets of patients whose tumors
for the addition of trastuzumab. overexpress Her2 [2, 171]. However, because of the

Table 34. Trastuzumab plus chemotherapy as neoadjuvant therapy for locally advanced HER2-positive breast cancer
Citation Clinical setting Neoadjuvant therapy Number of patients Response rate Pathology CR rate
[495] Locally advanced Trastuzumab × 3 weeks 35 23% –
[77, 78] Locally advanced Chemo alone 19 – 26%
Trastuzumab + Chemo × 24 weeks 23 – 65%
p = 0.016
[130] Stage II or III Trastuzumab + Docetaxel/Carboplatin × 77 95% 35%
18 weeks
[70] Stage II or III Trastuxumab + Paclitaxel q 3 weeks × 40 75% 18%
12 weeks
[334] Locally advanced Trastuzumab + Docetaxel + Cisplatin × 30 – 13%
12 wks
CR = complete response
Robert O. Dillman 349

variability in IHC methodology for detecting Her2, many Table 35. Trastuzumab plus chemotherapy in tumor types
of the higher estimates may be fallacious. So far there other than breast
are no tumor types other than breast cancer for which Screened
trastuzumab appears to be efficacious, even in trials patients who
restricted to patients with whose tumors are Her2- Clinical had Her2 + Patients Response
Citation setting tumors treated rate
positive by IHC (2+ or 3+). In ovarian cancer 95/837
(11%) were HER2-positive and only 3/45 (7%) responded [244] Lung non 103/619 51 36%
small (17%)
[48]. In non-small cell lung cancer (NSCLC) 24/209
cell IIIB
(11%) patients were Her2-positive tumors by IHC and & IV
only 1/24 (4%) had a response [116]. In another NSCLC [812] Lung non 77/360 21 38%
trial 13/69 had Her2-positive tumors by IHC, and 0/13 small (21%)
responded to trastuzumab [415]. In hormone-refractory cell IIIB
& IV
prostate cancer 0/18 Her2-positive patients had a [409] Lung non 28/179 64 28%
response [810]. small (16%)
As summarized in Table 35, several exploratory trials cell IIIB
have been conducted in which trastuzumab was com- & IV
bined with chemotherapy in Her2-positive patients with [337] Metastatic 57/109 44 70%
urothe- (49%)
tumor types other than breast cancer [244, 337, 409, 620, lial
621, 812]. Very few of the patients enrolled had tumors [620] Pancreas 16% 34 38%
that were Her2 3+ by IHC. For example, less than 5% of [621] With RT – 19 50% 2-years
patients in two of the NSCLC trials had tumors that were primary OS
Her2 3+ by IHC [244, 812]. Response rates did not esopha-
gus
appear to be higher than anticipated for chemotherapy
alone, but to determine the true contribution of trastu- OS = overall survival
zumab requires randomized trials. Gatzemeier et al. ran-
domized 101 patients with Her2-positive non-small cell
lung cancer to trastuzumab plus gemcitabine and cispla-
tin or the chemotherapy alone, and found no difference
Toxicity and Side Effects
in response rate (36% vs. 41%) or PFS (6.1 vs. 7.0 The toxicities associated with trastuzumab in pivotal
months), but only six patients had tumors that were 3+ trials were summarized earlier in this chapter in Table
Her2 by IHC, and five of them did have an objective 6. Black box warnings accompanying the label indica-
response to treatment [244]. Collectively these studies tion include cardiomyopathy and infusion reactions.
have been disappointing. This does not appear to be a Trastuzumab infusions can result in serious infusion
fruitful area for further investigation given that so few reactions including dyspnea and hypoxia, which rarely
non-breast cancers strongly over express Her2. have been fatal. Most of the time transfusion reactions
are mild, and are probably associated with binding to
Trastuzumab with other Biologicals circulating white blood cells via either cross reactive
Even though the efficacy of trastuzumab might be antigens or Fc receptors, but much milder than seen
enhanced by combining it with other biological response with rituximab. Typically, symptoms occurred during
modifiers, there has been limited exploration of this or within 24 h of trastuzumab infusion. Infusion should
approach because there are so many efficacious chemo- be interrupted for patients experiencing dyspnea or
therapy agents with which to combine trastuzumab. Two clinically significant hypotension, and patients should
trials have combined s.c. IL-2 with trastuzumab [225, be monitored until signs and symptoms completely
589]. In one trial there was a response rate of 9% in 45 resolve. The basis for the minor infusion reactions that
patients, but more than 10% of patients experienced are sometimes seen with initial infusions is not clear,
grade 3 or 4 pulmonary reactions [225]. In the other but may relate to cross reactivity with an antigen expressed
trial, one of ten patients had a clinical response [589]. In on some circulating cells, or a reaction related to binding
both trials the use of IL-2 was associated with an to Fc on some circulating cells.
increase in natural killer cells, but this did not correlate The major toxicity associated with trastuzumab is
with toxicity or clinical benefit. cardiac dysfunction or congestive heart failure, that is
350 Monoclonal antibody therapy

generally mild and reversible [374, 548, 658, 678, 712, Summary
730]. Trastuzumab treatment can result in left ventricu-
lar dysfunction and congestive heart failure; therefore The approval of trastuzumab in September 1998 marked
left ventricular function should be evaluated in all the first approval by the US FDA of a therapeutic mono-
patients prior to and during treatment. The frequency clonal antibody for the treatment of a solid tumor.
and severity of left ventricular cardiac dysfunction and/ Because trastuzumab has limited clinical benefit as a
or clinical congestive heart failure is highest in patients single agent, even in patients who overexpress HER2, it
who receive trastuzuamb concurrently with anthracy- is typically administered with chemotherapy agents that
cline-containing chemotherapy regimens, and is also have proven benefit in breast cancer, other than anthra-
higher in patients who have recently received anthra- cyclines, which should be avoided because of the increased
cyline-containing regimens. If left ventricular dysfunc- risk of cardiotoxicity. A decade after its introduction,
tion or clinical heart failure occurs, trastuzumab should trastuzumab with chemotherapy is now standard ther-
be discontinued until cardiac function has improved. apy in the neoadjuvant, adjuvant, and metastatic disease
Trastuzumab can often be resumed without recurrence settings for breast cancer patients whose tumors are
of left ventricular dysfunction. Her2-positive. It is controversial as to whether trastu-
As a single agent, trastuzumab alone was associated zumab should be combined with chemotherapy in
with a 4–5% frequency of cardiac dysfunction in patients patients who have previously progressed during treat-
with metastatic disease and in the adjuvant setting [118]. ment with trastuzumab with or without chemotherapy.
The risk of cardiac toxicity was higher in patients with Historically overexpression of HER2 was associated
metastatic breast cancer who received trastuzumab with with a poorer prognosis. Ironically, trastuzumab is so
chemotherapy. Cardiac dysfunction was noted in 11/91 effective, that the prognosis for HER2-positive breast
(12%) patients who received paclitaxel plus trastuzumab cancer patients is now better than for those that are
compared to only 1/95 (1%) who received paclitaxel HER2-negative.
alone, and in 28/143 (20%) who received AC plus tras-
tuzumab compared to 7/135 (3%) who received AC Bevacizumab (Avastin®, Genentech,
alone, 27% for combined therapy vs. 8% for anthracy-
cline-based chemotherapy alone, and 13% combined
South San Francisco, CA)
therapy vs. 1% for paclitaxel alone [674]. The higher Bevacizumab and Vascular Endothelial
rate of cardiotoxicity with the doxorubicin plus trastu- Growth Factor
zumab combination may be due in part to increased
drug delivery of doxorubicin to cardiac muscle by loose The anti-vascular endothelial growth factor (VEGF)
binding between doxorubicin and the Mab [182], or monoclonal antibody bevacizumab (Avastin) was approved
additive or synergistic cardiotoxic effects of both agents. in May 2004 on the strength of randomized trials in which
Other trials have also noted higher rates of cardiotoxic- the agent was combined with 5-FU-based chemotherapy
ity in patients who have received anthracyclines prior to for the treatment of metastatic colorectal cancer. In June
or concurrent with trastuzumab [376, 548, 674, 707]. 2006 it was also approved in combination with FOLFOX4
The cardiotoxicity of trastuzumab may relate to HER2 (5-fluorouracil, leucovorin, and oxaliplatin) for the second-
expression on cardiac muscle cells that are involved in line treatment of metastatic colorectal carcinoma. It also
tissue repair, since absence of the HER2/neu gene in has an indication combined with paclitaxel and carbopla-
knockout mice is associated with failure to develop an tin chemotherapy for the treatment of metastatic adeno-
embryonic heart [421]. Results from clinical trials sug- carcinoma of the lung. It is also an indicated for first-line
gest that cardiotoxicity is increased when trastuzumab treatment of metastatic breast cancer in combination with
is administered with or following cardiotoxic agents taxane chemotherapy. Bevacizumab is a recombinant
such as anthracylines. For this reason it is recommended humanized IgG1 Mab that is manufactured in CHO cells
that trastuzumab not be given in combination with an [570]. Unlike other commercially available antibodies
anthracycline or any other agent that is known to dam- that target antigens, bevacizumab binds to the VEGF
age the myocardium. Based on the adjuvant trials, when ligand rather than the VEGF receptor. As discussed ear-
trastuzumab is given after an anthracycline, heart failure lier in this chapter, VEGF is produced by many tumor
rates can be expected to be in the range of 4–5% during cells, is the central mediator of tumor angiogenesis, and
treatment, but there may be an increased risk that various lines of evidence suggest that VEGF would be a
extends beyond treatment. useful target for anticancer therapy [12, 55, 248, 420, 681].
Robert O. Dillman 351

The binding of bevacizumab to VEGF inhibited its bind- In the pivotal trial for regulatory approval, 923 patients
ing to the VEGF receptor in vitro, and had anti-angiogenic with metastatic colorectal cancer were randomized to
and anti-tumor activity in various animal model assay receive irinotecan, 5-FU and leucovorin (IFL) and beva-
systems [12, 55]. cizumab (IFL-BV), IFL and placebo (control), or FL
with bevacizumab (FL-BV) at a dose of 5 mg/kg every 2
weeks [335, 336]. In the first phase of the trial, 313
Clinical Trials with Bevacizumab patients were randomly assigned to these three arms,
Exploratory trials with bevacizumab then; after a planned initial analysis, the FL-BV arm was
Because it blocks VEGF, which is also important in normal discontinued after enrollment of 110 patients, not because
angiogenesis, there was concern that the agent might be of inferior results, but because IFL had become the stan-
associated with bleeding. In phase I trials in 25 patients dard control arm based on other trials in metastatic col-
there were no grade III or IV toxicities associated with i.v. orectal cancer. IFL-BV was superior to IFL-placebo for
doses up to 10 mg/kg [265]. There were no tumor responses, the 813 patients randomized to treatment with one of
but many patients had stable disease. Bevacizumab was these arms, in terms of response rate (p = 0.004), PFS
subsequently given in combination with a variety of che- (HR 0.54, p < 0.001), and OS (HR 0.66, p < 0.001) [335].
motherapy agents without undo toxicity [466]. The IFL-BV arm was associated with more grade 3 or 4
hypertension (11% vs. 2%). The FL-BV arm also was
Colorectal Cancer superior to the IFL-placebo arm [336].
Clinical trials of bevacizumab in colorectal cancer are In a trial of initial therapy for patients with metastatic
summarized in Table 36. A three-arm randomized phase colorectal cancer who were not considered optimal can-
II trial in metastatic colorectal cancer suggested that didates for first-line irinotecan treatment, 209 patients
bevacizumab augmented 5-FU based chemotherapy [359]. were randomized to receive FL + becacizumab at 5 mg/kg
In this trial 144 previously untreated patients were ran- or FL + placebo [361]. The addition of bevacizumab
domized to 5-fluorouracil and leucovorin (FU/LV) + resulted in a better PFS (HR = 0.50, p < 0.001), higher
low-dose bevacizumab (5 mg/kg q 2 weeks), or to FU/ response rate (p = 0.055), and a trend toward better sur-
LV + high-dose bevacizumab (10 mg/kg q 2 weeks) or to vival (HR = 0.79, p = 0.16). Hypertension was more
FU (500 mg/m2)/LV (500 mg/m2 alone. FU/LV was given frequent with bevacizumab.
weekly for the first 6 weeks of each 8-week cycle. Better Because of the emergence of oxaloplatin as an active
results were seen with the addition of bevacizumab, and agent in colorectal cancer, the Eastern Cooperative
there appeared to be an advantage for the lower dose. Oncology Group (ECOG) conducted a three-arm trial in

Table 36. Randomized trials of bevacizumab in the treatment of metastatic colorectal cancer
Citation Clinical setting Treatment arms Number of patients Response rate Median PFS Median OS
[359] Colorectal metastatic untreated FL + BV-5 35 40% 9 months 22 months
FL + BV-10 33 24% 7 months 16 months
FL alone 36 17% 5 months 14 months
[335, 336] Colorectal 1st-line metastatic IFL + BV-5 402 45% 11 months 20 months
IFL + placebo 411 35% 6 months 16 months
FL + BV-5 110 40% 9 months 18 months
[361] Colorectal 1st-line metastatic FL + BV-5 104 26% 9 months 17 months
FL + placebo 105 15% 6 months 13 months
[253] Colorectal relapsed post IFL FOLFOX4 + 290 23% 7 months 13 months
BV-10
FOLFOX 289 9% 5 months 11 months
BV-10 243 3% 3 months 10 months
[391] Pancreas metastatic BV-10 + 602 11% 6 months –
gemcitabine
gemcitabine
10% 6 months –
FL = 5-flurouracil, leucovorin
BV = bevacizumab
FOLFOX = folate (leucovorin), 5-fluroruracil, oxaloplatin
PFS = progression free survival
OS = overall survival
352 Monoclonal antibody therapy

which 829 patients whose cancer had recurred after IFL was the most significant complication associated with
therapy were randomized to receive oxaliplatin, fluorou- bevacizumab and included minor mucocutaneous hem-
racil, and leucovorin (FOLFOX4) with bevacizumab or orrhage, and major hemoptysis associated with centrally
to FOLFOX4 alone, or to bevacizumab alone [253]. In located squamous cell cancers accompanied by tumor
this trial bevacizumab was given at 10 mg/kg every 2 necrosis and cavitation.
weeks. As shown in Table 36, bevacizumab exhibited In a large two-arm randomized trial, 878 patients with
limited activity as a single agent, and FOLFOX4 plus previously untreated non-squamous NSCLC were ran-
bevacizumab was superior to FOLFOX alone for all key domized to PC + bevacizumab (15 mg/kg) or PC alone
endpoints, including OS (HR 0.75, p = 0.001), PFS (HR on a 3 week schedule [629]. The bevacizumab arm was
0.61, p < 0.0001) and response rate (p < 0.0001). There associated with a higher response rate (p < 0.001), better
were higher rates of hypertension and bleeding in the OS (HR = 0.79, p = 0.003), and PFS (HR 0.66, p <
FOLFOX plus bevacizumab arm. 0.001). Even though patients with squamous cell histol-
A metanalysis was performed using the raw data from ogy were not enrolled in this trial, there were still 5
three randomized trials that included 5-FU plus leuco- hemorrhagic deaths in the bevacizumab arm (1.2%) and
vorin and bevacizumab (FL-BV) as initial treatment for significant bleeding was more frequent in the bevaci-
patients with metastatic colorectal cancer [360]. For the zumab arm (4.4% vs. 0.7%, p < 0.001).
combined data, FL-BV (n = 249) was superior to FL (n In a large European trial 1,043 patients with previ-
= 241) in terms of response rate (34% vs. 24%, p = ously untreated non-squamous NSCLC were random-
0.019), PFS (9 vs. 6 months, HR = 0.63, p < 0.001), and ized to gemcitabine plus cisplatin with bevacizumab at
OS (18 vs. 15 months, HR 0.74, p = 0.008). In contrast 10 mg/kg or 5 mg/kg or placebo [463]. Results favored
to these excellent results in previously untreated patients, the bevacizumab arms, although the benefit was not as
in an expanded access trial 350 patients with metastatic impressive as in the U.S. trial with PC.
colorectal cancer, who had relapsed after or progressed In a phase I/II trial, 40 patients with metastatic non-
during both irinotecan and oxaloplatin based therapy, squamous NSCLC who had relapsed after previous
the response rate was only 4% in the first 100 patients chemotherapy were treated with bevacizumab 15 mg/kg
enrolled by investigator analysis, and only 1% based on i.v. every 2 weeks and erlotinib 150 mg orally daily
blinded central review [106]. [309]. Eight patients had a partial response.
ECOG conducted a trial with IFL and bevacizumab in
87 patients with untreated advanced colorectal cancer Breast
[252]. The response rate among 81 evaluable patients Miller et al. randomized 462 patients to p.o. capecit-
was 49%, but the dose of irinotecan and 5FU both had abine 2,500 mg/m2 twice daily days 1 through 14 every
to be reduced by 20 to 25% because of vomiting, diar- 3 weeks, with or without i.v. bevacizumab 15 mg/kg on
rhea and neutropenia. Bleeding occurred in 37 patients day 1 [484]. There was more grade 3 or 4 hypertension
(46%) and nine patients (11%) had grade 3 or 4 throm- requiring treatment (18% v 0.5%) in patients receiving
boembolic events. bevacizumab. The response rate was twice as high for
FOLFOX4 and bevacizumab was used to treat 53 the combination therapy as capecitabine alone, but this
patients with previously untreated metastatic colorectal did not result in superior survival, which is not surpris-
cancer [208]. The response rate was 68%. Hemorrhage ing since most of these patients had failed at least three
was not a problem in this trial, and only one patient had chemotherapy regimens prior to entering the trial.
a severe thromboembolic event. In a randomized trial bevacizumab plus paclitaxel
was compared to paclitaxel alone as initial therapy for
Non-Small Cell Lung 722 patients with metastatic breast cancer [482]. There
Randomized clinical trials involving bevacizumab in was an improvement in response rate and PFS, but not
tumor types other than colorectal cancer are summa- in OS, and there was an increased risk of complications
rized in Table 37. In a three-arm randomized phase II in in the bevacizumab arm. The addition of bevacizumab
non-small cell lung cancer (NSCLC), 99 previously was associated with a longer prolonged progression-free
untreated patients were randomized to treatment every 3 survival (median, 12 vs. 6 months; hazard ratio for pro-
weeks with paclitaxel 200 mg/m2 and carboplatin (AUC gression, 0.60; p < 0.001) and increased the objective
= 6) (PC) alone or in combination with bevacizumab at response rate (37% vs. 21%, p < 0.001). The overall
7.5 or 15 mg/kg [354]. The response rate and PFS were survival rate, however, was similar in the two groups
higher for the 15 mg/kg bevacizumab arm compared to (median, 27 vs. 25 months; hazard ratio, 0.88; p = 0.16).
PC alone with a trend toward better survival. Bleeding Grade 3 or 4 hypertension (15% vs. 0%, p < 0.001),
Robert O. Dillman 353

Table 37. Randomized trials of bevacizumab in the treatment of metastatic cancers other than colorectal cancer
Citation Clinical setting Treatment arms Number of patients Response rate Median PFS Median OS
[354] NSCLC IIIB or IV NSCLC PC + BV-15 35 32% 7 months 18 months
PC + BV-7.5 32 28% 4 months 12 months
PC 32 19% 4 months 15 months
[629] NSCLC IIIB or IV PC + BV-15 434 35% 6 months 12 months
non-squamous
PC 444 15% 4 months 10 months
[463] NSCLC IIIB or IV GC + BV-15 351 34% 6.7 months –
non-squamous
GC + BV-7.5 345 30% 6.5 months –
GC + Placebo 347 20% 6.1 months –
[484] Breast metastatic Cap + BV-15 231 19% 4.9 months 15 months
Capecitabine 231 9% 4.2 months 14 months
p = 0.001 NSD NSD
[482] Breast metastatic Pac + BV-10 368 37% 12 months
Pac 354 21% 6 months
[799] Renal cell metastatic BV-10 39 10% 4.8 months
BV-3 37 0% 3.0 months
Placebo 40 0% 2.5 months
[210] Renal cell metastatic IFN + BV-10 327 31% 10 months
IFN + Placebo 322 12% 5 months
p < 0.001 p > 0.001 p = 0.067
[391] Pancreas metastatic Gem + BV-10 301 11% 6 months
Gecitabine 301 10% 6 months
NSCLC = non small cell lung cancer
BV = bevacizumab, numbers refer to mg/kg
PC = paclitaxel, carboplatin
GC = gemcitabine, carboplatin
IFN = interferon-α
Gem = gemcitabine
Pac = paclitaxel
NSD = no significant difference
PFS = progression free survival
OS = overall survival
NSD = No significant difference

proteinuria (4% vs. 0%, p < 0.001), headache (2% vs. associated with a loss of suppressor gene function and
0%, p = 0.008), cerebrovascular ischemia (2% vs. 0%, induction of genes that are regulated by hypoxia, one of
p = 0.02), and infection (9% vs. 3%, p < 0.001), were which is the gene associated with production of VEGF.
more frequent in patients receiving paclitaxel plus For this reason, renal cell carcinoma was considered an
bevacizumab. excellent target for bevacizumab trials. In a randomized
The combination bevacizumab 10 mg/kg on days 1 double-blind, phase II trial, bevacizumab at 3 and 10 mg/kg
and 15 of a 28-day cycle in combination with docetaxel every 2 weeks was compared to placebo in patients with
35 mg/m2 on days 1, 8, and 15 was associated with a metastatic renal cell cancer [799]. After randomization
52% response rate in patients with metastatic breast of 116 patients, the trial was stopped early because there
cancer who had not been heavily treated with prior che- was a significant prolongation of progression of free
motherapy [579]. survival in the 10 mg/kg bevacizumab arm compared
with placebo (hazard ratio, 2.55; p < 0.001). At the time
Renal Cell the study was discontinued, the 5 mg/kg bevacizumab
Hypervascularity has long been noted as a feature of arm also had a slightly longer PFS compared to placebo
renal cell carcinoma. Most cases of sporadic clear cell that did not quite reach statistical significance (hazard
carcinoma of the kidney are associated with loss of ratio, 1.26; p = 0.053). There was no difference in sur-
heterozygosity von Hippel-Lindau (VHL) gene on chro- vival, but this analysis was of uncertain significance
mosome 3, and inactivation of the VHL allele. This is since cross-over to bevacizumab was permitted in the
354 Monoclonal antibody therapy

placebo group. Hypertension and asymptomatic protei- or bevacizumab plus the maximum tolerated dose of
nuria were the main side effects noted. thalidomide per dose escalation. The combination was
Large randomized phase III trials comparing inter- well-tolerated with more than 50% of patients able to
feron-alpha (IFN-α) plus bevacizumab with IFN-α escalate their thalidomide dose to at least 500 mg/day.
alone in patients with metastatic renal cell cancer Toxicities were similar for both treatments and there
have shown an advantage for the addition of the anti- were no objective responses. Progression free survival was
VEGF antibody. IFN-α was given at a dose of 9 MIU similar between groups, 3.0 months for bevacizumab plus
s.c. every other day thrice weekly and bevacizumab thalidomide and 2.4 months for bevacizumab alone.
10 mg/kg i.v. every 2 weeks in a large European trial
[210]. A similar comparison is ongoing in the US [597]. Gastric and Gastroesophageal Junction
Hypertension and proteniuria were much more common Adenocarcinoma
in the bevacizumab arm. Non-randomized trials involving bevacizumab in various
In a phase II trial patients with metastatic clear-cell tumor types are summarized in Table 38. Forty-seven
renal carcinoma were treated with bevacizumab 10 mg/ patients with metastatic or unresectable adenocarcinoma
kg i.v. every 2 weeks and erlotinib 150 mg orally daily of the stomach or gastroesophageal junction were treated
[280]. The objective response rate was 20%. The most every 3 weeks with bevacizumab 15 mg/kg on day 1,
common adverse events were mild to moderate rash and irinotecan 65 mg/m2, and cisplatin 30 mg/m2 on days 1
diarrhea which were attributed to erlotonib, and protei- and 8 [659]. Possible bevacizumab-related toxicity
nuria which was attributed to bevacizumab. included grade 3 hypertension (28%), grade 3 to 4
In dose-finding phase I–II trials, thalidomide, which thromboembolic events (25%) grade 3 to 4 thromboem-
also has anti-antigenesis properties was combined with bolic events (25%), gastric perforation (6%), myocar-
bevacizumab in patients with metastatic renal cell cancer dial infarction (2%), and significant upper gastrointestinal
whose disease had progressed after receiving placebo bleed (2%). The primary tumor was unresected in 40
in another trial [204]. Sequential cohorts of 10 to 12 patients, but only one patient had a significant upper
patients were treated with bevacizumab 3 mg/kg alone gastrointestinal bleed.

Table 38. Single arm phase II trials of bevacizumab with or without chemotherapy
Citation Clinical setting I.V. dose and schedule Number of patients Response rate
[106] Colorectal: metastatic refractory BV-5 q 2 weeks + bolus or 100 4%
to irinotecan & oxaloplatin continuous FL
[252] Colorectal: advanced, untreated BV-10 q 2 weeks + IFL 81 49%
[208] Colorectal: chemo-naïve metastatic BV-5 q 2 weeks + FOLFOX 53 68%
[579] Breast: metastatic breast 1st BV-10 q 2 weeks = docetaxel 27 52%
or 2nd line
[310] Lung: NSCLC non squamous, BV-15 q 3 weeks + erlotonib 40 20%
previously treated, recurrent
[659] Gastric and GE: adenocarcinoma BV-15 q 3 weeks + irinotecan + 34 65%
cisplatin
[809] Hepatocellular BV-10 q 2 week + GemOx 30 20%
[132] Pancreas: locally advanced, BV 2.5–10 q 2 weeks Capecitabine + 48 20%
inoperable RT
[66] Ovarian relapsed BV-15 q 3 weeks 62 21%
[87] Ovarian relapsed BV-15 q 3 weeks 44 16%
[390] Pancreas: metastatic BV-10 day 1 & 15 + Gemcitabine 52 21%
[281] Renal cell:clear cell, metastatic BV-10 q 2 weeks + erlotonib 59 25%
[143] Sarcoma: soft tissue BV-15 q 3 weeks + Doxorubicin 17 12%
[759] Glioblastoma BV-10 q 2 weeks + 23 61%
Anaplastic Astrocytoma Irinotecan 9 67%
BV = bevacizumab
IFL = irinotecan, 5-flurouracil, leucovorin
FL = 5-flurouracil, leucovorin
GemOx = gemcitabine, oxaloplatin
RT = radiation therapy
Robert O. Dillman 355

Hepatocellular events (7%), bleeding (2%), and wound-healing complica-


Bevacizumab has been given to patients with measur- tions (2%) [87]. There were three treatment-related
able unresectable or metastatic HCC [809]. For cycle deaths (7%). The rate of bowel perforation was unusually
1 (14 days), bevacizumab 10 mg/kg was administered high in this trial, but occurred in 24% of patients who had
alone i.v. on day 1. For cycle 2 and beyond (28 days/ received three prior chemotherapy regimens, compared to
cycle), bevacizumab 10 mg/kg was administered on 0% in patients who received less than three.
days 1 and 15, gemcitabine 1,000 mg/m2 was admin- It seems likely that the high rate of small bowel per-
istered as a dose rate infusion at 10 mg/m2/min fol- foration is the result of rapid elimination of bowel wall
lowed by oxaliplatin at 85 mg/m2 on days 2 and 16. tumor implants, and/or and additional insult to bowel
The most common treatment-related grade 3 to 4 tox- that has been damaged in some manner by chemotherapy
icities included leukopenia/neutropenia, transient ele- [87]. However, a broader experience based on multiple
vation of aminotransferases, hypertension, and fatigue. trials suggests that this complication is observed in only
about 5% of all ovarian patients [292]. The risk of this
complication appears to be increased in more heavily
Pancreas
treated patients. A three-arm trial has been started which
Bevacizumab has been a disappointment in cancer of the
compares paclitaxel plus carboplatin (PC) with placebo
pancreas. In one trial, 48 patients with inoperable pancre-
followed placebo, PC with bevacizumab followed by
atic adenocarcinoma received bevacizumab 2 weeks
placebo, and PC with bevacizumab followed by bevaci-
before radiotherapy, then every 2 weeks during radiother-
zumab for 15 months.
apy, and then after radiotherapy until disease progression
[132]. Each cohort included 12 patients at doses of 2.5,
Prostate
5.0, 7.5, and 10 mg/kg). Capecitabine was administered
Bevacizumab was combined with APC8015 (sipuleucel-
on days 14 through 52. Four had ulceration and bleeding
T) in the treatment of 22 patients with hormone-refractory
in the radiation field possibly related to bevacizumab.
prostate cancer [598]. Sipuleucel-T is a cellular prostate
Three patients had tumor-associated bleeding duodenal
cancer vaccine containing T-lymphocytes and dendritic
ulcers, and one had a duodenal perforation. [132]. In
cells loaded with a recombinant prostatic acid phosphatase
another trial, patients with previously untreated meta-
and granulocyte-macrophage-colony-stimulating factor
static pancreatic cancer received gemcitabine 1,000 mg/
fusion protein. Bevacizumab was given at a dose of 10 mg/
m2 IV over 30 min on days 1, 8, and 15 every 28 days and
kg i.v. on weeks 0, 2, 4, and every 2 weeks thereafter while
bevacizumab, 10 mg/kg IV after gemcitabine on days 1
sipuleucel-T was given i.v. weeks 0, 2, and 4. One patient
and 15 [390]. Grade 3 and 4 toxicities included hyperten-
had a >50% decrease in PSA.
sion in 19% of the patients, thrombosis in 13%, visceral
perforation in 8%, and bleeding in 2%.
Gliomas
Cancer and Leukemia Group B conducted a random-
In a phase II trial adult patients with recurrent anaplastic
ized phase III trial of gemcitabine plus bevacizumab vs.
astrocytoma or glioblastoma (grade III or IV glioma)
gemcitabine plus placebo [391]. There was no advantage
received bevacizumab at 10 mg/kg and irinotecan i.v.
for the addition of bevacizumab in terms of response,
every 2 weeks of a 6-week cycle [759]. The dose of irino-
rate, PFS or OS.
tecan was determined based on whether patients were taking
antiepileptic drugs that induced hepatic enzymes that
Ovarian Cancer accelerate metabolism of irinotecan. Patients taking
In contrast to reported results for other tumor types, enzyme-inducing antiepileptic drugs received irinotecan
single-agent bevacizumab is quite active in the treatment at 340 mg/m2, while other patients received irinotecan at
of ovarian cancer with response rates of 15–20% in 125 mg/m2. Although the significance of radiographic
patients who had progressive disease after two or more changes in treated gliomas is questionable, based on study
courses of chemotherapy [66, 87, 292]. The most frequent design the authors alleged an overall response rate of 63%
serious adverse events that were potentially related to for all patients. In other trials of bevacizumab patients
bevacizumab and noted by Burger et al. were hypertension with brain metastases were excluded because of concerns
(10%) and thromboembolic events (3%) [66]. Cannistra that there might be an increased risk of intracerebral
et al. treated reported a 16% response rate in 44 patients, hemorrhage. However, in this trial there were no instances
but serious toxicities included proteinuria (16%), GI per- of central nervous system hemorrhage, but four patients
foration (11%) hypertension (9%), arterial thromboembolic (12%) experienced thromboemboic complications.
356 Monoclonal antibody therapy

Sarcomas and waiting at least 2 to 3 weeks after surgery before


Seventeen patients with metastatic soft tissue sarcoma instituting bevacizumab therapy. Bowel perforation has
were treated with doxorubicin at 75 mg/m2 i.v. followed been a problem in the setting of gastrointestinal and ovarian
by bevacizumab 15 mg/kg i.v. every 3 weeks [143]. cancers that are prone to produce tumor implants in the
Dexrazoxane was given as a cardio-protectant once the bowel wall, and may be a result of anti-tumor effect.
total doxorubicin dose exceeded 300 mg/m2. Six patients Interestingly, the adverse events have varied somewhat
developed cardiac toxicity grade 2 or greater despite by tumor types, and some may reflect anti-tumor effects
close monitoring and standard use of dexrazoxane. rather than non-specific vascular effects. For instance,
massive hemoptysis has typically been limited to patients
with large centrally located lung cancers, and bowel per-
Mechanisms of Anti-Tumor Activity foration has been more common in colon cancer and
Bevacizumab probably promotes anti-tumor effects by a especially ovarian cancer, entities often associated with
number of mechanisms. The original concept for bevaci- metastatic implants on bowel surfaces. Although patients
zumab activity was that it would decrease tumor angio- with brain metastases have been excluded from many
genesis by blocking VEGF, resulting in a decrease in studies, in part because of a fear of intracranial hemor-
tumor blood supply which in turn would lead to cancer rhage, bleeding has not been a problem in patients with
cell death. However, it appears that bevacizumab treat- glioblastoma who have received bevacizumab.
ment is also associated with afferent vascular dilatation Hypertension and proteinuria occur in all settings,
and efferent vascular constriction of tumor vessels, which and may be due to effects on small renal vessels since
may help concentrate chemotherapy at the tumor site. VEGF is involved in repair of glomeruli endothelial
One study in patients with rectal cancer showed that the cells. Hypertension is usually readily controlled with a
bevacizumab decreased tumor perfusion, vascular volume, single anti-hypertensive agent. Protenuria can progress
interstitial fluid pressure, microvascular density and to full-blown nepthrotic syndrome. Patients should be
circulating endothelial cells while increasing the propor- monitored for protenuria and treatment discontinued if
tion of blood vessels with percytes [785]. The decrease in protenuria exceeds 2 g/24 h. Increased rates of expistaxis
interstial fluid pressure might allow easier penetration for have been noted in many trials, but increased rates of
the Mab itself, which might carry some chemotherapy vascular thrombosis have also been noted.
molecules with it. Another study in patients with inflam-
matory breast cancer demonstrated a decrease of in phos- Summary
phorylated VEGFR2 in tumor cells, a decrease in vascular Despite some safety concerns, bevacizumab has been a
permeability, and an increase in tumor cell apoptosis, but tremendous addition to our therapeutic armamenterar-
no significant changes in microvessel density or VEGF-A ium. Unlike most targeted therapies, it is potentially
expression [771]. In the pivotal trial of IFL and bevaci- useful in every type of solid tumor because of the impor-
zumab as initial treatment for metastatic colon cancer, tance of tumor angiogenesis. Bevacizumab seems to
levels of epithelial and stromal VEGF, stromal thrombos- enhance chemotherapy in virtually every tumor type in
pondin-2, and microvessel density failed to predict clinical which chemotherapy is active, and is already in wide-
benefit associated with the addition of bevacizumab as spread use in cancers of the colon, lung, breast, brain,
opposed to placebo [356]. ovary, and kidney. Although its single-agent activity
appears limited, bevacizumab alone may be useful in
the adjuvant setting, or following complete remission
Toxicities and Adverse Events after systemic therapy, because of the inhibition of
The toxicities associated with bevacizumab are summa- neoangiogenesis required to develop metastatic tumors.
rized earlier in this chapter in Table 6. The package insert
for the product warns of hemorrhage, hypertension, pro- Cetuximab (Erbitux®), Bristol-Myers
teinuria, impaired wound healing, GI perforation, and
congestive heart failure. The most serious, and sometimes Squibb
fatal, bevacizumab associated toxicities are gastroin- Cetuximab and the Epidermal Growth
testinal perforation, wound-healing complications, hem-
orrhage, arterial thromboembolic events, hypertensive
Factor Receptor
crisis, nephrotic syndrome, and congestive heart failure The rationale for targeting the epidermal growth factor
[211]. The impaired wound-healing has led to sugges- receptor (EGFR), which is expressed to some extent on
tions of waiting at least 4 to 6 weeks after bevacizumab all epithelial tumors, was addressed earlier in this chapter.
has been discontinued before taking a patient to surgery, The human epidermal growth factor receptor (EGFR),
Robert O. Dillman 357

and its ligands, such as transforming growth factor alpha The anti-EGFR chimeric Mab cetuximab (Erbitux)
(TGF-α, have long been recognized as a potential targets was approved in 2004 based on randomized trials in
for antibody-based therapy. EGFR is associated with which the agent was combined with the chemotherapy
tyrosine kinase activity, and is frequently overexpressed agent irinotecan in the treatment of metastatic colorectal
in cancers of the breast, ovary, bladder, head and neck, cancer. It is a recombinant chimeric monoclonal anti-
prostate, pancreas, and colon. EGFR are expressed at body that includes a murine Fv and human IgG1 kappa
high levels in about one third of all epithelial cancers, and constant regions. Unlike most monoclonal antibody
are associated with accelerated growth [480]. Autocrine products, cetuximab is manufactured in murine plasma-
activation and overexpression of EGFR appears to be cytoma cells. This cellular factory results in murine
crucial for the accelerated growth of many cancers and is glycosylation which may increase the risks of allergic
associated with increased expression of VEGF. reactions in some patients.
The murine antibody to EGFR-1 called 225 was
shown to block receptor function and inhibit cell growth
in cultures in nude mouse xenografts [471]. The Mab
Clinical Trials with Cetuximab
bound to the extracellular domain of EGFR causing Colorectal cancer
internalization of the receptor and antibody and “down- Many of the initial trials with cetuximab, as a single
regulation” of the receptor. For this reason most early agent and in combination with chemotherapy, were con-
development with anti-EGFR antibodies focused on ducted in patients with colorectal cancer, as summarized
immunoconjugates, especially ricin immunotoxins [472]. in Table 39.
Cetuximab, the chimeric form of the antibody, was actu- Trial designs included cetuximab alone, and cetuximab
ally more effective in the mouse tumor models than the in combination with irinotecan in patients with metastatic
murine antibody, apparently because of a higher affinity colorectal cancer that had previously progressed in the
for the EGFR target [571]. Internalization of the recep- face of irinotecan. In these trials cetuximab was given at
tor is slow enough that cetuximab is cytotoxic in vitro in an initial loading dose of 400 mg/m2 i.v. over 2 h, then
the presence of human effector cells and complement, 250 mg/m2 i.v. over 1 h weekly.
but its major anti-tumor mechanism is believed to relate Saltz et al. treated 57 patients whose metstatic cancer
to the receptor blockade preventing signal transduction had not responded to prior irinotecan alone or as part of
that is associated with cellular proliferation. In labora- combination chemotherapy [628]. Patients were required
tory tests against tumor cells and in animal models with to have EGFR demonstrated on formalin-fixed paraffin-
human tumor xenografts, cetuximab enhanced the chemo- embedded tumor tissue by IHC, but it is now known that
therapy effects of many different chemotherapy agents quantitative expression of EGFR is not predictive of
including platinums, campothecins, taxanes, fluoropy- response to cetuximab treatment. The most common
rimidines,, and gemcitabine. adverse events were an acne-like skin rash, predominantly

Table 39. Trials of cetuximab in patients with metastatic colorectal cancer


Median time to Median overall
Citation Regimen Clinical setting # of patients Response rate progression survival
[628] Cetuximab Prior irinotecan 57 9% 1.4 months 6.4 months
[424] Cetuximab Prior 5FU, irinotecan, 346 12% 1.4 months 6.6 months
oxaloplatin
[355] Cetuximab vs. Prior 5FU, irinotecan, 287 8% 41% 3 months 6.1 months
supportive care oxaloplatin
285 0% 24% 3 months 4.6 months
p = 0.001 p = 0.005
[136] Cetuximab Irinotecan refractory 111 11% 1.5 months 8.6 months
Cetuximab + irinotecan 218 23% 4.1 months 6.9 months
[245] Cetuximab + irinotecan Prior 5FU, irinotecan, 60 20% 3.1 6 months
oxaloplatin
[752] Cetuximab + irinotecan Refractory to irinotecan 55 25% 4.7 months 9.8 months
or oxaloplatin
[703] Cetuximab + FOLFOX First-line 43 72% 12.3 months 30 months
5FU = 5-fluorouracil
FOLFOX = folate (leucovorin), 5-fluororuracil, oxaloplatin
358 Monoclonal antibody therapy

on the face and upper chest (86%) and the constellation and cetuximab as the initial treatment for 43 patients
of asthenia, fatigue, malaise, or lethargy (56%). Three with metastatic colorectal cancer [703]. Cetuximab was
patients were reported to have had a grade 3 allergic reac- given day 1 at a dose of 400 mg/m2 during week 1, and
tion; two were withdrawn. then 250 mg/m2 weekly thereafter. Treatment was well-
Lenz et al. treated 346 patients whose metastatic tolerated and the response rate was 72%. In a randomized
cancer was considered refractory to fluoropyrimidines, trial of 337 patients, a preliminary report showed that
irinotecan and oxaliplatin [424]. EGFR positivity by cetuximab plus FOLFOX was superior to FOLFOX
IHC was an eligibility requirement, but degree of posi- alone in terms of response rate (46% vs. 36%, p = 0.064)
tivity did not correlate with clinical benefit. The most [46]. In a randomized trial of 1,198 patients, an initial
prevalent toxicity was the acneiform rash which was report showed that cetuximab plus FOLFIRI was
noted in 83% of patients, which was predictive of clin- superior to FOLFIRI alone in terms of response rate
ical benefit. (47% vs. 39%, p = 0.004) and PFS (8.9 vs. 8.0 months,
In a randomized trial Jonker et al. randomized 572 p = 0.048) [741].
patients who had progressed despite 5FU, irinotecan, Cetuximab is also being explored in rectal cancer.
and oxaloplatin, to standard dose cetuximab or obser- Based on a trial in 40 patients with rectal cancer, pre-
vation [355]. Although the objective response rate was operative radiotherapy in combination with capecit-
only 8%, OS and PFS were better in the cetuximab arm. abine and cetuximab is feasible [452]. Cetuximab in
Severe adverse events of rash and infection were more combination with capecitabine, weekly irinotecan, and
common in the cetuximab arm. Hypomagnesemia was radiotherapy is being evaluated as neoadjuvant therapy
also more common with cetuximab. Eleven patients for rectal cancer [324].
discontinued cetuximab because of infusion reactions.
In a regulatory pivotal trial Cunningham et al. ran- Squamous Cell Cancers of the Head and Neck
domized 329 patients with colorectal cancer, that had Cetuximab has also been extensively evaluated in
progressed during or within 3 months after discontinua- squamous cell cancers of the head and neck as shown in
tion of irinotecan, to receive irinotecan plus cetuximab Table 40. Vermorken et al. gave single-agent cetuximab
or cetuximab alone using a 2: 1 randomization [136]. at the standard dose and schedule to 109 patients who
Protocol prescribed irinotecan was to be the same as had recurrent and/or metastatic disease that progressed
used prestudy. The response rate was higher for the com- during platinum chemotherapy [751]. Acneiform skin
bination therapy (p = 0.007), as was PFS (p < 0.001). rash, which occurred in 49%, was the most common
Gebbia et al. treated 60 patients who had received at toxicity There was one death due to an infusion-related
least two prior therapies, and whose metastatic cancer was allergic reaction. The response rate was similar to that
considered refractory to oxaliplatin and irinotecan [245]. of single-agent cetuximab in colorectal cancer.
Standard cetuximab dosing was combined with irinotecan Phase I trials suggested that cetuximab could be
120 mg/m2 weekly for 4 out of 6 weeks. Responses were safely given with radiation therapy and might enhance
not predicted by EGFR expression. The main grade 3 and therapeutic benefit [602]. Bonner et al. conducted a
4 toxicities were mostly attributable to the chemotherapy, multinational trial comparing cetuximab plus radiother-
and included nausea (33%), diarrhea (27%), leukopenia apy to radiotherapy alone in 424 patients with locore-
(18%), asthenia (13%), and acneiform skin rash (13%). gionally advanced disease, that showed a benefit for the
Vincenzi et al. treated 55 patients whose metastatic addition of cetuximab including a 26% reduction in
cancer was considered refractory to oxaliplatin and iri- death with an increase in survival from 29 to 49 months
notecan [752]. Standard cetuximab dosing was combined (p = 0.03) [47]. There was no increase in toxicity other
with irinotecan 90 mg/m2 weekly. Skin toxicity was than the acneiform rash.
observed in 89% of patients. The most common grade 3 Pfister et al. treated 22 patients locoregionally advanced
to 4 adverse events were dermatologic (33%), diarrhea disease with cisplatin, cetuximab, and 70 Gy of radiother-
(16%), fatigue (13%) and stomatitis (7%). Fever was apy [553]. This trial was closed early because of several
noted 25%, typically in association with the first infusion adverse events related to infection and cardiac events.
of cetuximab, but no allergic reactions were recorded. Grade 3 or 4 cetuximab-related toxicities included acnei-
More recently cetuximab has been combined with form rash in 10% and hypersensitivity in 5%. With a
modern combination therapies such as FOLFOX and median follow-up of 52 months, the 3-year overall sur-
FOLFIRI as the initial treatment of patients with meta- vival rate is 76%, the 3-year progression-free survival rate
static colorectal cancer. Tabernero et al. used FOLFOX is 56%, and the 3-year locoregional control rate is 71%.
Robert O. Dillman 359

Table 40. Trials of cetuximab in patients with squamous cell cancer of the head and neck
Citation Treatment regimen Clinical setting Number of patients Response rate Median PFS
[751] Cetuximab Prior platinum, recurrent, 103 13% 1.6 months
metastatic
[47] Cetuximab or Placebo + 70 Gy RT Loco-regional advanced 211 – 24 months
213 – 15 months
p = 0.005
[553] Cetuximab + Cisplatin + RT Stage III or IV, M0 22 – 56% 3-year
[309] Cetuximab + cisplatin Refractory to cisplatin + 5FU 130 13% –
or paclitaxel
[26] Cetuximab + cisplatin refractory to cisplatin 96 10% 2.8 months
[72] Cisplatin + cetuximab Recurrent or metastatic 117 26% 4.2 months
Cisplatin + placebo 10% 2.7 months
p = 0.03
[54] Cetuximab + 5FU + Cis- Recurrent or metastatic 53 36%
or carboplatin
[102] Carboplatin Nasopharynx recurrent 59 12% 3.2 months
or metastatic
5FU = 5-fluoruracil
PFS = progression free survival

Herbst et al. treated 130 patients, who did not have Six patients (10%) experienced serious treatment-related
an objective response or relapsed within 90 days of adverse events during cetuximab.
completing two cycles of cisplatin/paclitaxel or cispla-
tin/fluorouracil, with standard cetuximab and cisplatin Cancers other than Colorectal and Head and Neck
(75 or 100 mg/m2 i.v.) every 3 weeks [310]. The most Exploratory trials with cetuximab have been conducted
common toxicities were anemia, acneiform skin rash, in a variety of other tumor types as shown in Table 41.
leukopenia, fatigue/malaise, and nausea/vomiting. Pinto et al. treated 34 patients who had advanced gastric
Seven patients (5%) experienced a grade 3 or 4 hyper- or gastroesophageal junction adenocarcinoma with
sensitivity reaction to cetuximab. cetuximab and the FOLFIRI regimen as first-line therapy
Baselga et al. treated 96 patients, who were refractory for up to 24 weeks, with cetuximab alone continued for
to cisplatin, with standard cetuximab and cisplatin at the patients whose tumors had not progressed [561]. Median
same dose and schedule during which progressive dis- survival was 12 months. Grade 3 to 4 toxicity included
ease had occurred [26]. Acneiform rash was the most neutropenia (42%), acneiform rash (21%), diarrhea
common toxicity. (8%), asthenia (5), stomatitis (5), and hypertransami-
Burtness et al. randomized 117 patients who had nasemia (5%) with one treatment-related death.
recurrent or metastatic disease to receive cisplatin every In pancreatic cancer, Xiong et al. treated 41 untreated
4 weeks with weekly cetuximab or placebo [72]. patients who had measurable locally advanced or meta-
Response rate was higher with the addition of cetux- static pancreatic cancer and whose tumors had EGFR
imab, but PFS and OS were not improved. Survival was expression by IHC, with cetuximab and gemcitabine
better for those patients who developed the skin rash. [797]. Median survival was 7.1 months and 1-year sur-
Bourhis et al. treated 53 patients who had recurrent or vival was 32%. The most common grade 3 to 4 adverse
metastatic disease with a combination of cetuximab, events were neutropenia (39.0%), asthenia (22.0%),
cisplatin or carboplatin, and escalating doses of 5FU as abdominal pain (22.0%), and thrombocytopenia (17.1%).
initial therapy [54]. Dermatologic toxicity was the most Despite the encouraging results of this phase II trial,
common adverse event, but the most common grade 3 or preliminary results of a randomized trial conducted by
4 adverse events were leucopenia (38%), asthenia the Southwest Oncology Group (SWOG) in 735 patients
(25%), thrombocytopenia (15%), vomiting (14%). with advanced pancreatic cancer showed no advantage
Chan et al. treated 59 patients who had recurrent or for the combination therapy over gemcitabine alone
metastatic nasopharyngeal cancer that had recurred with a response rate of only 12%, and the same median
after cisplatin, with cetuximab and carboplatin [102]. survival of approximately 6 months that has been
360 Monoclonal antibody therapy

Table 41. Clinical trials of cetuximab in patients with other tumor types other than colorectal or head and neck
Number of patients Response rate Median PFS Median OS
[561] Cetuximab FOLFIRI Gastric and GE junction 34 44% 8 months 12 months
untreated
[797] Gemcitabine Pancreas advanced 41 12% 3.8 months 7.1 months
[559] Cetuximab + Pancreas advanced 352 12% 3.5 months 6.5 months
Gemcitabine
[293] Cetuximab alone NSCLC previously treated 66 5% 2.3 months 8.9 months
[715] Paclitaxel + NSCLC untreated metastatic 31 26% 5 months 11 months
Carboplatin
[603] Gemcitabine & NSCLC untreated metastatic 35 29% 5.4 months 10.2 months
Carboplatin
[509] Cetuximab Renal cell metastatic 55 0% 1 month 49 months
29 months
p = 0.03
FOLFIRI = folate (leucovorin), 5-fluorouracil, irinotecan
GE = gastroesophogeal
NSCLC = non small cell lung cancer
PFS = progression free survival
OS = overall survival

observed in virtually all randomized trials involving Mechanisms of Anti-Tumor Activity


such patients [559].
and Toxicity
In NSCLC Hanna et al. observed a response rate of
only 5% for cetuximab alone in 66 patients with meta- Sustained blocking of EFGR is believed to be critical
static disease who had progressed after previous systemic for the anti-tumor effects of cetuximab. Based on ran-
therapy [293]. Grade 3 to 4 toxicities were uncommon, domized trials exploring single doses of 50, 100, 250,
but included acneiform rash (6%), anaphylactic reactions 400, or 500 mg/m2, a dose of 250 mg/m2 was predicted
(2%), and diarrhea (2%). Other studies have combined to nearly saturate epidermal growth factor receptors
cetuximab with chemotherapy. [237, 706]. Skin biopsies in 39 patients, before and after
Thienelt et al. treated 31 previously untreated patients i.v. infusions of cetuximab, confirmed a dose-dependent
with paclitaxel, carboplatin, and cetuximab [715]. Over relationship with suppression of EGFR expression [237].
80% of patients experienced dermatologic toxicity, with In these trials there was no association between dose and
13% grade 3 or 4. Robert et al. treated 35 patients with cutaneous rash, but rash predicted for clinical benefit.
gemcitabine and carboplatin [603]. The most common Pharmacokinetic trials of cetuximab with irinotecan
toxicities attributed to cetuximab were acneiform rash revealed no change in cetuximab clearance or metabolism
(89%), asthenia (31), fever/chills (20%), and nausea/ when combined with irinotecan compared to infusions
vomiting (17.1%). of cetuximab alone [154, 213].
In metastatic renal cell cancer, Motzer et al. observed Because all preclinical testing of cetuximab and its
no responses with single-agent cetuximab in 55 patients murine precursors were conducted in animals bearing
[509]. Grade 3 to 4 skin toxicity was noted in 17%. tumors that strongly expressed EGFR, most early trials
These results were so discouraging that cetuximab has restricted patient eligibility to those whose tumors had
not been evaluated further in kidney cancer. documentable expression of EGFR by IHC. However, in
A number of trials are in progress that combine cetux- clinical practice it is now generally accepted that IHC
imab with combined modality chemotherapy plus radia- testing for EGFR expression is not necessary for deter-
tion therapy regimens. A trial combining cetuximab mining appropriateness of cetuximab-based therapy,
with gemcitabine and radiation therapy was being because of inherent limitations in the assays and poten-
conducted in patients with locally advanced pancreatic tial for sampling error, and because responses have been
cancer [408]. A trial of cetuximab with temozolomide described in patients whose tumors were EGFR-negative
and radiation therapy is being conducted in patients by IHC. In eight colorectal cancer patients who were
with previously untreated glioblastoma [126]. Cetuximab refractory to irinotecan, and whose tumors were nega-
plus intensity modulated radiation therapy in non small tive for EGFR by IHC, four responded to cetuximab
cell lung cancer [349]. plus irinotecan therapy [301]. A small trial of 31 patients
Robert O. Dillman 361

suggested that EGFR gene copy number might predict locoregionally advanced squamous cell cancers of the
benefit [504]. However, in a much larger prospective head and neck. It will be interesting to see randomized
trial, cell surface expression of EGFR, EGFR kinase comparisons of chemotherapy plus radiation vs. cetux-
domain mutations, and EGFR gene amplification did imab plus radiation, but there are also trial comparing
not predict clinical benefit, probably because of the het- cetuximab plus chemoradiotherapy to chemoradiother-
erogeneity of tumors [355, 424]. A pilot study involving apy alone. There may be a role for cetuximab with che-
tumors from 39 colorectal cancer patients suggested that motherapy in NSCLC, but again bevacizumab with
cyclin D1 A870G and EGF A61G polymorphisms may chemotherapy is currently the standard initial therapy.
predict efficacy of single-agent Cetuximab [806].
The most common side effect associated with cetux-
imab and other anti-EGFR therapies is a characteristic
Panitumumab (Vectibix™) Amgen,
acneiform rash, which is most prominent on face, chest Thousand Oaks, California
and upper back [74]. In many trials the prevalence of this
Panitumumab and EGFR
side effect was over 80%. The rash usually appears within
a week of starting treatment. Serial punch biopsies in The epidermal growth factor receptor (EGFR) mono-
patients revealed two main reaction patterns: a superficial clonal antibody panitumumab (Vectibix) was approved
dermal inflammatory cell infiltrate, and a superficial in 2006 based on randomized trials in which the agent
folliculitis. The rash can be severe and should be treated was superior to placebo in patients with relapsed, refrac-
to decrease the risk of secondary infection and cellulites. tory, metastatic colorectal cancer. Formerly known as
Grade 3 to 4 dermatitis was reported in 5–15% of patients ABX-EGF, panitumumab was the first fully human
in most trials. Prophylactic topical lubricants are suffi- monoclonal antibody to receive regulatory approval and
cient in many patients, but others require treatment with the first totally human Mab approved for cancer therapy
tetracycline antibiotics or clindamycin. Other common [115, 120]. It is a human IgG2. Like the chimeric Mab
side effects include asthenia and diarrhea. Cetuximab cetuximab, the human Mab panitumumab binds to the
does not appear to increase the toxicity of any of the che- extracellular domain of EGFR causing internalization
motherapy agents with which it has been combined. of the receptor and antibody and “down-regulation” of
Black box warnings for cetuximab include infusion the receptor with disruption of potential downstream
reactions and cardiac arrest. The somewhat high rate of signal transduction that enhances proliferation and
severe infusion related reactions, many of which have resistance to apopotosis in normal and malignant cells.
been characterized as anaphylactic reactions, is of con- Panitumumab was produced with the hope that its clini-
cern. At this time it is not clear whether this is actually cal efficacy might be even greater than cetuximab
do to cross-reactivity with antigens on circulating cells because it is a totally human construct, which may offer
in some patients, effects of Fc interactions with effector advantages in terms of pharmacokinetics, decreased
cells, or a true allergic reaction to murine amino acids allergenic potential, and possibly enhanced ADCC. The
and/or glycosylation because of the murine plasmacy- anti-tumor activity of panitumumab was confirmed in vitro
toma cell line used to produce the product. There was a and in vivo, against many cancers, including lung, kidney
2% risk of sudden death in 208 patients with squamous and colorectal. In these models it was well tolerated and
cell cancer of the head and neck who received cetux- clinically active both as monotherapy and in combination
imab with radiation therapy [47]. chemotherapy agents. Panitumumab was originally made
in transgenic mice, but is manufactured in CHO cells.
Summary
Clinical Trials with Panitumumab
Cetuximab is widely used in combination with chemo-
therapy for patients who have relapsed after primary The few clinical trials published for panitumumab are
treatment for colorectal cancer. However, based on the summarized in Table 42. A pivotal trial for regulatory
comparative strengths and differences of clinical trials, approval compared panitumumab at 6 mg/kg i.v. every
bevacizumab with combination chemotherapy is usually 2 weeks to best supportive care in patients with meta-
preferred as the initial treatment. Cetuximab is increas- static colorectal cancer whose disease had progressed
ingly being used with radiation therapy in patients who during or after standard therapy with fluoropyrimidine-,
are considered poor candidates to receive combined oxaliplatin-, and irinotecan-containing chemotherapy
modality treatment with chemotherapy and radiation regimens [254, 255, 741, 742]. All patients had epider-
therapy, which is currently standard for treatment of mal growth factor receptor (EGFR)-expressing tumors.
362 Monoclonal antibody therapy

Table 42. Trials of panitumumab in patients with metastatic colorectal cancer


Median time to Median overall
Citation Clinical setting Treatment # of patients Response rate progression survival
[741] Progressed after prior chemo Panitumumab 231 10% 8 weeks –
Supportive care 232 0% 7.3 weeks –
p < 0.0001
[302] Progressed after prior chemo Panitumumab 148 9% 14 weeks 9 months
[35] No prior therapy IFL + panitumumab 19 46% 5.6 months 17 months
[35] No prior therapy FOLFIRI + 24 42% 10.9 months 22 months
panitumumab
IFL = irinotecan, 5-fluorouracil, and leucovorin
FOLFIRI = folate (leucovorin), 5-fluorouracil, irinotecan

Panitumumab produced a 10% objective response rate Toxicities


and a longer progression free survival compared to
supportive care alone. There was no difference in The toxicities and adverse events associated with pani-
survival, but approximately 75% of patients in the tumumab are summarized earlier in this chapter in Table 6.
supportive care alone arm crossed over to receive panitu- Panitumumab black box warnings include dermatologic
mumab after disease progression. Quality of life was also toxicities and infusion reactions. Panitumumab was
better in the patients who received panitumumab [668]. developed in the belief that it would be less toxic and
For 176 patients who progressed on the supportive care more active than cetuximab or other less than com-
arm, and then subsequently did receive panitumumab, pletely human antibodies. Panitumumab has been well
12% had an objective response and two patients had tolerated whether administered alone or in combination
complete responses [742]. with chemotherapy. However, there was a 1% rate of
Hecht et al. treated 148 patients whose metastatic col- anaphylaxis in trials submitted for registration of the
orectal cancer had progressed on chemotherapy that agent. During eight clinical trials, a very sensitive assay
included a fluoropyrimidine and irinotecan or oxalipla- detected anti-panitumumab responses in 25 of 604
tin, or both [302]. Panitumumab was given i.v. at a dose (4.1%) subjects, and eight developed neutralizing anti-
of 2.5 mg/kg weekly for 8 of each 9 weeks until disease bodies [445]. There is a report of at least one patient
progression or excessive toxicity. Skin toxicity occurred who had a severe infusion reaction during treatment
in 95% and 5% were grade 3 or 4. Four patients discon- with cetuximab, who was then successfully treated with
tinued therapy because of toxicity and one patient had panitumumab [332], but because most infusion reac-
an infusion reaction, but was able to resume treatment. tions are seen only with a first infusion, it is not clear
EGFR of at least 1+ by IHC was an eligibility require- that decreased toxicity will be an important benefit,
ment. There was no difference in response for 105 since most infusion reactions are due to antigen–antibody
patients who were judged as having high EGFR by IHC interactions rather than antibody reactions to murine
compared to 43 patients who were characterized as components of the chimeric or humanized Mab.
having a low EGFR. Similar to other agents targeting the epidermal growth
Berlin et al. tested the combination of irinotecan, 5-FU factor receptor pathway, skin rash has been the primary
and leucovorin, and panitumumab as initial therapy in toxicity recognized in association with panitumumab
patients with metastatic colorectal cancer [35]. This was therapy, and has occurred in 100% of patients receiving
a two-part, multicenter, phase II study of panitumumab doses of 2.5 mg/kg or higher. Hypomagnesaemia and
2.5 mg/kg weekly with bolus 5-FU (IFL) in the first part diarrhea were also most commonly reported. No grade
of the trial, and infusional 5-FU (FOLFIRI) in the second severe or life-threatening reactions were noted in the
part. Grade 3 to 4 diarrhea occurred in 11 patients (58%) large randomized trial in patients with colorectal cancer
in part 1 and six patients (25%) in part 2. All patients had [741, 742]. Other common adverse events noted were
dermatologic toxicity, but none was grade 4. The authors paronychia, fatigue, abdominal pain and nausea. The
concluded that panitumumab + FOLFIRI was better tol- most serious adverse events were severe dermatologic
erated than panitumumab + IFL. toxicity complicated by infectious sequelae and septic
Robert O. Dillman 363

death, infusion reactions, pulmonary fibrosis, hypomag- medical literature and cannot be considered all incon-
nesemia, and gastrointestinal problems including abdom- clusive because of the volume of unpublished results
inal pain, nausea, vomiting, diarrhea, and constipation. known only to certain companies who have pursued
commercial development of certain antibodies, and the
US FDA.
Summary
Panitumumab is the first fully human antibody to receive
regulatory approval, and joins cetuximab as an anti
Anti-Idiotype Antibodies
EGFR Mab. It remains to be seen whether the to totally The idiotype of malignant B-cell clones is perhaps the
human construct offers any meaningful advantages of best example of a tumor-specific antigen. More than 15
the chimeric construct. years before the first approval of a Mab for the treatment
of malignancy, the first antibody-mediated CR was
reported following administration of a murine anti-
Conclusions idiotype antibody [487]. The patient had follicular B
“Magic bullets” are now part of the standard anti-cancer cell lymphoma that had progressed after prior treatment
therapeutic armamentarium [168]. It has now been more with cyclophosphamide–vincristine–prednisone che-
than a decade since rituximab and trastuzumab became motherapy, and interferon-alpha biotherapy. The complete
the first Mabs approved for the treatment of cancer, and response persisted for over 7 years. Subsequent publica-
both have become well-established “blockbuster drugs.” tions established a 66% response rates among 45 patients
Alemtuzumab would be more widely used in the treat- with indolent lymphoma who received anti-idiotype
ment of hematologic malignancies if its target was more antibodies alone, or administered with single-agent
specific, and rituximab was not so safe and effective in chlorambucil, interferon-alpha, or interleukin-2 [146].
the treatment of B cell malignancies. While five of these Despite the promising early results with anti-idiotype
agents are limited in their scope of malignant targets antibodies, commercial development in this area has not
because of their specificity for surface antigens, bevaci- evolved because of the need to develop a specific anti-
zumab has the potential to be useful in every cancer set- idiotype antibody for each patient. Interest for commercial
ting because of the importance of the VEGF ligand for development increased when several investigators
tumor angiogenesis. Cetuximab and panitumumab are suggested that combinations of anti-idiotype antibodies
potentially useful in the same patient populations, and it commonly expressed on B-cell lymphomas might be a
will be interesting to see whether the totally human Ig useful clinical product [16, 394, 489, 614, 692, 702], but
has any meaningful clinical advantages over the chime- enthusiasm dwindled when the frequency of these
ric product. It is interesting that other than rituximab, “shared” or “cross reactive” idiotypes was found to be
these agents have rather limited anti-tumor effects, but much lower than originally suggested.. The company
all dramatically increase the effects of chemotherapy, IDEC of San Diego, CA, which was originally founded
which is what has led to their rapid and widespread adop- as an anti-idiotype company, subsequently acquired
tion in cancer therapy. The potential for synergistic and Analytical Biosystems of Sunnyvale, California, estab-
additive effects resulting from the use of Mab in combi- lished by Levy and Miller of Stanford, which was the
nation with other biological response modifiers has not first anti-idiotype company to pursue the anti-idiotype
been established, and certainly is not superior to the approach. IDEC eventually dropped development of a
common chemotherapy plus Mab combinations. composite anti-idiotype preparation called Specifid® in
order to focus its resources on a product called C2B8, a
chimeric anti-CD20 antibody that became the block-
buster product rituximab [168].
Other Antibodies and Antigens The largest experience with anti-idiotype antibodies
This section is organized by antigenic targets and covers was in lymphoma as initially reported by Meeker et al.
many other Mab that have investigated but are not [474] and subsequently by others at Stanford who were
approved for clinical use as anti-cancer therapy, although working with Ron Levy [60, 61, 146, 446, 458]. Among 17
some are clinically available based on other indications. patients receiving individualized anti-idiotype antibod-
Some trials that involved tracer quantities of radiola- ies at doses ranging from 400 mg to over 9 g, there were
beled Mabs and are also included if in fact patients nine partial responses including the sustained, complete
received at least several mg of unmodified antibody. The remission in the first patient treated, and others lasting
information described is based on publication in the from 1 to 6 months [478, 487]. Favorable responses were
364 Monoclonal antibody therapy

associated with increased T-cell infiltration of involved Urba et al. treated 36 patients with OKT3 antibody in
lymph nodes [446]. In some cases, resistance to therapy an effort to activate T cells in the hope of promoting an
was shown to result from emergence of idiotype-variant antitumor effect [737]. Five patients received a 30 ug
clones [478]. Stanford investigators combined IFN-α dose by i.v. bolus and the other 23 received 3-h infu-
with anti-idiotype antibodies based on the rationale that sions of 1, 10, 30, or 100 ug. An additional 8 patients
IFN-α might upregulate the idiotype antigen expression, received the anti-CD3 daily for 14 days by either bolus
and IFN-α is known to have antiproliferative effects on 3-h infusion, or 24-h infusion. The dose-limiting toxic-
follicular B-cell lymphoma cells. Of 12 patients treated ity in this study was headache, accompanied by signs
in this manner, who received total antibody doses rang- and symptoms of meningeal irritation. Eight of the 16
ing from 1.7 to 8.0 g, there were two CRs and seven PRs patients tested exhibited HAMA. No tumor responses
[61]. Another trial combined chlorambucil with anti- were observed.
idiotype antibodies and one CR and seven PR were Richards et al. treated 13 patients with OKT3. Six
described in 13 patients [458]. It was not clear that either patients received 50 ug, and 7 received 100 ug [593]. A
of these combination therapies was more effective than partial response was described in one patient with meta-
anti-idiotype antibodies alone because of changes in static renal cell carcinoma. Again, neurotoxicity was a
patient selection and the known efficacy of both IFN-α significant problem and was observed in 11/13 patients
or chlorambucil as single agents in the treatment of fol- after the first treatment. Headache and confusion were
licular lymphoma. Other groups have reported on lim- noted. In all patients, neurotoxicity was transient and
ited trials with anti-idiotype antibodies with limited interestingly, did not recur with re-treatment. In both of
success [99, 291, 581]. Rankin et al. noted minimal anti- these this study and that by Urba et al, a cerebral spinal
tumor effects in two lymphoma patients who received fluid lymphocytosis was noted in patients who under-
murine anti-idiotype Mab at individual doses from 5 to went lumbar puncture, and headache was a frequent
160 mg and total doses of 3.8 and 5.8 g respectively complaint [593, 737]. Because of the evidence that
[581]. Another report relates an initial experience with a OKT3 had stimulated an immune response in the central
mouse/human chimeric anti-idiotype in a patient with nervous system, Wiseman et al. conducted a trial with
B-cell lymphoma who had no tumor response [291]. OKT3 in patients with gliomas who had failed conven-
The brief and limited responses observed in CLL may tional therapy and evidence of progressive disease [789].
have been due to the relatively low doses of Mab admin- Patients received 25 to 75 ug of OKT3 over 1-h, fol-
istered in those trials [99]. lowed a day later by 300 mg/m2 of cyclophosphamide as
a dose intended to reduce effects of suppressor or regu-
Antigens Associated latory T cells. Three of nine patients were reported to
have had objective tumor regressions, based on brain
with Hematopoietic Cells studies with magnetic resonance imaging. Anticipated
Because of the ready availability of clinical material for side effects included headache, fever, stupor, nausea,
testing, many of the first murine Mab were developed emesis, and transient decreases in T-lymphocyte counts,
after immunization with hematopoietic cells. The vari- but no severe or life-threatening toxicity.
ous antigens identified on blood cells were classified by Several investigators have explored the use of the
a cluster designation (CD) numbering system [36]. T-lymphocyte cytokine interleukin-2 (IL-2) in combination
Many of these antibodies were utilized in early pilot, with OKT3 because of the potential for synergistic or
phase I, and phase I/II trials. additive T-cell stimulation. Sosman et al. treated 54
patients with doses of OKT3, ranging between 75 to
600 ug/m2 followed by high-dose bolus IL-2 therapy
CD3 [684]. The numbers of circulating T cells expressing the
CD3 is a pan-T cell antigen and an important receptor for IL-2 receptor were not increased, and the tumor response
activation of T lymphocytes. The first monoclonal anti- rate was no better than had been observed with IL-2
body approved for any use, was the murine antibody alone. Buter et al. gave 50 to 400 ug OKT3 with low-
OKT3 which now has the generic name muromonab-CD3 dose s.c. IL-2 to eight patients [76]. Neurotoxicity was
[638], approved to prevent kidney transplant rejection in the limiting toxicity at the highest dose. There was no
1984. Theoretically, CD3 might be a target as a tumor enhancement of activated lymphocytes and no responses.
associated antigen in T cell lymphoma, or to activate Another approach involves the production of antibodies
normal T cells to modulate or induce an indirect antitumor that are genetically fused to various cytokines to func-
action through other components of the immune system. tion as targeted biological response modifiers [388].
Robert O. Dillman 365

These multifunctional products are covered in other CD5


parts of this section based on the tumor antigen that is
being targeted. CD5 is a pan T cell antigen that is also co-expressed on
A humanized anti-CD3 antibody, initially called the B-lymphocytes of CLL and small B-cell lymphoma
HuM291, and now named visilizumab, has been studied (well-differentiated diffuse B-cell lymphoma), and the
in a variety of disorders to suppress activated T lympho- B-lymphoctyes of mantle cell lymphoma [188]. Normal
cytes, including ulcerative colitis, kidney rejection, and CD5+ B-cells are prevalent during fetal development, but
graft vs. host disease following allogeneic stem cell their number decrease after birth and with age, but increase
transplants [91, 367, 525, 563, 592]. Visilizumab is a again in the elderly. In the early years of Mab develop-
mutated IgG2 isotype whose Fc tail does not bind to Fc ment, several laboratories produced anti-CD5 antibodies
gamma-receptors, but can induce apoptosis of activated in response to immunization of mice with lymphocytes.
T lymphocytes. Doses from 0.1 to 15 mg/kg have been CD5 is expressed on most T cells. A number of the
infused in various trials, and infusion related toxicities early trials with mouse Mabs were conducted in patients
and CNS toxicities seem to be much less than were with T cell malignancies because of the early availabil-
observed with muromonab. To date there are no reports ity of antibodies that reacted with the CD-5 antigen on
of its use in cancer patients. T lymphocytes. Levy and colleagues treated eight T-cell
leukemia patients with the anti-CD5 momab Leu1, that
induces antigenic modulation, and two anti-leukemia
CD4 antibodies that did not induce antigenic modulation
CD4 is typically associated with helper T cells, and is [429]. Patients received 1 to 92 mg of Leu-1 alone or in
expressed on many T cell malignancies. Knox et al. combination with the other Mab. Transient reductions in
treated seven T-cell lymphoma patients with an anti- circulating leukemia cell counts were observed, but
CD4 chimeric Mab [397]. Doses of 10, 20, 40, and there were no sustained antitumor effects. There was no
80 mg were given i.v. twice a week for 3 consecutive obvious advantage to treating with a combination of
weeks. Circulating cells were coated with antibody, but antibodies in these trials.
there was no significant change in the number of T cells. Miller et al. reported an early success in a patient with
Some transient benefit was seen in all patients, but these CTCL who received 17 infusions of 1 to 20 mg of anti-
clinical results were not clearly better than those seen CD5 antibody Leu1 that was administered over a
with anti-CD5 murine Mab. 10-week period [485, 486]. He experienced a partial
Zanolimumab is a human IgG1 antibody against CD4 response including marked shrinkage of lymph nodes,
which may exert anti-tumor effects by several mecha- skin lesions, and a neck mass. There was also a transient
nisms [594]. Two are regulatory mechanisms that decrease in circulating T lymphocytes after each treat-
include rapid inhibition of signal transduction mediated ment. These same investigators treated five additional
by the CD4-associated tyrosine kinase p56lck, and then CTCL patients, and one with a large PTCL [488].
chronic down-regulation of CD4 molecules. The third is Individual patients received four to seven treatments
direct Fc-dependent lysis of circulating CD4+ T cells; over 2 to 10 weeks at doses of 250 ug to 100 mg infused
CD45RO+ cells are more sensitive to ADCC killing over 4–6 h for total doses of 13–761 mg. Four patients
than CD45RA+ cells. Zanolimumab is being evaluated had brief tumor regressions lasting from 1 to 4 months.
in cutaneous T-cell lymphoma (CTCL) and peripheral The IgG2a murine Mab T101, which reacts with the
T-cell lymphomas (PTCL) Kim et al. reported results of CD5 lymphocyte antigen, was one of the first mouse anti-
two phase two trials of zanolimumab in 37 patients with bodies to be extensively tested in human trials. The CD5
refractory CTCL, nine of whom were still in the myco- antigen rapidly internalizes after T101 binding, and the
sis fungoides (MF) stage of disease [387]. Patients antibody was not cytotoxic in in vitro assays of CMC or
received up to 4 months of weekly infusions with 280 to ADCC with human effector cells or human complement
980 mg per dose. The objective response rate was 32%. [181]. Dillman et al. treated two CTCL patients with mul-
Adverse events included infusion reactions with the tiple 2-h infusions of the anti-CD5 momab T101 [179],
initial depletion of peripheral T cells, and low-grade and subsequently treated an additional 10 CTCL patients
infections including eczematous dermatitis, especially with T-101 at doses from 10–500 mg, given over 24 h
in MF patients. Based on these encouraging results, [180]. With the prolonged infusions, brief antitumor
additional studies are being conducted in hopes of regu- responses lasting from days to weeks were seen in four
latory approval. patients. One of the most interesting responses was the
366 Monoclonal antibody therapy

dramatic remission of rheumatoid arthritis in a patient with one or more of the IgM anti-glycolipid momabs
with CTCL who was bedridden because of arthritis, and PMN-81, PMN-29, and PM-81 and a fourth antibody,
became ambulatory within 24 h of treatment with T101. In an IgG2b called AML-2-23, which reacts with a protein
other trials with T101, Foon et al. administered the anti- antigen [19]. None of these antibodies induced antigenic
body to several CTCL patients. Some patients had mini- modulation in vitro. Multiple 8 to 12 h infusions of 20 to
mal improvement in skin lesions [227]. Bertram et al. gave 70 mg of the various antibodies were associated with
T101 to eight patients with CTCL and to four other rapid, but transient decreases in circulating blasts and
patients with various T-cell lymphoproliferative disorders elevations of serum lactic dehydrogenase suggesting
[38]. One patient with CTCL had a partial response of his that leukemia cells had been destroyed, although sus-
cutaneous lesions that persisted for 3 months. Another tained remission of leukemia did not occur. Ball et al.
patient with convoluted T-cell lymphoma was alleged to treated another 16 acute myeloid leukemia (AML)
have had a complete remission based on a decrease in patients with 24-h continuous infusion of anti-CD20
bone marrow blasts from 6% to 3% which persisted for 2 Mab PM81 (also called MDX-11) at doses from 0.5 to
months following doses of 1–100 mg T101. 1.5 mg/kg in a dose escalation study [20]. Transient
Dillman et al. treated 10 CLL patients with T101 reductions in circulating blasts were noted, but there did
[177]. Two patients received 1 to 10 mg over 15 min, not appear to be an effect on marrow blasts at these rela-
two received weekly doses of 10 mg infused over 2 h, tively low doses. Circulating CD15 antigen presented a
and 6 received 24-h infusions of T101 at doses of 10, 50, problem in terms of immune complexes. The toxicities
100, 150, or 500 mg. Limited unsustained clinical ben- observed in this trial (fever, chills, hypotension, tachy-
efit was noted despite confirmation of in vivo binding to cardia) were the same as observed with other Mab that
CLL cells in blood, lymph nodes, and bone marrow. The bind to circulating cells.
same T101 antibody was studied by Foon et al. in 13
patients with CLL [228]. Successive three-patient
cohorts received doses of 1, 10, 50, 100 mg and one
CD19
patient received 140 mg. At all doses there binding to CD 19 is a 90 kD glycoprotein differentiation antigen
circulating and bone marrow cells could be demon- expressed on normal and malignant B cells that might be
strated, and a rapid but transient decline in circulating a useful regulatory target on B cells [753]. The CD19
leukemia cell counts. Durable responses were not seen. molecule acts as an internalizing receptor that is physi-
cally and functionally associated with certain protoonco-
gene phosphokinases. Hekman et al. treated 6 lymphoma
CD10 patients with an IgG2a murine Mab against the B-cell
CD10 was originally known as the common acute lym- CD19 [303]. Total doses from 225 to 1,000 mg were
phoblastic leukemia antigen (CALLA). CD10. Ritz and given over 4 h without major toxicity. One patient was
colleagues explored the mouse mab J-5 that reacts with felt to have had a partial remission that lasted 8 months
CD 10, in four patients with acute lymphocytic leukemia following his first treatment, and then 9 months follow-
(ALL) [600]. Doses of 1 to170 mg of J-5 were infused ing a second treatment. Because of the internalization
over 15 min to 2 h. In those patients with circulating (modulation) of this antigen, recent studies have focused
blasts, treatment was followed by a rapid decrease in cir- more on immunoconjugate approaches.
culating CALLA+ blasts, but a large number of CALLA-
negative lymphoblasts persisted. However, once J-5 was
no longer detectable in the serum, the lymphoblasts reex-
CD20
pressed CALLA, consistent with antigenic modulation or This is the target for the chimeric Mab rituximab, which
internalization of the surface antigen in the presence of was discussed in detail in this chapter in the section on
the antibody. There was no decrease in marrow blasts commercially available antibodies. It is also the target
even though J-5 binding was demonstrated. There was no of the commercially available radiolabeleled antibodies
significant toxicity noted in these patients although all I-131 tositumomab (Bexxar) and Y-90 ibritumomab
three patients who had circulating blast cells had tempera- tiuxetan (Zevalin) [174]. The earliest evidence of the
ture elevations in association with therapy. therapeutic potential of targeting CD20 was reported by
Press et al. [569] who used the anti-CD20 momab 1F5
to treat four patients with refractory B-cell lymphoma.
CD15 Total treatment was given over 5 to 10 days. Two
CD15 is a granulocyte-associated antigen. In prelimi- patients had a 90% reduction in circulating B cells. The
nary trials Ball et al. had treated three AML patients total doses delivered were 52 mg in a patient who had
Robert O. Dillman 367

progressive disease, 104 mg in a patient who had stable et al. conducted a phase I trial involving 46 patients who
disease, 1,032 mg in a patient who had a minor response, had relapsed or refractory CLL [84]. Lumiliximab was
and 2,380 mg in a patient who had a partial remission given i.v. at 125, 250, 375 mg/m2 or 500 mg/m2 weekly
including a 90% reduction in lymph nodes that persisted for 4 weeks or 500 mg/m2 thrice during week 1 then
for 6 weeks. 500 mg/m2 weekly for the next 3 weeks, or 500 mg/m2
This same antibody, which is now called tositu- thrice a week for 4 weeks. Little toxicity was reported at
momab, is given as part of I-131 radioimmunotherapy. any dose level, but there were no objective responses.
In one randomized trial that included a control arm in
which 36 patients received two 450 mg doses of tositu-
CD25
momab over 1 to 2 weeks, there was a 19% objective
response rate [150]. In a similarly designed randomized Daclizumab (Zenapax®, Roche Laboratories,
trial of Y-90 radioimmunotherapy, 4 weeks of standard Nutley, NJ)
dose rituximab had an objective response rate of 56% The interleukin-2 receptor, CD25, is overexpressed in
[790]. activated T-cells and many T cell malignancies. It is com-
Because of the tremendous clinical success of ritux- posed of 3 subunits including an alpha chain (p55), a beta
imab, a number of companies have made new CD20 chain (p75), and a gamma chain (p65) which combine
Mab with variations in Fv and Fc in efforts to enhance noncovalently to bind the important lymphokine interleu-
therapeutic benefit. It will be interesting to see how kin-2 (IL-2) which is not expressed on resting lympho-
many of the products are tested in randomized trials cytes, but is often overexpressed in activated and
against rituximab as opposed to going for a unique malignant lymphocytes, especially T-lymphocytes [762].
indication in a subset of CD20 positive malignancy. The anti-CD25 Mab daclizumab (Zenapax™, Roche
Laboratories, Nutley, NJ) was approved by the US FDA
in 1998 for the marketing indication of kidney transplant
CD22 rejection [766], but there is a strong rationale for it having
CD22 is a modulating antigen widely expressed on B clinical activity in certain hematopoietic malignancies
lymphocytes. Epratuzumab, a humanized IgG1 antibody [761, 764]. It is a humanized Mab derived by genetic
that targets CD22, was originally developed for radioim- engineering modification of a murine anti-Tac (55 kD
munotherapy, although preliminary work with anti- subunit of the IL-2 receptor, IL-2Rα) Mab. The human-
CD22 antibodies focused on immunoconjugates because ized form of this antibody was originally called anti-
of the internalization of the antigen after antibody bind- TAC-H. Later daclizumab was constructed using a human
ing. Leonard et al. treated 55 patients who had recurrent IgG1 constant framework [358]. The murine version was
indolent or large B-cell lymphoma with epratuzumab at unable to effect ADCC with human effector cells, but the
doses of 120 to 1,000 mg/m2 over 30 to 60 min weekly humanized construct was cytolytic in ADCC assays. As
for four treatments [425]. There was an 18% response expected, daclizumab was much less immunogenic in
rate among 51 evaluable patients with a median duration monkeys than the mouse Mab. Daclizumab was approved
of response or more than a year. All nine responses were as part of combination prophylaxis regimens for the pre-
in patients with follicular lymphoma. One complete vention of renal allograft rejection. These trials utilized
response persisted for more than a year in a lymphoma five weekly 1 mg/kg doses of daclizumab. Daclizumab
patient who was considered refractory to the anti CD20 was well-tolerated at that dose in kidney transplant
ximab rituximab. The treatment with this agent has been patients who were also receiving other immunosuppres-
well-tolerated with no surprising toxicities described. sive agents (cyclosporine, cyclosporine plus corticoster-
Subsequently clinical trials were launched in both folli- oids, or those two agents plus azathioprine). Because of
cular and large B cell lymphoma using four weekly doses its immunosuppressive effects, daclizumab is also being
of 360 mg/m2, but these were not completed. tested in other transplant settings for the treatment of graft
vs. host disease (GVHD) [572].
CD23
Daclizumab and T Cell Malignancy
CD23 has been characterized as a low-affinity IgE The expression of CD25 is increased in malignant cells
receptor that mediates allergic responses, but it is also of adult T-cell leukemia which is induced by the human
widely expressed on CLL cells and its expression is T-cell lymphotrophic virus I (HTLV-I) [272]. Since very
used to help differentiated CLL and SLL from mantle few normal cells express IL-2Rα, but abnormal T cells
cell lymphoma. Lumiliximab is an IgG1 anti-CD23 chi- in patients with lymphoid malignancies do, targeting the
meric Mab that is in clinical trials in CLL [587]. O’Brien IL-2Rα with daclizumab might be of therapeutic benefit.
368 Monoclonal antibody therapy

Waldmann et al. used the original murine anti-Tac peripheral leukemic cells, and schedules that sustained
(T-activated cells) Mab (predecessor to the humanized daclizumab in the blood, durable responses were not
daclizumab to treat 19 patients with adult T-cell leuke- obtained. There are other anti-CD25 Mabs being inves-
mia [765]. Patients received 20, 40, 50, 60, or 100 mg tigated, including the chimeric Mab basiliximab, that
doses over 8 to 445 days with variable total doses ranging could be investigated in lymphoid malignancy [352].
from 220 mg over 9 days, 490 mg over 51 days, and
220 mg over 445 days. The Mab was given as 3–11 infu-
sions per treatment course. One patient, who had previ-
CD30
ously failed aggressive combination chemotherapy had CD30 is an excellent target for immunotherapy of
a complete response that lasted for 5 months, and then a Hodgkin’s lymphoma (HL) because it is overexpressed on
partial response which lasted 6 months after retreatment. Hodgkin’s and Reed-Sternberg cells and the lymphocytes
There were two complete and four partial responses of some anaplastic lymphomas, but has limited expression
observed in this trial as well as one mixed response. The on normal tissue, which makes it a potential target for
duration of remissions ranged from 2 months to 3 years. Mab therapy [402]. Like many targets on hematologic
Another patient had a reduction in peripheral leukemia cells, it may not only be a target for CDC and ADCC, but
cells, but had an increase in malignant lymphadenopa- there is also evidence CD30 may be a regulatory target
thy at the same time. Despite the reactivity with circu- involved in signal transduction. There are a number of dif-
lating T cells, these patients reportedly had no significant ferent anti-CD30 products under investigation including
toxicity other than low-grade fever in two patients, and unmodified Mab, bispecific Mab in which CD30 is one of
hives in one patient. However, these patients were the targets, and various immunoconjugates. MDX-060 is
already markedly immunosuppressed, and they may a human IgG1 kappa anti-CD30 Mab that has exhibited
have been further immunosuppressed by the anti-T-cell anti-tumor effects in preclinical models. Ansell et al.
antibody. While on study, one patient developed treated 72 patients who had CD30+ tumors including 63
Kaposi’s sarcoma, one developed Pneumocystis carinii with Hodgkin’s lymphoma, seven with anaplastic large-
pneumonia, and a third developed Staphylococcal-A cell lymphoma, and two with CD30+ T-cell lymphoma
septicemia. Further evidence of immunosuppression [10]. Treatment was well-tolerated during treatment with
was the observation that only one of the nine patients escalating i.v. doses of 1, 5, 10, and 15 mg/kg, with only
treated developed HAMA. The CD25 IL-2 receptor is 7% of patients experienced grade 3 or 4 treatment-related
also sometimes expressed in Hodgkin’s disease; so, adverse events. Clinical responses were observed in six
antibodies such as daclizumab might have activity in patients (8.3%) and more than one third of the patients had
selected patients. a long duration of stable disease.
Few formal studies of daclizumab in T cell malignan- The bifunctional antibody HRS-3/A9 (BiMAb) targets
cies have been published, therefore most hematologists the Fc gamma receptor CD16 that is expressed on
prefer to use other products that target CD25, such as various effector cells including NK cells, and CD30
denileukin diftitox (Ontak), the fusion product of IL-2 antigen on Hodgkin’s cells. Fifteen patients with refrac-
and diptheria A chain as treatment for CD25 positive tory Hodgkin’s disease were treated with this antibody
malignancies [627]. CD25 is probably a better target for in a phase I/II trial [295]. The maximum tolerated dose
immunoconjugates therapy, such as immunotoxins, was 16 mg/m2 and the major side effects were fever,
because binding leads to internalization of CD25. rash and painful lymph nodes. One complete, one
Theoretically daclizumab therapy may only eliminate partial, one mixed and three minor responses were
circulating cells that express large numbers of CD25 noted. Nine patients developed HAMA. In a subsequent
molecules, since CDC and ADCC correlate with the trial 16 patients with refractory Hodgkins disease were
extent of antibody binding. This limitation is supported randomized to either 25 mg by continuous i.v. infusion
by the study by Koon et al. who treated 10 CD25+ leu- daily for 4 days, or 25 mg i.v. over 1 h every other day
kemia patients with daclizumab in a study focused on for four doses [296]. There was a 25% response rate
pharmacokinetics of the antibody and pharmacodynamics including one CR.
of interaction with the CD25 target. [403]. They confirmed Another bifunctional product consists of F(ab’) frag-
that high numbers of CD25+ circulating tumor cells ments derived from the murine anti-CD30 Mab Ki-4 and
were predictive of accelerated pharmacokinetics, and the humanized CD64-specific Mab H22. Ten patients
high levels of CD25 antigen expression correlated with were treated with escalating doses of 1, 2.5, 5, 10, and
the extent of target cell clearance. Despite clearance of 20 mg/m2 per day i.v. on days 1, 3, 5, and 7 [49]. All five
Robert O. Dillman 369

doses were well tolerated. Mild infusion reactions were had significant decreases in bone marrow blasts and one
observed and four patients had a partial response. was considered a CR.

CD33 CD38
M195 is a murine IgG2a monoclonal antibody that CD38 antigen is present on the majority of neoplastic
reacts with the CD-33 antigen that is expressed on early plasma cells and some CLL, and other B cell popula-
myeloid precursors. This receptor is rapidly modulated tions. Stevenson et al. developed a mouse/human chi-
or down regulated in the presence of M195. Scheinberg meric antibody to CD38 that consisted of human IgG1
et al. treated 10 AML patients using escalating doses of chemically linked to mouse Fab was able to mediate
M195 (1, 5, and 10 mg/m2) up to a total dose of 76 mg ADCC in laboratory studies [693] The same group has
[633]. Radioisotope tracer studies and bone marrow also developed a CDR-grafted humanized IgG1 from
biopsies demonstrated binding to bone marrow cells, the same murine antibody [206], but noted little differ-
but rapid antigenic modulation took place. Sustained ence between the two in various in vitro assays, including
antitumor effects were not seen. down regulation of the receptor. Clinical results with
Because of the theoretical limitations of HAMA, a these products have been disappointing [694].
CDR-grafted human IgG1 version of M195 (HuM195,
lintuzumab) was developed that is able to effect ADCC
with human effector cells in vitro [89]. Thirteen AML
CD40
patients received six i.v. doses over 3 weeks along with a Interaction with CD40 activates antigen-presenting cells
radiolabelled tracer dose. A decrease in marrow blasts and enhances immune responses, but targeting CD40 that
was only noted in one patient. Intermittent dosing with is expressed on solid tumors induces apoptosis. CP-870,893
3 mg/m2/day was found to saturate available binding sites is a fully human anti-CD40 Mab that was given to 29
[88]. In a subsequent trial 10 patients with refractory patients in a phase I trial [756]. Weekly doses from 0.01 to
AML were given higher doses of HuM195 as daily infu- 0.3 mg/kg were associated with grade 1 to 2 infusion reac-
sions of 12, 24, and 36 mg/m2 on days 1 to 4 and 15 to 18 tions that included fever, chills and rigors as well as brief
[90]. One patient had a complete remission that lasted decreases in lymphocytes, monocytes and platelets, but
more than 2.5 years. Mild infusion reactions including there was also evidence of B cell activation. Abnormalities
fever and rigors were noted. In a successor trial, 50 refrac- in D-dimer and liver function tests were noted 24 to 48 h
tory AML patients were given HuM195 as first salvage after infusion, and one patient experienced a deep venous
therapy in 24 patients and as second or subsequent sal- thrombosis. Despite these low doses and the cross reactiv-
vage therapy in 26 patients [217]. Patients were random- ity with circulating cells, four metastatic melanoma patients
ized to receive 12 or 36 mg/m2 on days 1 to 4 and 15 to 18. were judged to have had a PR (14%).
Three responses were recorded, all at the 12 mg/m2 dose.
Infusion-related fevers and chills were the predominant
toxicities, and were similar for the two doses.
CD45
Feldman et al. randomized 191 patients with primary The CD45 antigen is expressed on all hematopoietic
refractory or initial relapsed AML to combination che- cells. Anti-CD45 Mabs cause marrow suppressive
motherapy consisting of mitoxantrone, etoposide, and effects that range from leucopenia to marrow aplasia in
cytarabine (MEC) with or without lintuzumab. There some rodent models, so CD45 may be a useful therapeu-
was no significant difference in response rate, 36% for tic target for hematoproliferative malignancies. Krance
MEC plus lintuzumab vs. 28% for MEC alone (p = 0.28) et al. gave the rat anti-human CD45 MAbs, YTH25.4
nor in median survival. Mild antibody infusion-related and YTH54.12, which bind to different epitopes on
toxicities including fever, chills, and hypotension were CD45, to 14 patients with hematologic malignancies
common in the lintuzumab arm. who were scheduled to undergo stem cell transplantion
HuM195 was preceded by low-dose s.c. IL-2 in 13 [407]. Escalating doses were given; the maximum toler-
patients with relapsed or refractory, followed AML ated dose was 400 μg/kg/day for 4 days. Treatment was
[406]. After 5 weeks of s.c. IL-2 patients received i.v. followed by marked reduction in circulating lymphoid
infusions of HuM195 at a dose of 3 mg/m2 twice a week and myeloid cells, but had little impact on normal mar-
for 3 weeks. Infusion reactions that included nausea, row cells despite reducing the percentage of leukemic
rigors, and fever were frequently observed. Two patients blasts in two of three patients who a leukemic marrow.
370 Monoclonal antibody therapy

CD74 modulation of the CD122 IL-2/IL-15 beta receptor on


the surfaces of the TLGL cells, but there was no sustained
Milatuzumab (hLL1 or IMMU-115) is a humanized decrease in leukemia cell numbers. Clinical trials with
anti-CD74 monoclonal antibody [689]. CD74 is a surface the huMikbeta1 are planned. Higher doses of the human-
membrane protein that functions as a survival receptor ized Mab may be associated with more clinical activity,
in that it enhances cell proliferation and survival [685]. but perhaps also toxicity.
It also functions as an MHC class II chaperone. It is
expressed on lymphoid cells, especially malignant B
cells, and carcinomas of the kidney, lung, and stomach, HLA DR
and by certain sarcomas, but has limited expression on LYM-1 is an IgG2a murine Mab that reacts with a polymor-
other normal tissues. In vitro milatuzumab does not phic HLA-Dr antigen on all B cells, but the target antigen is
induce ADCC or CDC, probably because of the rapid not shed nor does it modulate in the presence of antibody.
internalization of this receptor molecule. However, Ten patients with refractory B cell lymphomas received
despite the rapid internalization of milatuzumab after weekly i.v. infusions of escalating doses of LYM-1 over 4
binding to CD74, preclinical models have suggested weeks without any objective tumor responses [330].
anti-tumor activity with unconjugated antibody, in addi- Apolizumab (hu1D10) is a humanized Mab against a
tion to immunoconjugates [688]. Unlike rituximab, polymorphic epitope on HLA DRbeta. Rech et al. treated
CD74 is also highly expressed on the malignant plasma six patients who had relapsed or refractory 1D10-positive
cells of multiple myeloma [73]. CD74 is also expressed non-Hodgkin’s lymphoma with granulocyte colony
on T cells and NK cells, and therapy is associated with stimulating factor (GCSF) and apolizumab at doses rang-
depletion of lymphocytes in primate models. The product ing from 0.15 to 1.5 mg/m2 [582]. There were no clear
is being explored in B cell lymphoma, multiple myeloma, anti-tumor effects. One patient had skin rash, one throm-
and CLL. bocytopenia, and one auto-immune hemolytic anemia.

CD80 HM1.24
Galiximab reacts with CD80, which is expressed on a
HM1.24 has been touted as a human plasma cell specific
variety of lymphoid and myeloid cells. Czuczman et al.
antigen that is over expressed on myeloma cells.
treated 37 patients suffering from relapsed or refractory B
Anti-HM1.24 inhibits proliferation of plasma cells that
cell lymphoma with four weekly i.v. infusions of galix-
overexpress HM1.24. A humanized IgG1/kappa anti-
imab at doses of 125, 250, 375, or 500 mg/m2 [142]. Four
HM1.24 (AHM) has been developed that exhibits strong
of 35 evaluable patients had an objective response (11%).
ADCC activity in vitro with human effector cells against
Treatment was well-tolerated with mild nausea, head-
myeloma KPMM2 and ARH77 human myeloma cells
ache, and fatigue the most common toxicities noted.
in the presence of human PBMCs myeloma cells [540].
It also shows promising activity in animal xenograft
CD122 models that is diminished by pretreating animals with
an anti-Fc gamma receptor II/III antibody [369].
CD122 is the beta-subunit shared by the IL-2 and IL-15
receptors [763]. Both IL-2 and IL-15 activate T cells,
but IL-2 also induces apoptosis of self-reactive T cells Anti CTLA4 (Cytotoxic T Lymphocyte
while IL-15 stimulates their persistence. Mikbeta1 and Antigen 4)
its humanized version (HuMikbeta1) bind to CD122.
In some cell models HuMikbeta1 is more effective in CTL-associated antigen 4 (CTLA-4) can inhibit T-cell acti-
blocking IL-2 stimulation then IL-15 stimulation. The vation and helps maintain peripheral self-tolerance of anti-
murine Mikbeta 1 was given to 12 cytopenic patients gens expressed on both normal and tumor cells. CTLA-4
with T cell large granular lymphocyte leukemia (T-LGL) is expressed on the surface of activated T-lymphocytes
in a phase I dose-escalation trial in which 0.1, 0.5 or where it suppresses the induction of immune responses
1.5 mg/kg i.v. doses were infused on days 1, 4, 7, and 10 that normally follow the interaction between T-cell recep-
[505]. At these doses there were no significant toxicities tors and HLA molecules on the antigen-presenting cells.
and HAMA was not detected, but such patients are Mouse experiments showed that blockade of CTLA4
already cytopenic and immunosuppressed. The investi- could yield anti-tumor effects. The two anti-CTLA 4 Mabs,
gators were able to demonstrate saturation and down- ipilimumab (MDX-010) and tremelimumab (CP-675, 206;
Robert O. Dillman 371

formerly known as ticilimumab), have been extensively mumab in 39 patients, including 34 with melanoma and
tested with promising activity [414]. However, both have four with renal cell cancer [591]. Among 29 melanoma
been associated with significant immune-related adverse patients who had measurable disease, there were two
events (IRAE) including dermatitis, inflammatory bowel durable CR and two durable PR lasting more than 2
disease, uveitis, arthritis, hypophysitis, and others. years. Dose-limiting toxicities were IRAE including
Generally, these IRAE have reversed after cessation of autoimmune phenomena such as diarrhea, dermatitis,
therapy and use of i.v. or topical corticosteroids. However, vitiligo, panhypopituitarism and hyperthyroidism. The
colectomy has been required in several severe cases of maximum tolerated dose for a single i.v. infusion was
inflammatory bowel disease, and several IRAE-associated 15 mg/kg. Among 30 melanoma patients who received
deaths have been reported [31]. Despite this toxicity pro- tremelimumab at a dose of 10 mg/kg monthly (n = 20) or
file, it is likely that one or both of these agents may receive 15 mg/kg every 3 months (n = 10) 4/12 patients with
regulatory approval. IRAE had an objective tumor responses compared to
only 1/18 without IRAE (p = 0.046) [590]. The IRAE
Ipilimumab and responses were associated with increased production
Striking anti-tumor activity has been seen in melanoma, of IL-2 and lack of these was associated with increased
but mostly in conjunction with substantial autoimmune production of IL-10. IRAE attributed dermatitis, colitis,
toxicity [133, 531]. Downey et al. reported a 17% hepatitis, and hypophysitis with panhypopituitarism. In a
response rate among 139 metastatic melanoma patients preliminary report on 20 patients, there was uveitis or
who were treated with ipilimumab, mostly in conjunction vision changes noted during extensive opthalamalogic
with peptide vaccines [190]. The details of the dose evaluations during and after treatment [696].
escalations used in these trials had been previously
reported [455, 558]. IRAE, including enterocolitis, Tumor Necrosis Factor-Related Apoptosis-
arthritis and uveitis, were documented in 62% of
patients and were highly correlated with a measurable
Inducing Ligand (Trail) Receptor
anti-tumor effect. The response rate was 36% among Tumor necrosis factor (TNR)-related apoptosis-inducing
24 patients who had colitis compared to 11% among ligand (TRAIL) is a TNF family member capable of induc-
113 patients who did not (p = 0.006) [31]. The three ing apoptosis by binding to its two death receptors (DR)
patients who experienced a CR had grade 4 toxicities. DR4 (TRAIL-R1) and DR5 (TRAIL-R2) [64]. There are at
Anti-tumor activity has been demonstrated in renal cell least two Mab being investigated that target the tumor
cancer. Yang et al. treated 61 metastatic renal cell patients necrosis factor-related apoptosis-inducing ligand (TRAIL)
with either 3 mg/kg followed by 1 mg/kg every 3 weeks (n receptors. Both are fully human Mabs. Mapatumumab
= 21) or 3 mg/kg every 3 weeks [800]. Partial responses (HGS-ETR1, TRM-1) targets DR4 (TRAIL-R1) [760].
were noted for 6/61 patients (10%), five at the higher dose. Lexatumumab (HGS-ETR2), targets DR2 (TRAIL-R2)
Grade 3 to 4 toxicity was documented in 33% of patients, [467]. TRAIL receptors are upregulated by chemotherapy.
most of which appeared to be auto-immune mediated, DR4 and DR5 may be useful targets for both hematologic
especially enteritis and endocrine deficiencies. The malignancies and solid tumors.
response rate was 35% among 17 patients who experi-
enced colitis and only 2% among 44 patients who did not Mapatumumab (HGS-ETR1, TRM-1)
(p = 0.002) [31]. Tolcher et al. treated 49 solid tumor patients with 158
A small number of prostate cancer patients have been courses of mapatumumab as i.v. doses ranging from
treated with ipilimumab [676]. Fourteen patients with 0.01 to 10 mg/kg infused over 30 to 120 min [724].
metastatic hormone-refractory prostate cancer received Initially mapatumumab was given as a single dose, then
ipilimumab at a dose of 3 mg/kg i.v. dose. Two patients every 28 days, and then 10 mg/kg every 14 days.
had a 50% decline in PSA level. Treatment was gener- TRAIL-R1 was expressed in 68% of patients whose
ally well tolerated and only one patient developed grade tumors were assayed. No objective tumor responses
3 dermatititis that required systemic corticosteroids. were seen. There was minimal toxicity with mild infu-
sion reactions being the most common adverse event
Tremelimumab (CP-675,206, Ticilimumab) reported, including fatigue, fever, and myalgia.
Striking anti-tumor activity has been seen in melanoma,
but mostly in conjunction with substantial autoimmune Lexatmumab (HGS-ETR2)
toxicity [85, 709]. Ribas et al. conducted a phase I dose Plummer et al. treated 37 solid tumor patients with 120
escalation trial of seven different dose levels of tremeli- cycles of lexatumumab at doses ranging from 0.1 to
372 Monoclonal antibody therapy

20 mg/kg every 21 days [564]. The maximum tolerated cells and antibodies blocking this integrin inhibit angio-
dose was 10 mg/kg based on dose-limiting toxicities genesis in preclinical models.
that included asymptomatic elevations of serum amy- Mullamitha et al. treated 24 patients with CNTO 905,
lase, transaminases, and bilirubin in three of seven a fully human Mab to anti-alphaV integrins that inhibits
patients treated with 20 mg/kg. There were no tumor angiogenesis and tumor growth in preclinical models
responses. [512]. CNTO 95 (0.1, 0.3, 1.0, 3.0, and 10.0 mg/kg) was
infused on days 0, 28, 35, and 42. CNTO 95 was gener-
ally well tolerated with only infusion-related fever being
Nuclear Factor-Kappa B Ligand the most common adverse event. At the 10.0.mg dose, a
Nuclear factor-kappa B (NF-ΚB) is a family of ubiqui- patient with cutaneous angiosarcoma had a 9-month
tous transcription factors that are believed to control partial response and a patient with an ovarian carcino-
inflammation. NF-KB normally resides in the cytoplasm sarcoma had a lesion that became negative by positron
of cells, but in response to a variety of stimuli, including emission tomography, although a stable lesion persisted
inflammatory cytokines, growth factors, carcinogens by computerized axial tomogragphy.
and ionizing radiation, NF-ΚB translocates to the McNeel et al. treated 25 solid tumor patients with
nucleus where it upregulates the expression of over 400 MEDI-522, a second generation humanized anti-alphaV-
different gene products associated with cell survival, beta3 antibody designed for antiangiogenic therapy, in a
proliferation, invasion, and angiogenesis [5, 194]. phase I study in which patients received doses ranging
Activation of NF-ΚB has been linked to oncogenesis from 2 to 10 mg/kg/wk [477]. Treatment was well toler-
and the biologic behavior of cancer cells, including ated and no dose-limiting toxicities were seen in this dose
resistance to chemotherapy and radiation therapy via range. The major adverse events observed were grade 1
transcription of anti-apoptotic proteins, leading to and 2 infusion-related reactions (fever, rigors, flushing,
increased proliferation of cells and tumour growth. and tachycardia), and low-grade constitutional and gas-
Activation of the NF-ΚB signal pathway follows the trointestinal symptoms (fatigue, myalgias, and nausea),
interaction of NF-ΚB ligand and receptor (RANKL). and asymptomatic hypophosphatemia. There were no
Denosumab is a fully human monoclonal antibody to objective responses observed, but three patients with
the receptor activator of nuclear factor-kappa B (NF-ΚB) metastatic renal cell cancer had prolonged stable disease
ligand [430]. Lipton et al. gave denosumab to 255 women Serial biopsies of skin showed detected MEDI-522 both
who had breast cancer-related bone metastases not previ- in quiescent and in angiogenically active skin blood
ously treated with i.v. bisphosphonates [439]. Denosumab vessels as well as in the dermal interstitial space [805].
was administered s.c. every 4 weeks (30, 120, or 180 mg) Posey et al. treated metastatic cancer patients with
or every 12 weeks (60 or 180 mg). Treatment was well vitaxin, a humanized monoclonal antibody, which has
tolerated and there was evidence of suppression of bone specificity for the integrin alphaV-beta 3 (vitronectin
resorption. Anti-cancer activity was not noted. receptor) which inhibits tumor cell mediated angiogen-
Body et al. conducted a small randomized, double- esis in pre-clinical animal models [565]. Vitaxin was
blind trial of single doses of s.c. denosumab (0.1, 0.3, given i.v. at doses of 10, 50 or 200 mg in cohorts of three
1.0, or 3.0 mg/kg) or i.v. pamidronate (90 mg), in 29 patients. There was no significant toxicity noted in these
patients with breast cancer and 25 with multiple three dose levels. There were no objective anti-tumor
myeloma [45]. Patients received a single dose of either responses. An every 3 week schedule of 200 mg appeared
agent and a placebo of the other. Treatment was well- to sustain serum levels.
tolerated. The magnitude of decrease in bone resorption
markers was similar to pamidronate, but the effects Human Epidermal Growth Factor Receptor
lasted longer with denosumab.
Antigens (EGFR1)
Cetuximab and panitumumab, the two FDA-approved
Integrins
products that target EGFR1, were discussed earlier in
Integrins are cell adhesion receptors that mediate inter- this chapter. A number of other Mabs have been devel-
cellular communication. Integrins are implicated in oped that target this receptor. Mab ch806 is a chimeric
tumor cell survival, angiogenesis, cancer invasiveness, antibody that detects a unique epitope on EGFR that is
and metastasis, and therefore are a potential target of exposed only on mutanted or ligand-activated forms of
cancer therapy [520]. The integrin alphaV-beta3 is an the receptor on cancer cells in which it is overexpressed
adhesion molecule expressed by proliferating endothelial [648, 649]. In preclinical studies murine 806 blocked
Robert O. Dillman 373

EGFR-mediated signal transduction resulting in anti- but at this dose dermatologic toxicities were common,
tumor effects on human tumor xenografts, while in vivo including acne, papillary rash, dry skin, and paronychia,
studies in human showed tumor specific targeting of but also headache, fatigue, and diarrhea, and case of pan-
ch806. ABX-EGF is a fully human anti-EGFR Mab that creatitis that was possibly treatment related. There were
was given to 88 patients with metastatic renal cell can- no objective tumor responses. Matuzumab was given at
cer at doses of 1.0, 1.5, 2.0, or 2.5 mg/kg weekly [612]. weekly doses of matuzumab (100, 200, 400 or 800 mg)
Three patients had an objective response and the majority concurrently with paclitaxel at 175 mg/m2 every 3 weeks
of patients had stable disease for at least 2 months. A dose- in 18 patients with NSCLC [401]. No additive toxicity was
related acneiform rash was the most common toxicity, noted even at the 800 mg dose. Low grade acneiform
and occurred 100% of patients who received at least skin rash occurred in 14 patients. There were five objec-
three doses of ABX-EGF at 2.5 mg/kg/wk. tive tumor responses. Escalating doses of matuzumab
The humanized anti-EGFR Mab h-R3 was given to (400 mg weekly, 800 mg biweekly, or 800 mg weekly)
29 patients with newly diagnosed malignant gliomas were given with gemcitabine to three groups of patients
including 16 glioblastoma, 12 anaplastic astrocytoma and with advanced pancreatic adenocarcinoma [270]. Skin
one anaplastic oligodendroglioma (AO) [580]. Following toxicity was the most frequent side effect. There were
debulking surgery, patients received six 200 mg infu- three partial responses in patients who received 800 mg
sions of h-R3 weekly in during radiotherapy. Interestingly, of Mab weekly.
no patients developed acneiform rash, and there were no A bispecific antibody called MDX-447 was constructed
allergic reactions. Median survival was 17months for by cross-linking antigen binding F(ab’) fragments) of a
the glioblastoma patients. Mab h-R3 was given at four humanized Fab anti-EGFR to a humanized Fab which
dose levels weekly for 6 weeks in combination with binds to CD64, the high-affinity immunoglobulin G
radiotherapy, to 24 patients with advanced head and receptor known as Fc gamma receptor I. Escalating
neck cancer [135]. In this trial there wee some infusion weekly doses from 1 to 15 mg/m2 were administered to
reactions, but no dermatologic or allergic toxicities 36 patients with various refractory EGFR-positive can-
were noted. The overall survival of patients was consid- cers including cancers of the kidney, bladder, prostate,
ered encouraging. head and neck, and skin bladder, ovarian, prostate cancer
HuMax-EGFr is a fully human Mab that was given in and skin cancer, with and without G-CSF [138]. MDX-
escalating doses of up to 8 mg/kg to 28 patients with 447 infusions were associated with decreases in mono-
squamous cell cancer of the head and neck [28]. Most cytes and increases in plasma cytokines in association
frequently reported adverse event was rash. There were with fever, chills, hypotension, and myalgias. Fury et al.
seven responses noted among 11 patients who were treated reported on 41 patients treated with MDX-447 at doses
at the higher doses. from 1 to 40 mg/m2 and 23 patients who received 1 to
Matuzumab is a humanized anti-EGFR Mab formerly 15 mg/m2 of MDX-447 in combination with GCSF [242].
known as EMD 72000 that has been explored in patients Hypotension was the major dose-limiting toxicity in both
with a variety of cancers. Nine patients with squamous cohorts, and accrual to MDX-447 plus GCSF was stopped
cell cancer of the head and neck were treated in a phase because of excessive toxicity in association with higher
I trial in which two doses were given before surgery and levels of TNF-α and IL-6. The maximum tolerated dose
three doses after surgery [41]. Successive patient groups for MDX-447 alone was 30 mg/m2 i.v. weekly. There were
received a single dose of 100, 200 or 400 followed by no objective responses.
four weekly doses of 50, 100, and 200 mg, respectively.
Treatment was well-tolerated. The most frequent adverse Human Epidermal Growth Factor Receptor
events attributed to the antibody were fever and a tran-
sient elevation of liver enzymes, but there was no significant
Antigens (EGFR2)
skin toxicity. In another dose-escalation trial 22 patients Trastuzumab, the only FDA-approved product that tar-
with various solid tumors received matuzumab at five gets EGFR2, was discussed earlier in this chapter. Other
different dose levels randing from 400 to 2,000 mg/wk antibodies have been developed that also target HER3.
[744]. The maximum-tolerated dose was 1,600 mg/wk Pertuzumab (rhuMAb 2C4) is a humanized construct
because of severe headaches at the 2,000 mg dose. of the murine 2C4 Mab that binds to the HER-2 receptor
At these dose levels mild acneiform rash was the most and blocks its ability to dimerize with other HER recep-
common toxicity. Matuzumab was given to 37 women tors. In a phase I trial pertuzumab was given at doses of
with intra-abdominal platinum-resistant ovarian cancer at (0.5 to 15 mg/kg) i.v. every 3 weeks to 21 patients with
doses of 1,600 mg/wk [656]. Therapy was well tolerated, various advanced solid malignancies [3]. Treatment was
374 Monoclonal antibody therapy

well-tolerated. Two objective responses were recorded. IL-6, TNF-α, and IFN-γ were seen in association with the
Patients with relapsed ovarian cancer were treated with infusion reactions. Objective tumor responses were seen
pertuzumab, including 61who received a loading dose in 5 patients.
of 840 mg followed by 420 mg every 3 weeks, and 62 The bispecific antibody MDX-H210 was constructed
patients who received 1,050 mg every 3 weeks [266]. by cross-linking antigen binding F(ab’) fragments of
Pertuzumab was well tolerated, but diarrhea occurred in Mab H22 to CD64, the high-affinity immunoglobulin G
69% (11% grade 3, no grade 4). Five patients had receptor FcgammaRI) and mAb 520C9 to HER2 [770].
asymptomatic left ventricular ejection fraction decreases In a dose escalation trial of single i.v. infusions in
to less than 50%. There were five PRs. Herbst et al. gave patients with breast and ovarian cancer, one partial
pertuzumab once every 3 weeks to 43 previously treated response was observed in a patient with breast cancer
NSCLC patients [311]. Treatment was well-tolerated, [740]. Elevated plasma levels of TNF-α, IL-6, G-CSF
but there were no objective responses. Agus et al. gave and neopterin were detected at higher doses of the anti-
pertuzumab as a loading dose of 840 mg followed by body. Seven patients with HER2-positive prostate can-
420 mg for subsequent cycles, to 41 patients with hor- cer received 1 to 8 mg/m2 of MDX-H210 as 2-h infusions
mone refractory prostate cancer who had also experi- three times a week [642]. Over this dose range MDX-
enced disease progression after taxane chemotherapy H210 was well tolerated other than mild infusion-related
[4]. Pertuzumab was well tolerated; grade 1 to 3 diar- fever, chills, myalgias and malaise, in association with
rhea was the most common toxicity. There were no increased plasma levels of TNF-α and IL-6. No dose-
tumor responses. A trial using the same doses of pertu- limiting toxicicty was noted. In another study, 25
zumab was conducted in 35 patients with hormone patients with HER2+ advanced prostate cancer were
refractory prostate cancer who had not been treated with treated with s.c. GM-CSF at a dose of 5 μg/m2 for 4 days
chemotherapy, and another 33 were treated with a fixed of each week along with MDX-H210 at 15 μg/m2 weekly
dose of 1,050 mg [153]. Treatment was well tolerated, for 6 weeks [346]. Toxicities leading to withdrawal from
but there were no tumor responses. Attard et al. com- the trial included grade 3 heart failure, dyspnea and
bined pertuzumab (1,050 mg fixed dose and later the allergic reactions, but 35% of patients had at least a 50%
840 mg loading dose, then 420 mg) with docetaxel che- decrease in serum PSA levels.
motherapy with both agents given together every 3
weeks [13]. Based on toxicity they recommended further
evaluation with docetaxel 75 mg m2 and 420 mg pertu-
Insulin-Like Growth Factor Receptor
zumab following a loading dose of 840 mg. The anti insulin-like growth factor-I receptor Mab
Ertumaxomab (2B1) is a bispecific murine monoclonal CP-751,871 was given in doses up to 20 mg every 3
antibody that binds to the extracellular domains of HER2/ weeks to 24 patients with various malignancies [290].
neu and FcgammaRIII. Twenty women with metastatic A maximum tolerated dose was not identified over this
breast cancer, all but one of whom had received prior dose range. Mild adverse events included hyperglycemia,
chemotherapy were treated in a phase I trial [50]. The hyperuricemia, elevated hepatic enzymes, anorexia,
initial dose of 2.5 mg/m/day was associated with dose- nausea, diarrhea, and fatigue. There were no objective
limiting toxicities in three of the first eight; so the remaining responses.
12 patients received 1 mg/m/day. Objective antitumor
responses were not seen. A trifunctional version of this
antibody which also binds to CD3 on T lymphocytes was
Transferrin Receptor
tested at doses of 10–200 μg weekly for up to three doses The murine Mab 42/6 reacts with the human transferrin
in 17 patients with various malignancies [384]. The maxi- receptor thereby blocking the iron transport protein
mum tolerated dose was 100 μg because of a systemic transferrin, which is crucial for iron transport by cells
inflammatory response syndrome and acute renal failure [732]. In vitro studies showed that 42/6 had inhibitory
in a patient treated with 150 μg and severe hypotension effects on a various types of cancer cells [704], [705]. In
and respiratory distress syndrome in one patient and a phase I trial 33 treatments at doses of 2.5 to 300 mg/m2
exacerbation of heart failure in two patients who received were given as 24-h infusions to 27 patients [57]. 42/6
200 μg. The most common drug-related adverse events was well tolerated, although one patient experienced an
were mild, transient infusion reactions including fever, allergic-type response associated with a HAMA
rigors, headache, nausea, and vomiting, but grade 3 to 4 response during a second course of treatment. Limited
lymphocytopenia occurred in 76% of patients and ele- mixed tumor responses were observed in three patients
vated hepatic enzymes in 47%. Elevated levels of IL-2, with hematological malignancies, but there were no
Robert O. Dillman 375

partial or complete remissions. HAMA was detected cross-reactivity with granulocytes, and associated with
33% of patients. transient decreases in total leukocyte counts [178].
There were no tumor responses. Other trials utilizing
small dose of murine anti-CEA Mab that did not cross-
Vascular Ligands and Receptors react with granulocytes were associated with a mini-
Bevacizumab, which reacts with the VEGF ligand, is the mum of side effects, and were taken up into tumors, but
only FDA-approved product in this group. Bevacizumab not associated with tumor responses [451], [30], [288].
was reviewed in detail earlier in this chapter. Several In December 1992 the 111-indium conjugate of a
other Mab products that target VEGF or VEGFR are murine Mab B-72.3 (OncoScint™) became the first Mab
under development. approved specifically for use in cancer patients, albeit as
IMC-1C11 is a chimeric Mab that has anti-angiogenesis an imaging agent for patients with ovarian and colon
properties based on its binding to the VEGF receptor 2 cancer rather than a therapeutic agent. approved for clin-
rather than the VEGF ligand. In a dose escalation study, ical use for radioimmunodetection of cancer in patients
14 patients with colorectal carcinoma and hepatic metas- with ovarian cancer and colorectal cancer in Numerous
tases received weekly doses at 0.2, 0.6, 2.0, or 4.0 mg/ patients received doses of 10 to 40 mg of antibody along
kg for 4 weeks [566]. There were no serious toxicities, with 1 mg doses of radiolabelled antibody [454]. Tumor
although minor bleeding occurred in four patients regressions were not noted in the trials in patients with
treated at the two lowest dose levels. HACA was known colorectal cancer. However, humanized forms of
detected in half the patients and altered pharmacokinetics this antibody appeared promising based on in vitro assays
of IMC-1C11 in two patients. There were no objective of ADCC. Khazaeli et al. treated 12 patients with 3 to
tumor responses. 7 mg of B-72.3 chimeric IgG4 antibody [381]. No toxicities
HuMV833 is a humanized IgG4 anti-VEGF Mab that or tumor effects were seen at these low doses.
was given to 20 patients who had advanced solid tumors Haisma et al. treated 20 colorectal cancer patients
by i.v. infusion on days 1, 15, 22, and 29 [348]. Treatment with the human IgM Mab 16.88, labeled with 131iodine
was well-tolerated over the dose range of 0.1 to 10 mg/ for tumor localization [282]. Patients received an 8 mg
kg per dose. One patient with ovarian cancer experi- dose followed 1 week later by 200, 500, or 1,000 mg of
enced a partial response. Dose of 1 and 3 mg/kg were antibody. Tumor uptake was seen in at least one tumor
selected for further investigation. site in 80% of the patients, but no tumor regressions
J591 targets the external domain of prostate-specific were reported.
membrane antigen (PSMA), but is also expressed on the
neovasculature of various solid tumors. J591 was given
every 3 weeks in doses of 5, 10, 20, 40, 60, or 100 mg,
Epithelial Cell Adhesion Molecule (EpCAM)
along with a 2 mg tracer dose of Mab conjugated to One of the targets for many murine Mab produced in
10 mCi of indium-111 to 20 patients with a variety of different laboratories by immunizations with epithelial
malignancies [507]. In a second trial 27 patients received cancer cells was a surface glycoprotein called 17-1A,
doses of 5, 10, 20, 40, or 80 mg and some patients GA733-2, or KSA. This molecule was expressed on
received weekly therapy for up to 6 weeks [490]. Tumor most malignant epithelial cells and some normal human
localization was confirmed in both studies and there cells. It was subsequently recognized that this antigen
were no dose limiting adverse events. No tumor responses is what is now called epithelial cell adhesion molecule
were observed in either study. (EpCAM) [820]. Loss of membranous EpCAM is associ-
ated with nuclear beta-catenin localization, reduced
cell-cell adhesion, and increased migratory potential of
Carcinoembryonic Antigen (CEA) cells, which suggests that blocking this molecule could
Numerous clinical studies with antibodies directed be associated with anti-cancer effects.
against carcinoembryonic antigen (CEA) and other anti-
gens of gastrointestinal mucosa have been reported. 10.6.2.27.1
Several different murine IgG1 anti-CEA antibodies at Edrecolomab (17-1A, Panorex®), was the most heavily
doses from 1 to 80 mg were infused into 30 colorectal studied anti-EpCAM Mab [268]. This was actually the
cancer patients utilizing single 1- to 2-h infusions with first therapeutic Mab approved for the treatment of
tracer doses of 111-Indium labeled antibody [183]. With cancer internationally, but it never received approval in
one such Mab three patients had dramatic infusion reac- the U.S. Edrecolomab is a murine IgG2a that reacts with
tions with fever and rigors which turned out to be due to a 37–40 kD glycoprotein found on various adenocarci-
376 Monoclonal antibody therapy

nomas and on normal epithelial tissues [315]. The at 3-week intervals for 2 treatment courses. There were
EpCAM antigen is not shed into the circulation nor is it no responses among seven patients who received 200 to
internalized. Edrecolomab may produce anti-cancer 400 mg every other day up to a total dose of 4.8 to 7.6 g.
effects by various mechanisms [404], [316]. It was one There were no responses among six patients who
of the few murine Mabs that could effect ADCC with received 500 mg 3 days a week, to a total dose of 12 g.
human mononuclear cells [312]. Edrecolomab induced Enthusiasm for edrecolomab as a potentially useful
a high frequency of HAMA in humans [651], [653]. anti-cancer agent peaked after publication of the results
Some of these HAMA (Ab2) react with the idiotype of of a multicenter randomized German trial of adjuvant
the 17-1A mouse antibody (Ab1). As part of the idiotype therapy that was conducted during 1990–1992 in
network control of B-cell proliferation, the Ab2 induces patients with resected Dukes C colorectal cancer [822].
additional antibodies against itself, including antibodies There were 90 patients randomized to observation,
(Ab3) that react with the idiotype of the Ab2. Such an and 99 to a post-operative regimen of edrecolomab
Ab3 is actually an endogenous human antibody with the 500 mg i.v., then 100 mg i.v. once a month for 4 months.
same reactivity against the tumor antigen as Ab1. This Mild side effects were observed in association with the
phenomenon has been observed in patients who have infusions, including fever, chills, abdominal pain,
received edrecolomab [316], [215]. Single injections of nausea, and diarrhea, but there were four episodes of
edrecolomab at doses of 15–1,000 mg were well toler- anaphylaxis during 371 infusions (1.1%). After a
ated, but 50% of patients developed HAMA after a median follow up of 5 years, there was a statistically
single injection [651], [653]. Uptake of 17-1A into significant difference in disease-free and overall sur-
tumors was demonstrated by radiolabeled antibodies vival curves with a 30% reduction in death rate and
and by immunohistochemistry [666, 496]. There were 27% reduction in tumor recurrence in the 17-1A group.
no objective responses among 40 patients who received The magnitude of improvements in key endpoints were
single infusions of 15–1,000 mg of 17-1A during phase similar to those observed in U.S. trials of 5-fluououra-
I trials in patients with various gastrointestinal adeno- cil and levamisole that had led to acceptance of 5-FU
carcinomas [651], [653]. In a phase II trial of 20 patients based chemotherapy in the adjuvant treatment of colon
who received 200–850 mg of 17-1A, one response was cancer. After a median follow up of 7 years, the appar-
claimed in a patient with recurrent rectal cancer [240]. ent benefits of edrecolomab persisted with a 23%
In a trial of 25 patients who received one to four doses reduction in recurrence and 32% reduction of death in
of 400 mg of 17-1A, one patient was interpreted as hav- the antibody group [595]. On the basis of this trial,
ing a complete response [443]. Eleven patients experi- Edrecolomab was approved for the treatment of Dukes
enced mild gastrointestinal toxicity during treatment. C colorectal cancer in Germany in 1995.
Subsequently, this group treated eight patients with a These promising results were followed by four addi-
17-1A chimeric antibody and confirmed the longer tional phase III trials in the United States and Europe.
serum half-life and reduced rate of HAMA, but there Trials in North America compared 5-FU + levamisole, or
were no responders even though the IgG4 human subclass 5-FU + leucovorin to the same agents plus edrecolomab in
heavy chain enhanced ADCC [444]. Dukes C colon cancer using the same dose and schedule
Additional trials with 17-1A were conducted in used in the German trial. The European trials involve
patients with colorectal carcinoma by Mellstedt and col- comparisons among 17-1A antibody alone, 5-FU + leuco-
leagues who reported one objective response among 52 vorin, and 5-FU + leucovorin + 17-1A. Edrecolomab was
patients who were treated in various ways [240], [821]. also studied in combination with chemotherapy and radio-
There were no responses among 10 patients who therapy in Dukes B and C rectal cancer. Results of the
received a single 200 to 400 mg dose of 17-1A every large randomized European adjuvant trial did not demon-
4–6 weeks. There was one minor response among 10 strate a benefit for the use of edrecolomab as adjuvant
patients who received 200 to 400 mg of 17-1A preceded therapy [574]. There were 2,761 patients randomized in
by 400 mg/m2 of the chemotherapy agent cyclophosph- the three-arm trial. The major toxicities observed in the
amide, which was given in an effort decrease the edrecolomab-alone arm were diarrhea in 32%, although
frequency of HAMA. There was one CR and two minor severe or life-threatening diarrhea was noted in only 2%.
responses among 14 patients who received the same Various types of hypersensitivity reactions to edrecolomab
dose/schedule of Mab preincubated with autologous were documented in 25% of patients. Results in the
peripheral blood mononuclear cells. There were no edrecolomab-alone arm were inferior to the 5FU-containing
responses in five patients who received a total dose of arms (p < 0.05), and there was no evidence of better results
3.6 g of 17-1A given as 400 mg daily on days 1, 3 and 6, when the antibody was combined with 5FU (p = 0.53).
Robert O. Dillman 377

Three-year survival rates were 76% for 5FU/leuovorin, subsequent trial 20 patients with advanced colorectal
75% for the antibody/chemotherapy combination, and cancer received edrecolomab 400 mg at day 3 of a
70% for the antibody alone. The differences in 3-year 10-day treatment cycle with the simultaneous adminis-
event free survival also revealed inferior results for anti- tration of s.c 250 μg/m2 GM-CSF once daily and 2.4
body alone, and no advantage to the chemotherapy plus million U/m2 IL-2 twice daily for 10 days in a planned
antibody combination. monthly treatment cycle [321]. Doses of the various
In another trial 377 patients with stage 2 colon cancer, drugs had to be decreased because of infusion/allergic
stratified according by whether the primary tumor was type reactions. There was one PR. In a trial of similar
T3 or T4, were randomized post-operatively to treat- design, 12 colorectal cancer patients were treated with
ment with 900 mg edrecolomab or observation [297]. 300 μg GM-CSF and 6 million units IL-2 s.c. daily from
The study was stopped because of discontinuation of day 1 to 10 with 400 mg edrecolomab on day 3 of the
drug supply in Germany after negative results from first cycle, and reduced to150 mg on subsequent cycles
other trials became public. After a median follow-up of up to a maximum of four cycles [221]. There were no
3.5 years there was no difference in overall survival or tumor responses. Edrecolomab was combined with
disease-free survival. chemotherapy, GM-CSF, and IFN-α for 27 patients
Several trials have explored edrecolomab in combina- with metastatic colorectal cancer in one trial [434],
tion with other biologicals with or without chemotherapy. and with capecitabine alone in 27 patients with various
Edrecolomab was combined with IFN-γ in an effort to metastatic adenocarcinomas [457]. It was unclear whether
increase Fc receptors on effector cells and increase the the clinical activity seen was any better than what might
expression of the antigen. Weiner et al. gave 150 mg of have been expected with the same chemotherapy alone.
17-1A on days 2 to 4 in combination with 1.0 MIU/m2 of
IFN-γ on days 1 to 4, to 19 colorectal cancer patients, but Other anti EpCAM antibodies
no antitumor responses were noted [774]. In a second trial Elias et al. treated NSCLC patients with the IgG1
27 patients with colon or pancreatic cancer were given momab KS1/4 that reacts EpCAM [205]. Five patients
IFN-γ at doses up to 40 MIU/day for 4 days followed by received sequential doses of 1, 10, 60, 100, and 1,000 mg
400 mg 17-1A on day 5 [773]. No objective tumor over 2 weeks for a total of 1,661 mg. Minor upper
responses were not seen and the authors concluded that gastrointestinal toxicity was seen in some patients. No
the low dose of IFN-γ was as effective as higher doses.. antitumor responses were seen.
Saleh et al. treated 15 colorectal cancer patients with The anti-EpCAM human-engineered Mab ING-1 was
0.1 mg/m2 IFN-γ on days 1 to 15 and 400 mg of 17-1A at a given to 25 advanced adenocarcinoma patients at doses
dose of 400 mg on days 5, 7, 9 and 22 [622]. No significant of 0.03 mg/kg, 0.10 mg/kg, and 0.30 mg/kg [151]. Two
objective tumor responses were described. patients experienced reversible, but dose limiting pan-
Weiner et al. conducted a phase II multicenter trial of creatitis; so, 0.10 mg/kg was felt to be the maximum
edrecolomab in patients with unresectable pancreatic tolerated dose. There were no responses observed in the
carcinoma [776]. A dose of 500 mg was given i.v. thrice first 25 patients, or seven additional patients who were
weekly for 8 weeks. Because of rapid progression of treated with repeated weekly doses of 0.10 mg/kg.
disease in several patients, only 16/28 patients received Adecatumumab (MT201) is a human recombinant
the planned total dose of 12 grams. There was one dura- IgG1 Mab that binds EpCAM. Twenty patients with
ble PR. Tempero et al. treated 30 patients who had hormone refractory prostate cancer were treated with two
advanced, measurable pancreatic cancer with IFN-γ at a adecatumumab infusions on days 0 and 14 in over a dose
dose of 1 MIU/m2 daily for 4 days and 150 mg of 17-1A range of 10 to 262 mg/m2 [528]. Mild infusion reactions
admixed with autologous leukocytes on days 2, 3 and 4 including fever and nausea were the most common
[713]. One patient had a CR that persisted for 4 months. adverse events. No tumor responses were reported.
The median survival for this group of patients was only Catumaxomab is a trifunctional Mab that binds
5 months. There was no evidence of increased HLA-DR human EpCAM and CD3. Escalating doses of catumax-
expression on monocytes or lymphocytes following the omab from 2 to 7.5 μg were infused over 8 h in patients
administration of IFN-γ. with advanced NSCLC who were premedicated with
GM-CSF and IL-2 have also been combined with corticosteroids and antihistamines to reduce infusion
edrecolomab in an effort to enhance the immune reactions [654]. Hepatic transaminasemia was the dose-
response. In a trial of 20 patients who received 17-1A limiting toxicity. The MTD was determined to be 40 mg
plus GM-CSF to increase effector cells and the expres- of dexamethasone followed by 5 μg of catumaxomab.
sion of Fc receptors, there were two CRs [575]. In a No tumor responses were reported.
378 Monoclonal antibody therapy

SK-1 is a human Mab generated using hybridoma reported. A bifunctional momab variant of MOv18
fusion methods with lymphocytes from regional lymph called OC/TR combines the anti-ovarian binding of the
nodes of colon cancer patients. that recognizes a glyco- MOv18 with anti-CD3 to target T lymphocytes. The
protein expressed on the majority of colon cancer tissues product was given as an F(ab’)2 in an effort to avoid sys-
that appears to be in the EpCAM family of molecules temic toxicity related to removal of T cells in the reticu-
[398], [801]. In the initial dose escalation study colon loendothelial system [721]. However, significant
cancer patients received 2, 4, or 10 mg given three times. infusion reactions still occurred except at the lowest i.v.
Treatment was well-tolerated; no tumor response were doses among five patients, suggesting that the binding to
reported [399]. CD3 itself led to the acute release of cytokines that
mediated the side effects. This Mab has also been given
i.p. as one or two 5-day cycles with IL-2 to 28 patients
CA125 who had residual limited disease after surgical debulk-
Oregovomab (B43.13, OvaRex) is a murine Mab that ing and chemotherapy [86]. Responses were confirmed
targets CA125 [32]. Because CA125 is shed into the cir- in seven patients based on surgical restaging, although
culation, infusion of oregovomab is associated with one had concurrent progression in retroperitoneal nodes.
immune complexes which probably would complicate a Most of the responders subsequently progressed outside
direct therapeutic approach. However, in patients who the peritoneal cavity, but three of the CRs lasted 18 to 24
received low doses of oregovmab, it was possible to months. HAMA was detected in 21/25 patients. Elevated
demonstrate antibody and T cell responses to CA125 plasma levels of TNF-α, IL-6, G-CSF and neopterin
that were not present before injection of the antibody; so were detected following infusions of higher doses of
it has been hypothesized that these immune complexes Mov18. A chimeric version of this Mab has been devel-
are taken up by dendritic cells leading to an anti-CA125 oped and given to small numbers of patients [748]. An
immune response that may be therapeutic [526]. In a IgE construct has also been engineered [366].
phase II pilot study 13 heavily pretreated, platinum-re-
sistant ovarian cancer patients CA125 > 35 U/mL, were Other Anti-Adenocarcinoma
given 2 mg of oregovomab i.v. at weeks 0, 2, 4, 8, and
12, followed by additional dose every 3 months for up
Monoclonal Antibodies
to 2 years [200]. More than half the patients exhibited L6
enhanced anti-CA125 immune responses, four patients L6 is a mouse Mab that reacts with a 24 kD epithelial
showed decreases in CA125 levels, and three patients antigen with expression on most adenocarcinomas. It
had progression-free disease for more than 2 years. In a mediates CMC with human complement and ADCC
phase III trial 145 patients with stage 3 or 4 ovarian can- with human NK cells and macrophages. The L6 antigen
cer, who had achieved a complete response following is now believed to be a distant member of the transmem-
other therapy, were randomized to oregovomab or brane-4 superfamily (TM4SF). Goodman et al. treated
placebo administered at weeks 0, 4, and 8, and then five metastatic breast cancer patients with daily doses
every 12 weeks for up to 2 years [33]. There was no dif- from 5 to 400 mg/m2 of murine L6 for 7 days to a total
ference between the two arms in time to recurrence, dose of 35 to 2,800 mg [262]. One patient with hormone
13.3 months for oregovomab and 10.3 months for pla- receptor negative cancer, which had progressed on the
cebo (p = 0.71). chest wall despite chemotherapy and radiation therapy,
Abagovomab (formerly ACA125) is a murine anti- had a CR after receiving a 400 mg dose. The response
idiotypic Mab antibody to a Mab that reacts with CA125 did not become apparent until about 5 weeks after treat-
(Ab2). It is being explored as a vaccine in ovarian can- ment, and it took about 14 weeks before the CR was
cer and appeared promising in phase I trials based on the attained. Doses from Doses from 5 to 400 mg/m2 were
induction of a human anti-CA125 antibody (Ab3) given daily for 7 days to treat nine patients with advanced
response [554, 616]. This approach is discussed in more ovarian cancer [262]. No responses were seen. No evi-
detail in the vaccine chapter of this text. dence of anti-tumor effects in three patients with NSCLC
MOv18 is a chimeric Mab that reacts with a folate who received L6 [262]. Among five colorectal cancer
receptor on ovarian cancer cells. In a phase I trial 15 patients, one patient had a partial response following
ovarian cancer patients received single i.v. doses rang- seven daily 2-h infusions of L6 followed by 1 week of
ing from 5 to 75 mg [497]. At doses of 50 mg and above rest, and then 4 days of s.c. IL-2 [811].
all patients experienced side effects that included fever, A humanized chimeric form of L6 was developed and
headache and nausea of mild degree. No responses were administered as single infusions ranging from 350 to
Robert O. Dillman 379

750 mg/m2, but no responses were seen in a phase II trial and altered biodistribution [405]. A dose-dependent in
of 21 patients [263]. Subsequent work with this Mab vitro T-cell proliferation was observed in 13/15 patients,
focused on radioimmunotherapy and a chemotherapy but no tumor responses were seen. A single-chain Fv
immunoconjugate. (scFv) and Fab fragment from this antibody have been
engineered that retain their specificity for MUC1 [552].
Monoclonal antibody 494/32 The huHMFG-1 (AS1402) antibody is a humanized
Buchler et al. conducted a prospective randomized IgG1 directed against MUC1 and is currently in clinical
trial of observation or adjuvant therapy with a 10-day trials for the treatment of breast carcinoma [502].
course of 370 mg of the murine IgG1 Mab 494/32 in 61 Seventeen patients with MUC1-positive cancers were
patients who had undergone a Whipple resection for treated with the murine anti-MUC1 BrevaRex mAb-
pancreatic cancer [63]. Analysis after 10 months AR20.5, which was given i.v. in doses of 1, 2 or 4 mg
showed no significant difference in survival between every 2 to 4 weeks for a total of six doses, with the intent
the two treatment groups who had median survivals of of forming immunogenic complexes as a vaccination
428 days for the treatment group, compared to 386 approach [152]. MUC1-specific anti-T cell responses
days for the control group. were confirmed in five patients.
Ryan et al. administered three different IgM anti-
breast human Mabs, selected based on their patterns of c30.6 anti-colorectal cancer antibody
tissue reactivity, to 10 patients with metastatic breast The mouse Mab m30.6, which had been used as a diag-
cancer [615]. One patient each was treated at dose levels nostic tool to detect colorectal cancer cells, was engi-
of 1, 2, 4, and 11 mg, and then 3 patients were treated at neered to create the chimeric Mab c30.6. Clinical trials
20 mg, and three other patients received 22 mg in addi- were initiated after demonstration of in vitro and in vivo
tion to tracer doses of 111-Indium labeled antibody. anti-tumor effects in preclinical models. Four patients
No tumor regressions were seen in this pilot study or with metastatic colorectal cancer received 3 mg of
relatively low doses of antibody. 123I-labeled c30.6, and the subsequent 13 patients were
Saleh et al. investigated momab D612 in a dose esca- treated with single doses up to 50 mg/m2 [769]. The
lation trial of multiple doses of 10 to 180 mg/m2 over 8 most frequent side effect was infusion-related erythema
days in patients with various gastrointestinal malignan- and a severe burning sensation on the face, neck, ears,
cies [625]. The dose limiting toxicity was a secretory chest, palms, soles, and genitalia, which was reduced by
diarrhea. HAMA was detected in 18/21 patients. The premedication with blockade of type I and II histamine
dose of 40 mg/m2 was selected for phase II trials. No receptors. No anti-tumor responses were seen.
responses were reported. In a subsequent study, 14
patients with metastatic gastrointestinal cancer received A33
the D612, 40 mg/m2, days 4, 7, and 11 along with 80 μg/kg/ The A33 antigen is a cell membrane surface glycopro-
day of recombinant human monocyte colony-stimulating tein that is sole or predominantly expressed on human
factor [626]. Secretory diarrhea occurred in 10 of 14 gastrointestinal epithelium and on nearly all colorectal
patients. There were no anti-tumor responses. cancers [300]. HuA33, is a CDR-grafted humanized
Mab against the A33 antigen. In an initial phase I study
Mucin Proteins and Human Milk Fat Globulin 11 patients with metastatic, chemotherapy-resistant
MUC1 is a member of a family of mucin transmem- colorectal cancer were given huA33 at dose of 10, 25, or
brane glycoproteins expressed on most glandular epi- 50 mg/m2/week every 4 weeks [778]. Significant toxicity
thelia [672]. Many adenocarcinomas over-express and occurred in four patients who had developed a strong
shed MUC1, and elevated MUC1 serum levels can serve HAHA response against the Mab. Two patients experi-
as a marker of tumor burden and progressive disease. enced infusion reactions that included fevers, rigors,
Human milk fat globulin (HMFG1) antigen, which is facial flushing, and blood pressure changes, while the
over-expressed in more than 90% of breast cancers and other two patients had serum-sickness type reactions
serous ovarian cancers in an underglycosylated form, is associated with skin rash, fever, and myalgia. One
the same as MUC1. Mab HMFG1 is a mouse IgG1 that patient, who was HAHA negative, had a PR. In a second
reacts with epitopes of MUC1 on high molecular weight phase I trial 12 colorectal cancer patients were given
human milk fat globulin glycoprotein antigen. Kosmos lower doses of huA33, 0.25, 1.0, 5.0, and 10 mg/m2
et al. treated 15 ovarian cancer patients with intraperito- along with a tracer dose of radiolabeled Mab prior to
neal HMFG1 rather than i.v. based on evidence that high surgery [647]. At these doses there was no significant
circulating levels of antigen result in immune complexes toxicity and tumor localization was confirmed.
380 Monoclonal antibody therapy

Lewis Y Antigen Prostate Cancer Associated Antigens


Lewis Y antigen is a blood group antigen that is highly
expressed on the surface of epithelial tumors. Hu3S193, Low doses of IgG1 murine Mabs reactive with prostatic
a humanized Mab that binds to the Le(y) antigen, was acid phosphatase (PAP) or prostate specific antigen
given as four weekly doses to eight colorectal cancer (PSA) were given to 19 patients with metastatic prostate
patients, six breast cancer patients, and one NSCLC cancer [183, 286]. Tracer doses of 111Indium-labeled
patient, at doses of 5, 10, 20, and 40 mg/m2 [648, 649]. antibodies showed uptake in some sites of tumor. There
All patients had tumors that were Le (y)-positive. Mild were no significant toxicities associated with treatment,
transient nausea and vomiting was observed following but nearly half of patients developed HAMA. No tumor
the 40 mg/m2 dose. Indium-111 tracer studies confirmed responses were observed. J591 is a humanized IgG1
uptake in metastatic sites. No objective tumor responses Mab that targets the extracellular domain of the trans-
were recorded. A similar trial with huS193 was carried membrane prostate-specific membrane antigen (PSMA).
out in 10 patients with Le (y)-positive small cell lung Fourteen patients with hormone refractory progressive
cancer [410]. Tumor uptake of radiolabeled Mab was metastatic prostate cancer received 10, 25, 50, and
confirmed. Toxicities, which again were mild, and only 100 mg of J591 with 2 mg of Indium-111 tracer [506].
seen at the 40 mg/m2 dose, included two patients with There were no significant toxicities and tumor uptake
nausea/vomiting, and one who developed urticaria. was confirmed. Investigators have reported results for
more than 2,000 prostate cancer patients who received
111-In CYT-356 (Capromab, Prostascint™) [298, 680].
G250 (CAIX, MN/CA9) Renal Cell Uptake in regional lymph nodes has been confirmed in
Cancer Antigen some patients, but there have not been reports of anti-
tumor effects. but tumor responses could not be mea-
The monoclonal antibody G250 (mAbG250) was pro-
sured in this trial design.
duced by immunizing against human renal cell carci-
noma (RCC). The G250 antigen was isolated and
determined to be the same as the MN/CA9 antigen of Melanoma and Neuroectodermal
HeLa cells which has carbonic anhydrase activity. The Associated Antigens
G250 antigen, which is also referred to as G250MN pro-
tein, and carbonic anhydrase IX (CAIX), is found on Melanoma Glycoprotein Antigens
95% of clear cell RCC, is rarely expressed on other types Many of the first Mab developed against solid tumors were
of kidney cancer or cancers from other organs [412]. The to melanoma-associated antigens, including melanotrans-
associated carbonic anhydrase activity has been impli- ferrin (p97, p96.5 or gp95 antigen) and high molecular
cated in regulation of cell proliferation in response to weight chondroitin sulfated proteoglycan core protein
hypoxia. For many years it has been proposed as a good (p240, p280). None of the trials utilizing anti-p97 and anti-
target for Mab-directed therapy [734]. p240 murine Mab resulted in objective tumor responses,
Initial studies were done with the murine Mab G250. but this may be due to the limited evidence in vitro for
In a phase I study, 16 RCC patients received five differ- CMC or ADCC with these murine Mabs and/or lack of
ent doses of 131-I labeled G250 a week prior to nephre- regulatory significance for these antigens, and the rela-
ctomy [536]. Good tumor localization was confirmed tively small doses that were delivered in these trials.
and no significant side effects. The murine IgG2a G250 Dillman and Halpern infused 1 to 50 mg of mouse Mabs
was converted to a mouse kappa light chain/human IgG1 into melanoma patients as single 2-h infusions, with or
chimeric Mab, cG250 [700]. In a dose escalation trial, without a 1 mg tracer dose of 111Indium radiolabeled Mab
13 patients received doses of 5, 10, 25, or 50 mg/m2 [183, 287]. Twenty-four patients received IgG1 Mabs
given weekly i.v. for 6 weeks [144]. There were no seri- directed against the p97 antigen [287, 679], and another 28
ous toxicities and one patient had a CR. In a phase II received IgG2a Mabs directed against the p240 antigen
trial 36 RCC patients, 21 of whom were pre-treated with [183, 288]. No definitive tumor responses were seen in
IL-2 and or IFN-α, were given weekly doses of 50 mg patients with measurable disease. Similar experiences were
cG250 was i.v. for 12 weeks [43, 749]. There were no reported by other investigators utilizing relatively low
serious adverse events. There were no objective quantities of these same Mabs with radiolabeled tracers
responses by the end of treatment, but one of ten patients [392, 514, 515]. Oldham et al. treated eight patients with
who continued treatment for an additional 8 weeks 9.2.27, an IgG2a murine Mab that reacts with the p240
eventually developed a CR. Doses of 5, 10, 25, or 50 mg/m2 glycoprotein antigen [535]. Patients received twice weekly
were given weekly by i.v. infusion for 6 weeks. escalating doses of 1, 10, 50, 100, and 200 mg. There was a
Robert O. Dillman 381

relationship between dose and tumor penetration and anti- of 10 mg/m2, 30 mg/m2 or 50 mg/m2 [453]. Toxicity was
gen saturation based on immunohistochemical staining of substantial and included hypoalbumenemia in all cases,
tumor biopsies which showed in vivo localization in s.c. and a death at the highest dose. There was one mixed
tumors in 6/8 patients following doses of 50 mg or greater. tumor response.
No tumor responses were seen. Goodman treated four In another phase I trial with murine R24, 37 patients
melanoma patients with a combination of two mouse with advanced melanoma received 1, 10, 20, 40, or
Mabs, an IgG1 anti-P97 and an IgG1 anti-p240, and a fifth 80 mg/m2 with five to six patients at each dose [393].
patient received anti-p97 alone [261]. Escalating antibody Urticaria was the most frequent adverse event. A dose-
doses were administered as 6-h infusions daily for 10 days limiting toxicity of pulmonary capillary leak syndrome
to a maximum dose of 50 mg per infusion. No objective occurred in three of five patients at the highest dose.
tumor regressions were observed. Hepatic enzyme elevation and chest pain were noted
even at lower doses. Two responses were seen at the
Melanoma Disialoganglioside Antigens lowest dose.
Many early murine Mabs derived from immunization R24 has been given in combination with a variety of
with melanoma cells reacted with disialoganglioside different immunostimulatory cytokines in the hopes of
glycolipid antigens, such as GD2 and GD3, which are enhancing antitumor effects. Caulfield et al. gave five
commonly expressed on neuroectodermal tissues [573]. daily 6-h infusions of escalating dose of R24 in combi-
nation with intramuscular with IFN-α2a to 15 mela-
Anti-GD3 noma patients [100]. Toxicities were similar to those
The murine anti-GD3 Mab R24 was given to 21 patients seen in other trials of R24. No tumor regressions were
at doses of 1, 10, 30, or 50 mg/m2 every other day for 2 reported, which was disappointing in view of the appar-
weeks for a totals of 8, 80, 240, or 400 mg/m2 [328, ent activity of R24 and IFN-α as single agents.
738]. At higher doses all patients developed urticaria Several trials of R24 and IL-2 have been conducted.
and pruritus that typically occurred within 2 to 4 h Bajorin et al. evaluated the combination of IL-2 and
following treatment and often appeared around tumor murine R24 in 20 patients with metastatic melanoma in
or at sites of previous tumor excision. In patients who a phase I trial in which IL-2 was given at a dose of 6
received 30 and 50 mg/m2, uptake of Mab in tumor was MIU/m2 i.v. over 6 h on days 1–5 and 8–12, while the
readily demonstrated using biopsies and immunohis- anti-GD3 antibody R24 was given on days 8–12 at 1, 3,
tochemical analysis. Significant tumor regressions were 8, or 12 mg/m2 per day with five patients evaluated at
seen in four patients: 2/6, 1/6, 1/6, and 0/3 for each each dose level [17]. Some in vitro T-cell activation was
successive dose level. Responses were first noted within demonstrated and one patient had a PR in soft tissue
2 weeks of completing treatment, but continued to sites lasting 6 months, and two other patients had minor
increase for several months. responses [779]. R24 combined with low-dose continu-
An effort was made to intensify the administration ous infusion IL-2 resulted in one PR and two minor
of mouse R24 since treatment was associated with a responses in 28 melanoma patients who were treated in
high rate of urticaria for patients receiving cumulative a dose escalation trial [682]. At higher doses two patients
doses of 400 mg/m2 over 2 weeks [18]. Eight patients experienced chest and abdominal discomfort that neces-
received R24 at doses of 800 and 1,200 mg/m2 over 1 sitated dose reductions. In a separate trial, Creekmore et
week by continuous i.v. infusion along with prophy- al. gave a higher dose of continuous infusion IL-2 fol-
lactic histamine blockers. Severe toxicity was noted. lowed by R24 [134]. This sequencing of the agents was
One patient developed anaphylaxis. All patients devel- associated with responses in 10/28 patients while con-
oped lymphopenia and decreases in serum complement. current administration was associated with responses in
A cytokine induced vascular leak syndrome occurred only 1/17 patients. However, in the sequential trial, there
in seven patients during the first 2 days of therapy. were also five patients who never received R24 because
Serum sickness was observed in six patients within a of the severity of IL-2-related toxicity.
week, which coincided with the ability to detect R24 has also been given to melanoma patients in com-
HAMA. The maximum tolerated dose was felt to be bination with GM-CSF, M-CSF, or TNF-α. Chachoua et al.
800 mg/m2 based on dose-limiting toxicities that gave s.c. GM-CSF for 21 days at a dose of 150 ug/m2/day,
included hypertension, chest pain, and vision changes and gave R24 by continuous i.v. infusion on days 8–15 at
at the 1,200 mg/m2. doses of 0, 10, or 50 mg/m2 [101]. There were no tumor
In another phase I trial in 11 patients with advanced responses observed with GM-CSF alone in five patients,
melanoma, R24 was given i.v. daily for 5 days at doses or the lower R24 dose in six patients, but two responses
382 Monoclonal antibody therapy

were seen at the 50 mg/m2 dose in nine patients. However, in a phase I trial in 13 melanoma patient, who received
4/9 were unable to complete this single course of therapy 5 to 100 mg of Mab [624]. As has been seen in other trials
because of toxicity. Minasian et al. treated 19 metastatic of anti-GD2 antibodies in adult patients, the major tox-
melanoma patients with a 14-day continuous infusion of icity associated with this therapy was abdominal/pelvic
recombinant human M-CSF at a dose of 80 ug/kg/day in pain syndrome that necessitated use of narcotic analgesics.
combination with R24 which was administered daily by No tumor responses were seen in this cohort of patients.
i.v. infusion at doses of 1, 3, 10, 0 and 50 ug/m2/day on days KM871 is an IgG1 kappa chimeric Mab to GD3, that
6 to 10 [492]. There were no objective tumor responses, was given to 17 metastatic melanoma patients at dose
although three patients did have a mixed response with levels of 1, 5, 10, 20, or 40 mg/m2 at 2-week intervals for
regression of some lesions. In a pilot study R24 was three doses [645]. Specific targeting of melanoma tissue
combined with two different doses of TNF in eight was demonstrated using indium-111 tracer Mab. No
patients with melanoma [491]. One patient had a dra- dose-limiting toxicity was observed although three
matic tumor lysis syndrome associated with hemorrhagic patients experienced pain and/or erythema at tumor sites
tumor necrosis in multiple visceral sites of disease. that was not related to dose. One patient had a partial
Combining data from 11 R24 trials cited above reveals response that lasted 11 months. KW-2871 is another
a cumulative response rate of 6/77 for R24 alone, and IgG1 kappa chimeric Mab to GD3 that was given to 17
6/127 for R24 in combination with other biologicals. metastatic melanoma patients who received an initial
10 mg/m2 and then 2 weeks later were stratified into four
Anti-GD2 cohorts to receive four doses of KW-2871 at 2-week
The IgG3 anti-GD2 mouse Mab 3F8 was used by intervals at doses of 20, 40, 60, and 80 mg/m2 [232]. The
Cheung et al. to treat melanoma patients who received maximum tolerated dose for this schedule was 40 mg/m2
5, 20, 50, or 100 mg/m2 as 8-h infusions given daily for based on grade three dose-limiting toxicities of laryn-
2 to 4 days [109]. The study was closed at the 100 mg/m2 gospasm and chest tightness at doses of 80 and 60 mg/
dose because all patients treated at that dose level devel- m2. Urticaria were noted in all 16 patients who were not
oped hypertension. Treatments were associated with focal premedicated with antihistamines.
pain at tumor sites and pain especially over the abdomen, Neuroblastomas also express disialogangliosides.
back, and extremities which was attributed to reactivity Treatment of eight children with the 3F8 anti-GD2
with neural tissue. Urticaria, fever, nausea, vomiting mouse Mab produced two CRs, one at 5 mg/m2, and the
and sweats were also noted. Inflammatory actions were other at 20 mg/m2 [109]). The abdominal/pelvic pain
observed around tumors, and partial responses were syndrome was not as problematic in these pediatric
reported for 2/9 patients and two others had a mixed patients as it had been in adults with melanoma. 3F8
response. Six melanoma patients were given a combina- was used to treat 16 neuroblastoma patients who had
tion of R24 and 3F8 at doses of only 1–10 mg/m2; but no stage 4 disease [111]. Adverse reactions included pain,
tumor regressions were seen. fever, urticaria, hypertension, hypotension and anaphy-
Saleh et al. conducted a phase I trial of a different lactoid reactions. Three patients survived more than 6
murine anti-GD2 Mab 14G2a, an IgG2a switch variant years. In an effort to eradicate minimal residual disease,
of the IgG3 anti-GD2 Mab 14.18, in which 12 patients 3F8 was given after completion of chemotherapy, to 34
with melanoma received single i.v. infusions of doses neuroblastoma patients, most of whom had bone and/or
from 10 to 120 mg [623]. Treatment was complicated by bone marrow metastases [110]. Only 23 were in CR at
abdominal and pelvic pain, which necessitated narcotic the time 3F8 was given. Pain, fever, and urticaria were
analgesia for control. All 12 patients developed HAMA. the most frequent adverse events noted. Fourteen
There was one PR. Murray et al. gave Mab 14G2a to 11 patients were still alive after 3 to 10 years of follow up.
patients with metastatic melanoma as part of a phase I Murray et al. reported two PR among five neuroblas-
trial [516]. There were no objective remissions although toma patients who were treated with the 14.18 Mab as
two patients exhibited mixed responses to the antibody. part of a phase I trial [516]. They also noted that pediat-
Significant adverse events were noted including gener- ric patients tolerated the agent much better than adults.
alized pain, fever, rash, paresthesias, weakness, hypona- The anti-GD2 Mabs have been combined with vari-
tremia and postural hypotension were the significant ous cytokines in an effort to enhance anti-tumor effi-
toxicities observed. The investigators suggested 100 mg/ cace. Yu and colleagues treated 17 neuroblastoma
m2 as the maximum tolerated dose of this Mab. patients, aged 2 to 8 years, with GM-CSF and the chi-
A chimeric version of the anti-GD2 antibody (ch14.18) meric Mab ch14.18, which was engineered with a
with a constant region of human IgG1κ has been tested human IgG1κ modification of the murine 14.18 [804].
Robert O. Dillman 383

Toxicity was minimal in these children as opposed to extramedullary myeloma with a murine anti-IL6 Mab
the experience in adult patients with melanoma. [29]. Three patients had an objective antiproliferative
Significant tumor responses were noted in eight patients, effect based on labeling index, and one had a minor
including three complete remissions. 3F8 and GM-CSF regression of a tumor mass. Most patients had some
were given to 19 neuroblastoma patients whose disease worsening of neutropenia and thrombocytopenia.
was resistant to initial therapy [411]. One cycle consisted Moreau et al. have used a combination of a murine anti-
of s.c. GM-CSF alone for 5 days, and then GM-CSF i.v. IL-6 monoclonal antibody with dexamethasone and
followed hour later by i.v. 3F8 on days 6–10, and 13–17 high dose melphalan in the autologous transplant setting
along with additional s.c. doses of GM-CSF on days 11 [501]. A major obstacle to treatment with anti-IL-6 is
and 12. The treatment was given in an outpatient setting. the very high rates of IL-6 production in some patients
Side effects were mild and included manageable pain with myeloma, which would need to be reduced by
and occasional rash. Complete responses were recorded another therapeutic modality [610]
for 12/19 patients.
Choi et al. treated 23 melanoma and four sarcoma IL-15
patients with a combination of ch14.18 and R24 anti- Interleukin-15 (IL-15) is a proinflammatory cytokine that
bodies together with continuous IL-2 [114]. When com- stimulates T lymphocytes and NK cells. HuMax-IL15 is
bined with IL-2, the maximum tolerated dose for both a human IgG1 anti-IL-15 Mab that suppresses T cell
Mab was 5 mg/m2/day because of dose-limiting toxici- proliferation and induces apoptosis in vitro. HuMax-IL15
ties including severe allergic reactions and pain with was given for 12 weeks to patients with rheumatoid
both products at the 7.5 mg/m2/day dose. Two melanoma arthritis in a double-blinded phase I/II dose-escalation,
patients had PRs. placebo-controlled trial that enrolled 30 patients [27].
L612 HuMAb is a human IgM that binds to ganglio- At the doses used in this trial there, HuMax-IL15 was
side GM3 and effects CDC with human complement. well tolerated and there were no significant effects on
Nine patients with measurable metastatic melanoma were subsets of T lymphocytes. This product may be devel-
given a continuous infusion of L612 at a dose of 960 mg, oped mostly for the treatment of auto-immune disease,
1,440 mg, or 1,920 mg over 48 h [341]. Mild pruritus and but could also be considered for certain hematologic malig-
skin rash were noted. No objective responses occurred, nancies in which IL-15 stimulation may be important.
but one patient reported pain in s.c. tumor sites.
Gastrin Releasing Peptide (Bombesin)
Peptides such as human gastrin-releasing peptide (GRP)
Other Ligand Targets and the peptide bombesin, are sometimes produced by
lung cancers. Mulshine et al. infused the anti-GRP
Interleukin-6 murine Mab 2A11 into 12 patients with NSCLC, at dose
Interleukin-6 (IL-6) is a growth factor for plasma cells of 1, 10, or 100 mg/m2 i.v. thrice weekly for 4 weeks
that is involved in autocrine/paracrine growth regulation [513]. No tumor regressions were noted. In a phase II
[107]. Soluble IL-6 receptors and L-6 are typically ele- trial, 12 patients with small cell carcinoma of the lung
vated in the advanced stages of myeloma. Klein et al. were given 2A11 at dose and schedule of 250 mg/m2
treated a patient whose primary plasma cell leukemia thrice weekly for 4 weeks [375]. In this study one patient
whose disease was refractory to chemotherapy, with achieved a complete radiographic remission which
daily i.v. anti-IL-6 Mab [396]. Serial monitoring during lasted 5 months, and four patients had stable disease. No
the 2 months of treatment demonstrated a decrease in significant toxicities were observed. In an accompany-
serum calcium, serum paraprotein, C-reactive protein, ing radioimmunodetection study, tumor uptake of
and the percentage of cells in S-phase in the bone mar- 111-Indium conjugated 2A111 was detected in 11/12
row, but an objective tumor response could not be patients [105]. The receptor for GRP/bombesin could
claimed. There were no major toxicities but decreases in also be a target for Mab therapy [807].
both platelet and white blood cell counts were noted. A
chimeric anti-IL-6 Mab was given to 12 patients with Fibroblast Activation Protein
myeloma at daily doses of 5, 10, 20, 40, and total doses Sibrotuzumab (BIBH 1) is a humanised version of the
of 140, 280, 560 mg, and 1,120 mg given as two 14-day murine anti-FAP mAb F19 which reacts with fibroblast
treatment cycles [747]. There were no responses, and activation protein (FAP). FAP is highly expressed by acti-
much of the anti-IL-6 antibody was complexed to circu- vated fibroblasts of the tumor stroma in many malignan-
lating IL-6. Bataille et al. treated 10 patients with cies including breast, colon and lung carcinomas [476].
384 Monoclonal antibody therapy

In a phase I trial, 26 patients, including 20 with metastatic autoimmune diseases and associated with similar risks for
colorectal cancer and six with NSCLC, were given sibrotu- infection. The specific marketing indications for adali-
zumab at doses of 5, 10, 25, or 50 mg/m2 weekly for 12 mumab are rheumatoid and psoriatic arthritis, ankylosing
weeks, with trace 131-I labeled Mab during weeks 1, 5, and spondylitis, and Chron’s disease [739]. At this time there
9 [646, 323]. There was minimal toxicity during 218 infu- are no published clinical trials in which adalimumab has
sions of sibrotuzumab administered during the first 12 been used in the treatment of human malignancy.
weeks and only one dose limiting toxicity was observed.
Tumor uptake of the Mab was confirmed. There were no Anti-Idiotype Vaccines
tumor responses.
In a phase II trial 25 metastatic colorectal cancer As discussed earlier in this chapter, the anti-idiotype
patients were given i.v. weekly 100 mg doses of sibrotu- cascade provides a rationale for developing Ab2 Mab
zumab for 12 scheduled weeks [777]. Five patients products as a vaccine to induce an endogenous Ab3
experienced infusion reactions that included rigors or response against the target antigen.
chills, flushing, nausea, and one episode of bronchos- In the nomenclature that is now in use, the generic
pasm. HACA were detected in three patients after four names of such products have “ab” at the beginning of
to 12 infusions. There were no tumor responses. their name.

Tumor Necrosis Factor Alpha (TNF-α) Anti-Idiotype Vaccines in Adenocarcinomas


Infliximab (Remicade®, Centocore, Malvern, PA) After clinical trials suggested that murine Mab 17-1A
Infliximab is a chimeric IgG1 kappa that neutralizes could induce an anti-idiotype cascade, Herlyn et al. tried
tumor necrois factor alpha (TNF-α) by blocking binding to induce endogenous human anti-tumor antibodies in
of the ligand to its receptor because of its own high affin- patients with colorectal cancer [314]. They gave patients
ity binding to the ligand. It is used in the treatment of a repeated s.c. injections of a goat antibody against the
variety of chronic inflammatory autoimmune disorders idiotype of Mab 17-1A. The goat antibody induced
including Crohn’s disease, ulcerative colitis, rheumatoid human antibodies against the same antigen targeted by
and psoriatic arthritis, uveitis, and ankylosing sondylitis 17-1A. A significant tumor response was seen in 1/30
[739]. Not surprisingly, sepsis and opportunistic infec- patients so treated.
tions are significant complications of TNF-α inhibition. Foon et al. treated 32 patients following resection of
Infliximab was given to 19 patients with cytokine- locoregionally advanced colorectal cancer, with 2 mg
resistant renal cell carcinoma (RCC) at doses of 5 mg/kg s.c. injections of CeaVac, an anti-idiotype antibody to
at weeks 0, 2, and 6, and then every 8 weeks and then to an anti CEA antibody, every other week for four injec-
18 RCC patients at doses of 10 mg/kg at weeks 0, 2, and tions and then monthly until tumor recurrence or pro-
6, and then every 4 weeks [294]. Severe adverse events gression [229]. CeaVac induced a potent anti-CEA
included a grade 3 hypersensitivity reaction and a death humoral and cellular immune response in all 32 patients,
from pulmonary infection with sepsis. Clinical activity including 15 patients who received 5-FU chemotherapy
was promising with partial responses observed in 3/19 during vaccination.
patients using the first dose/schedule and 11/18 using the The anti-idiotype Mab, an anti-HMFG antibody, vac-
second dose schedule. In a trial that enrolled patients cine 11D10 (TriAb), was given to 45 patients with meta-
with a variety of advanced malignancies, 21 patients static breast cancer that had responded to standard
received infliximab at 5 mg/kg and 20 patients were chemotherapy, before and after high dose chemotherapy
treated at 10 mg/kg at 0 and 2 weeks and then every 4 and autologous stem cell rescue [583]. Idiotype-specific
weeks [59]. These doses and schedule were not associ- humoral and T-cell proliferative responses were demon-
ated with any dose-limiting toxic effects. There were no strated in most patients despite the immunosuppressive
objective responses in this heterogeneous patient popu- effects of high-dose chemotherapy. The 48% 3-year
lation. In both these trials neutralization of plasma overall survival rate is similar to what has been reported
TNF-α was demonstrated shortly after the first infusion. in other series of stem cell transplant in patients with
advanced breast cancer.
Adalimumab (Humira®, Abbott, North Chicago, IL) In a multicenter cooperative group trial, 56 patients
Adalimumab is a recombinant human IgG1 that blocks the with colorectal cancer who had undergone curative
binding of TNF-α to its cell surface receptors. Like inflix- resection of their liver metastases, received a combina-
imab, it is also used in a variety of chronic inflammatory tion of two anti-idiotype monoclonal antibody vaccines,
Robert O. Dillman 385

one to CEA(CeaVac) and the other to HMFG (TriAb) with NeuGc-containing ganglioside that is present on
[567]. About 40% of patients were progression-free various solid tumors. Up to six i.d. injections of 1 to
after 2 years, which is similar to what is seen with 2 mg of anti-Id mAb were given every 2 weeks. In 20
surgery alone. patients with advanced melanoma, the treatment was
The anti-idiotypic antibody vaccine ACA125, that is well tolerated and 16 patients exhibited strong Ab3
designed to induce endogenous immune responses against responses against GM3 [7]. In locally advanced or met-
tumor antigen CA125, was given to 119 women with astatic breast cancer, eight out of nine patients who
advanced ovarian carcinoma [588]. A specific anti-anti- received four or more injections exhibited strong Ab3
idiotypic antibody (Ab3) response was detected in 68% responses against GM3 [159]. Similar reactions were
and endogenous CA125 antibodies were detected in 50%. seen among nine patients with advanced NSCLC who
Ab3-positive patients had a much longer survival than received injections of 1E10 following a good response
Ab3-negative patients (23 vs. 5 month, p < 0.0001). to chemotherapy and/or radiotherapy [521].
In a randomized phase II trial, prior to surgery 67
patients with colorectal cancer were randomized to Summary
receive the human anti-idiotypic Mab 105AD7 with or
without BCG or to no treatment [735]. Vaccinations The hybridoma and recombinant DNA technologies,
were initiated in 32 patients prior to surgery and were combined with advances in our understanding of immu-
planned to be continued for up to 2 years. Most of the nology, tumor biology, and ligand–receptor–signal trans-
immunized patients exhibited immune responses, but duction cell physiology, laid the foundation for monoclonal
specific immune data and comparative data have not antibody therapy of cancer. During1980–1990 exploratory
been reported. clinical trials confirmed many of the points to consider
when developing Mab therapy. During 1990–1995 many
murine mab were engineered to produce chimeric and
Anti-Idiotype Vaccines in Melanoma humanized antibodies. During 1997–2007 six unconju-
Mittelman and colleagues gave anti-idiotype antibodies gated Mab that have clinical indications specifically for
directed Mab that targeted the gp240 glycoprotein anti- the treatment of human malignancy became available for
gen, in effort to induce endogenous antibodies against widespread clinical use. Several of these have revolution-
gp240 on melanoma cells. In a phase I trial involving 16 ized cancer therapy and have become the highest reve-
patients, s.c. injections of MAF11-39 from 0.5 mg to nue-generating cancer drugs ever. Hundreds of other
4 mg were well tolerated; so a phase II trial was carried Mab are in development and/or clinical trials, including
out in which an additional 21 patients were given 2 mg several totally human Mab. Many of these may be in
on days 0,7, and 28 [493]. At the 2 mg dose 16 patients wide-spread clinical use by the time this chapter is pub-
exhibited a response to gp240 and one patient had a CR. lished. The next several years will see the introduction of
In another trial the MK-23 anti-idiotype antibody was many more Mab into the clinic and continued evolution of
conjugated to keyhole limpet hemocyanin (KLH) and their integration into cancer therapy, and perhaps eventu-
coadministered with BCG in 25 patients [494]. Three ally replacement of many current forms of therapy.
PRs were observed, and 14 of 23 patients developed
endogenous human antibodies against gp240.
Foon et al. treated 47 melanoma patients with TriGem, References
an anti-idiotype antibody to a Mab that binds to disialo- 1. Adair JR. Engineering antibodies for therapy. Immunol Rev 1992;
ganglioside GD2, using weekly s.c. injections of 1, 2, 4, 130:5–40.
or 8 mg in combination with QS-21 as an adjuvant [230]. 2. Agus DB, Bunn PA Jr, Franklin W, et al. HER-2/neu as a therapeu-
tic target in non-small cell lung cancer, prostate cancer, and ovarian
An anti-anti-idiotype (AB3) response was detected in cancer. Semin Oncol 2000;27 (6 suppl 11):53–63.
40/47 patients. One patient had a complete response. 3. Agus DB, Gordon MS, Taylor C, et al. Phase I clinical study of
pertuzumab, a novel HER dimerization inhibitor, in patients with
advanced cancer. J Clin Oncol 2005;23:2534–43.
1E10 Anti-Idiotype Vaccines 4. Agus DB, Sweeney CJ, Morris MJ, et al. Efficacy and safety of
single-agent pertuzumab (rhuMAb 2C4), a human epidermal
to Gangliosides growth factor receptor dimerization inhibitor, in castration-
A series of trials have been conducted with aluminum resistant prostate cancer after progression from taxane-based
therapy. J Clin Oncol 2007;25:675–81.
hydroxide-precipitated 1E10, which is an anti-idiotype
5. Ahn KS, Sethi G, Aggarwal BB. Nuclear factor-kappa B: from
murine Mab (Ab2) specific to an Ab1 Mab that reacts clone to clinic. Curr Mol Med 2007;7:619–37.
386 Monoclonal antibody therapy

6. Albrecht H, DeNardo SJ. Recombinant antibodies: from the laboratory 25. Baselga J, Carbonell X, Castaneda-Soto NJ, et al. Phase II study
to the clinic. Cancer Biother Radiopharm 2006;21:285–303. of efficacy, safety, and pharmacokinetics of trastuzumab mono-
7. Alfonso M, Diaz A, Hernandez AM et al. An anti-idiotype vaccine therapy administered on a 3-weekly schedule. J Clin Oncol
elicits a specific response to N-glycolyl sialic acid residues of gly- 2005;23:2162–71.
coconjugates in melanoma patients. J Immunol 2002;168:2523–9. 26. Baselga J, Trigo JM, Bourhis J, et al. Phase II multicenter study of
8. Ansell SM, Witzig TE, Kurtin PJ, et al. Phase 1 study of interleu- the antiepidermal growth factor receptor monoclonal antibody
kin-12 in combination with rituximab in patients with B-cell cetuximab in combination with platinum-based chemotherapy in
non-Hodgkin lymphoma. Blood. 2002;99:67–74. patients with platinum-refractory metastatic and/or recurrent
9. Ansell SM, Geyer SM, Maurer MJ, et al. Randomized phase II squamous cell carcinoma of the head and neck. J Clin Oncol
study of interleukin-12 in combination with rituximab in previ- 2005;23:5568–77.
ously treated non-Hodgkin’s lymphoma patients. Clin Cancer Res 27. Baslund B, Tvede N, Danneskiold-Samsoe B, et al. Targeting inter-
2006;12(20 Pt 1):6056–63. leukin-15 in patients with rheumatoid arthritis: a proof-of-concept
10. Ansell SM, Horwitz SM, Engert A, et al. Phase I/II study of an study. Arthritis Rheum 2005;52:2686–92.
anti-CD30 monoclonal antibody (MDX-060) in Hodgkin’s 28. Bastholt L, Specht L, Jensen K, et al. Phase I/II clinical and pharma-
lymphoma and anaplastic large-cell lymphoma. J Clin Oncol cokinetic study evaluating a fully human monoclonal antibody
2007;25:2764–9. against EGFr (HuMax-EGFr) in patients with advanced squamous
11. Arnould L, Gelly M, Penault-Llorca F, et al. Trastuzumab-based cell carcinoma of the head and neck. Radiother Oncol 2007;85:24–8.
treatment of HER2-positive breast cancer: an antibody-dependent 29. Bataille R, Barlogie B, Lu ZY, et al. Biololgic effects of anti-inter-
cellular cytotoxicity mechanism? Br J Cancer 2006;94:259–67. leukin-6 murine monoclonal antibody in advanced muliple
12. Asano M, Yukita A, Matsumoto T, et al. An anti-human VEGF myeloma. Blood 1995;86:685–91.
monoclonal antibody, MV833, that exhibits potent anti-tumor 30. Beatty JD, Duda RB, Williams LE, et al. Preoperative imaging of
activity in vivo. Hybridoma 1998;17:185–90. colorectal carcinoma with 111-In-labeled anticarcinoembryonic
13. Attard G, Kitzen J, Blagden SP, et al. A phase Ib study of pertu- antigen monoclonal antibody. Cancer Res 1986; 46:6494–502.
zumab, a recombinant humanised antibody to HER2, and doc- 31. Beck KE, Blansfield JA, Tran KQ et al. Enterocolitis in patients
etaxel in patients with advanced solid tumours. Br J Cancer with cancer after antibody blockade of cytotoxic T-lymphocyte-
2007;97:1338–43. associated antigen 4. J Clin Oncol 2006;24:2283–9.
14. Avner BP, Liao SK, Avner B, et al. Therapeutic murine monoclonal 32. Berek JS. Immunotherapy of ovarian cancer with antibodies: a
antibodies developed for individual cancer patients. J Biol Response focus on oregovomab. Expert Opin. Biol Ther 2004;4:1159–65.
Modif 1989;8:25–36. 33. Berek JS, Taylor PT, Gordon A, et al. Randomized, placebo-con-
15. Aviles A, Leon MI, Diaz-Maqueo JC, et al. Rituximab in the trolled study of oregovomab for consolidation of clinical remission
treatment of refractory follicular lymphoma – six doses are better in patients with advanced ovarian cancer. J Clin Oncol
than four. J Hematother Stem Cell Res 2001;10:313–6. 2004;22:3507–16.
16. Axelrod O, Silverman GJ, Dev V, et al. Idiotypic cross-reactivity of 34. Berinstein NL, Grillo-Lopez AJ, White CA, et al. Association of
immunoglobulins expressed in Waldenstrom’s macroglobulinemia, serum rituximab (IDEC-C2B8) concentration and anti-tumor
chronic lymphocytic leukemia, and mantle zone lymphocytes of response in the treatment of recurrent low-grade or follicular non-
secondary B-cell follicles. Blood 1991;77:1484–90. Hodgkin’s lymphoma. Ann Oncol 1998:9:995–1001.
17. Bajorin DF, Chapman PB, Wong G, et al. Phase I evaluation of a 35. Berlin J, Posey J, Tchekmedyian S, et al. Panitumumab with irino-
combination of monoclonal antibody R24 and interleukin 2 in tecan/leucovorin/5-fluorouracil for first-line treatment of meta-
patients with metastatic melanoma. Cancer Res 1990;50:7490–95. static colorectal cancer. Clin Colorectal Cancer. 2007, 6:427–32.
18. Bajorin DF, Chapman PB, Wong GY et al. Treatment with high 36. Bernard A, Boumsell L. The clusters of differentiation (CD)
dose mouse monoclonal (anti-GD3) antibody R24 in patients with defined by the First International Workshop on Human Leucocyte
metastatic melanoma. Melanoma Res 1992;2:355–62. Differentiation Antigens. Hum Immunol 1984;11:1–10.
19. Ball ED, Bernier GM, Cornwell III GG, et al. Monoclonal antibodies 37. Bernengo MG, Quaglino P, Comessatti A, et al. Low-dose intermittent
to myeloid differentiation antigens: in vivo studies of three patients alemtuzumab in the treatment of Sezary syndrome: clinical and immu-
with acute myelogenous leukemia. Blood 1983; 62:1203–1210. nologic findings in 14 patients. Haematologica 2007;92:784–94.
20. Ball ED, Selvaggi K, Hurd D, et al. A phase I clinical trial of sero- 38. Bertram JH, Gill PS, Levine AM, et al. Monoclonal antibody T101
therapy in patients with acute myeloid leukemia with an immuno- in T-cell malignancies: a clinical, pharmokinetic and immunologic
globulin M monoclonal antibody to CD15. Clin Cancer Res correlation. Blood 1986; 68:752–61.
1995;1:965–72. 39. Bhattacharya-Chatterjee M, Foon KA. Anti-idiotype antibody vac-
21. Barker E, Mueller BM, Handgretinger R, et al. Effect of a chimeric cine therapies of cancer. Cancer Treat Res 1998;94:51–68.
anti-ganglioside GD2 antibody on cell-mediated lysis of human 40. Bienvenu J, Chvetzoff R, Salles G, et al. Tumor necrosis factor
neuroblastoma cells. Cancer Res 1991; 51:144–9. alpha release is a major biological event associated with rituximab
22. Bartsch R, Wenzel C, Altorjai G, et al. Results from an observa- treatment. Hematol J 2001;2:378–84.
tional trial with oral vinorelbine and trastuzumab in advanced 41. Bier H, Hoffmann T, Hauser U, et al. Clinical trial with escalating
breast cancer. Breast Cancer Res Treat 2007;102:375–81. doses of the antiepidermal growth factor receptor humanized
23. Baselga J, Tripathy D, Mendelsohn, et al. Phase II study of weekly monoclonal antibody EMD72000 in patients with advanced
intravenous recombinant hmanized anti-p185HER2 monoclonal anti- squamous cell carcinoma of the larynx and hypopharynx. Cancer
body in patients with HER2/neu-overexpressing metastatic breast Chemother Pharmacol 2001;47:519–24.
cancer. J Clin Oncol 1996;14:737–44. 42. Birch JR, Racher JR. Antibody production. Advanced Drug
24. Baselga J, Norton L, Albanell J, et al. Recombinant humanized Delivery Reviews 2006;58:671–85.
anti-HER2 antibody (Herceptin) enhances the antitumor activity of 43. Bleumer I, Knuth A, Oosterwijk E, et al. A phase II trial of chime-
paclitaxel and doxorubicin against HER/neu overexpressing human ric monoclonal antibody G250 for advanced renal cell carcinoma
breast cancer xenografts. Cancer Res 1998;58:2825–31. patients. Br J Cancer 2004;90:985–90.
Robert O. Dillman 387

44. Bodey B, Bodey B Jr, Siegel SE, Kaiser HE. Genetically engi- 62. Brusamolino E, Rusconi C, Montalbetti L, et al. Dose-dense
neered monoclonal antibodies for direct anti-neoplastic treatment R-CHOP-14 supported by pegfilgrastim in patients with diffuse
and cancer cell specific delivery of chemotherapeutic agents. Curr large B-cell lymphoma: a phase II study of feasibility and toxicity.
Pharm Des 2000;6:261–76. Haematologica 2006;91:496–502.
45. Body JJ, Facon T, Coleman RE, et al. A study of the biological 63. Buchler M, Friess H, Schultheiss K-H, et al. A randomized controlled
receptor activator of nuclear factor-kappaB ligand inhibitor, deno- trial of adjuvant immunotherapy (murine monoclonal antibody
sumab, in patients with multiple myeloma or bone metastases from 494/32) in resectable pancreatic cancer. Cancer 1991;68:1507–12.
breast cancer. Clin Cancer Res 2006;12:1221–8. 64. Buchsbaum DJ, Forero-Torres A, LoBuglio AF. TRAIL-receptor anti-
46. Bokemeyer C, Bondarenko I, Makhson A et al. Cetuximab plus bodies as a potential cancer treatment. Future Oncol 2007;3:405–9.
5-FU/FA/Oxaliplatin (FOLFOX-4) versus FOLFOX-4 in the 65. Burdette S, Schwartz RS. Current concepts: Immunology. Idiotypes
first-line treatment of metastatic colorectal cancer: OPUS, a ran- and idiotypic network. N Engl J Med 1987 317:219–24.
domized phase ii study. Proc Am Soc Clin Oncol 2007:25:172s 66. Burger RA, Sill MW, Monk BJ et al. Phase II trial of bevacizumab
[abstract 4035]. in persistent or recurrent epithelial ovarian cancer or primary peri-
47. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab toneal cancer: a Gynecologic Oncology Group study. J Clin Oncol
for squamous-cell carcinoma of the head and neck. N Engl J Med 2007;25:5165–71.
2006;354:567–78. 67. Burnett KG, Masuho Y, Hernandez R et al. Human monoclonal
48. Bookman MA, Darcy KM, Clarke-Pearson D, et al. Evaluation of antibodies to breast cancer cells. In: Monoclonal Antibodies,
monoclonal humanized anti-HER2 antibody, trastuzumab, in Diagnostic and Therapeutic Use in Tumor and Transplantation.
patients with recurrent or refractory ovarian or primary peritoneal Chatterjee SN, ed. Littleton, MA, PSG Publishing, 1985, pp
carcinoma with overexpression of HER2: a phase II trial of the 47–62.
Gynecologic Oncology Group. J Clin Oncol 2003;21:283–90. 68. Burris H 3rd, Yardle D, Jones S, et al. Phase II trial of trastu-
49. Borchmann P, Schnell R, Fuss I, et al. Phase 1 trial of the novel zumab followed by weekly paclitaxel/carboplatin as first-line
bispecific molecule H22xKi-4 in patients with refractory Hodgkin treatment for patients with metastatic breast cancer. J Clin Oncol
lymphoma. Blood 2002;100:3101–7. 2004;22:1621–9.
50. Borghaei H, Alpaugh RK, Bernardo P, et al. Induction of adaptive 69. Burstein HJ, Kuter I, Campos SM, et al. Clinical activity of trastu-
Anti-HER2/neu immune responses in a Phase 1B/2 trial of 2B1 zumab and vinorelbine in women with HER2-overexpressing
bispecific murine monoclonal antibody in metastatic breast cancer metastatic breast cancer. J Clin Oncol 2001;19:2722–30.
(E3194): a trial coordinated by the Eastern Cooperative Oncology 70. Burstein HJ, Harris LN, Gelman R, et al. Preoperative therapy with
Group. J Immunother 2007;30:455–67. trastuzumab and paclitaxel followed by sequential adjuvant doxo-
51. Borup-Christensen P, Erb K, Jensenius JC, et al. Human-human rubicin/cyclophosphamide for HER2 overexpressing stage II or III
hybridomas for the study of antitumor immune response in patients breast cancer: a pilot study. J Clin Oncol 2003;21:46–53.
with colorectal cancer. Int J Cancer 1986; 37:683–688. 71. Burstein HJ, Harris LN, Marcom PK, et al. Trastuzumab and
52. Boucher Y, Leunig M, Jain RK. Tumor angiogenesis and intersti- vinorelbine as first-line therapy for HER2-overexpressing meta-
tial hypertension. Cancer Res 1996;56:4264–6. static breast cancer: multicenter phase II trial with clinical out-
53. Boue F, Gabarre J, Gisselbrecht C, et al. Phase II trial of CHOP comes, analysis of serum tumor markers as predictive factors, and
plus rituximab in patients with HIV-associated non-Hodgkin’s cardiac surveillance algorithm. J Clin Oncol 2003;21:2889–95.
lymphoma. J Clin Oncol 2006;24:4123–8. 72. Burtness B, Goldwasser MA, Flood W, et al. Phase III randomized
54. Bourhis J, Rivera F, Mesia R, et al. Phase I/II study of cetuximab in trial of cisplatin plus placebo compared with cisplatin plus cetuximab
combination with cisplatin or carboplatin and fluorouracil in in metastatic/recurrent head and neck cancer: an Eastern Cooperative
patients with recurrent or metastatic squamous cell carcinoma of Oncology Group study. J Clin Oncol 2005;23:8646–54.
the head and neck. J Clin Oncol 2006;24:2866–72. 73. Burton JD, Ely S, Reddy PK, et al. CD74 is expressed by multiple
55. Brekken RA, Overholser JP, Stastny VA, et al. Selective inhibition myeloma and is a promising target for therapy. Clin Cancer Res
of vascular endothelial growth factor (VEGF) receptor 2 (KDR/ 2004;10:6606–11.
Flk-1) activity by a monoclonal anti-VEGF antibody blocks tumor 74. Busam KJ, Capodieci P, Motzer R, et al. Cutaneous side-effects in
growth in mice. Cancer Res 2000;60:5117–24. cancer patients treated with the antiepidermal growth factor recep-
56. Bremer K. Semi-extended, six weekly rituximab infusions in pre- tor antibody C225. Br J Dermatol 2001;144:1169–76.
treated advanced low-grade B cell non-Hodgkin’s lymphoma: a 75. Buske C, Dreyling MH, Eimermacher H et al. Combined immuno-
phase II study. Anticancer Drugs 2003;14:809–15. chemotherapy (R-CHOP) results in significantly superior response
57. Brooks D, Taylor C, Dos Santos B, et al. Phase Ia trial of murine rates and time to treatment failure in first line treatment of patients
immunoglobulin A antitransferrin receptor antibody 42/6. Clin with lymphoplasmacytoid/immunocytoma. Blood 2004;104;50a
Cancer Res 1995;1:1259–65. [abstract 162].
58. Brown BA, Davis GL, Saltzgaber-Muller J, et al. Tumor specific 76. Buter J, Janssen RAJ, Martens A, et al. Phase I/II study of low-dose
genetically engineered murine/human chimeric monoclonal anti- intravenous OKT3 and subcutaneous interleukin-2 in metastatic
body. Cancer Res 1987; 47:3577–83. cancer. Eur J Cancer 1993;29A:2108–13.
59. Brown ER, Charles KA, Hoare SA, et al. A clinical study assessing 77. Buzdar AU, Ibrahim NK, Francis D, et al. Significantly higher
the tolerability and biological effects of infliximab, a TNF-alpha pathologic complete remission rate after neoadjuvant therapy with
inhibitor, in patients with advanced cancer. Ann Oncol 2008; [Epub trastuzumab, paclitaxel, and epirubicin chemotherapy: results of a
ahead of print] randomized trial in human epidermal growth factor receptor 2-pos-
60. Brown SL, Miller RA, Horning SJ, et al. Treatment of B-cell lym- itive operable breast cancer. J Clin Oncol 2005;23:3676–85.
phomas with anti-idiotype antibodies alone and in combination 78. Buzdar AU, Valero V, Ibrahim NK, et al. Neoadjuvant therapy with
with alpha interferon. Blood 1989;73:651–61. paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophosph-
61. Brown SL, Miller RA, Levy R, et al. Antiidiotype antibody therapy amide chemotherapy and concurrent trastuzumab in human epidermal
of B-cell lymphoma. Semin Oncol 1989;16:199–210. growth factor receptor 2-positive operable breast cancer: an update of
388 Monoclonal antibody therapy

the initial randomized study population and data of additional patients 98. Case DC Jr, Desch CE, Kalman LE, et al. Community-based trial
treated with the same regimen. Clin Cancer Res 2007;13:228–33. of R-CHOP and maintenance rituximab for intermediate- or
79. Byrd JC, Waselenko JK, Maneatis TA, et al: Rituximab therapy in high-grade non-Hodgkin lymphoma with first-cycle filgrastim
hematologic malignancy patients with circulating blood tumor for older patients. Clin Lymphoma Myeloma 2007;7:354–60.
99. Caulfield MJ, Murthy S, Tubbs RR, et al. Treatment of chronic
cells: association with increased infusion-related side effects and
lymphocytic leukemia with an anti-idiotypic monoclonal anti-
rapid tumor clearance. J Clin Oncol 1999;17:791–5. body. Cleve Clin J Med 1989;56:182–8.
80. Byrd JC, Murphy T, Howard RS, et al. Rituximab using a thrice 100. Caulfield MJ, Barna B, Murthy S, et al. Phase Ia/Ib trial of an
weekly dosing schedule in B-cell chronic lymphocytic leukemia anti-GD3 monoclonal antibody (R24) in combination with inter-
and small lymphocytic lymphoma demonstrates clinical activity feron alpha (rHuIFNa-2a) in patients with malignant melanoma.
and acceptable toxicity. J Clin Oncol 2001;19:2153–64. J Biol Response Mod 1990;9:319–28.
81. Byrd JC, Kitada S, Flinn IW et al. The mechanism of tumor cell 101. Chachoua A, Oratz R. Liebes L, et al. Phase Ib trial of granulo-
clearance by rituximab in vivo in patients with B-cell chronic lym- cyte-macrophage colony-stimulating factor combined with
phocytic leukemia: evidence of caspase activation and apoptosis murine monoclonal antibody R24 in patients with metastatic
melanoma. J Immunother 1994;16:132–41.
induction. Blood 2002;99:1038–43.
102. Chan AT, Hsu MM, Goh BC, et al. Multicenter, phase II study of
82. Byrd JC, Peterson BL, Morrison VA et al. Randomized phase 2 cetuximab in combination with carboplatin in patients with recur-
study of fludarabine with concurrent versus sequential treatment rent or metastatic nasopharyngeal carcinoma. J Clin Oncol
with rituximab in symptomatic, untreated patients with B-cell 2005;23:3568–76.
chronic lymphocytic leukemia: results from Cancer and Leukemia 103. Chan A, Martin M, Untch M, et al. Vinorelbine plus trastuzumab
Group B 9712 (CALGB 9712). Blood 2003;101:6–14. combination as first-line therapy for HER 2-positive metastatic
83. Byrd JC, Rai K, Peterson BL, et al. Addition of rituximab to flu- breast cancer patients: an international phase II trial. Br J Cancer
darabine may prolong progression-free survival and overall sur- 2006;95:788–93.
104. Chatterjee SK, Tripathi PK, Chakraborty M, et al: Molecular mim-
vival in patients with previously untreated chronic lymphocytic
icry of carcinoembryonic antigen by peptides derived from the
leukemia: an updated retrospective comparative analysis of structure of anti-idiotype antibody. Cancer Res 1998;58:1217–24.
CALGB 9712 and CALGB 9011. Blood 2005;105:49–53. 105. Chaudhry A, Carrasquillo JA, Avis IL, et al. Phase I and imag-
84. Byrd JC, O’Brien S, Flinn IW, et al. Phase 1 study of lumiliximab ing trial of a monoclonal antibody directed against gastrin-re-
with detailed pharmacokinetic and pharmacodynamic measurements leasing peptide in patients with lung cancer. Clin Cancer Res
in patients with relapsed or refractory chronic lymphocytic leuke- 1999;5:3385–93.
mia. Clin Cancer Res 2007;13:4448–55. 106. Chen HX, Mooney M, Boron M, et al. Phase II multicenter trial
85. Camacho LH. Novel therapies targeting the immune system: of bevacizumab plus fluorouracil and leucovorin in patients with
CTLA4 blockade with tremelimumab (CP-675,206), a fully advanced refractory colorectal cancer: an NCI Treatment Referral
human monoclonal antibody. Expert Opin Investig Drugs Center Trial TRC-0301. J Clin Oncol 2006;24:3354–60.
2008;17:371–85. 107. Chen YH, Shiao RT, Labayog JM, et al. Modulation of interleu-
86. Canevari S, Stoter G, Arienti F, et al. Regression of advanced ovar- kin-6/interleukin-6 receptor cytokine loop in the treatment of
ian carcinoma by intraperitoneal treatment with atuologous T lym- multiple myeloma. Leuk Lymphoma 1997;27:11–23.
phocytes retargeted by a bispecific monoclonal antibody. J Natl 108. Cheresh DA, Honsila CJ, Staffileno LK, et al. Disialoganglioside
Cancer Inst 1995;87:1463–69. GD3 on human melanoma serves as a relevant target antigen for
87. Cannistra SA, Matulonis UA, Penson RT et al. Phase II study of monoclonal antibody-mediated tumor cytolysis. Proc Natl Acad
bevacizumab in patients with platinum-resistant ovarian cancer or Sci USA 1985;82:5155–59.
peritoneal serous cancer. J Clin Oncol 2007;25:5180–6. 109. Cheung NK V, Lazarus H, Miraldi FD, et al. Ganglioside GD2
88. Caron PC, Scheinberg DA. Anti-CD33 monoclonal antibody M195 specific monoclonal antibody 3F8: A phase I study in patients
for the therapy of myeloid leukemia. Leuk Lymphoma with neuroblastoma and malignant melanoma. J Clin Oncol
1993;11(suppl 2)1–6. 1987;:1430–40.
89. Caron PC, Schwartz MA, Co MS, et al. Murine and humanized con- 110. Cheung NK, Kushner BH, Cheung IY, et al. Anti-G(D2) antibody
structs of monoclonal antibody M195 (anti-CD33) for the therapy of treatment of minimal residual stage 4 neuroblastoma diagnosed
acute myelogenous leukemia. Cancer 1994;73:1049–1056. at more than 1 year of age. J Clin Oncol 1998a;16:3053–60.
90. Caron PC, Dumont L, Scheinberg DA. Supersaturating infusional 111. Cheung NK, Kushner BH, Yeh SD, et al. 3F8 monoclonal anti-
humanized anti-CD33 monoclonal antibody HuM195 in myeloge- body treatment of patients with stage 4 neuroblastoma: a phase II
nous leukemia. Clin Cancer Res 1998;4:1421–8. study. Int J Oncol 1998b;12:1299–306.
91. Carpenter PA, Lowder J, Johnston L, et al. A phase II multicenter 112. Chia S, Clemons M, Martin LA, et al. Pegylated liposomal doxoru-
study of visilizumab, humanized anti-CD3 antibody, to treat ste- bicin and trastuzumab in HER-2 overexpressing metastatic breast
roid-refractory acute graft-versus-host disease. Biol Blood Marrow cancer: a multicenter phase II trial. J Clin Oncol 2006;24:2773–8.
Transplant 2005;11:465–71. 113. Chiorazzi N, Wasserman RL, Kunkel HG, et al. Use of Epstein-
92. Carrasquillo JA, Bunn PA, Keenan AM, et al. Radioimmuno- Barr virus-transformed B-cell lines for the generation of immu-
detection of cutaneous T-cell lymphoma with 111-In-T101 mono- noglobulin-producing human B-cell hybridomas. J Exp Med
clonal antibody. N Engl J Med 1986; 315:673–80. 1982; 156:930–5.
93. Carter P, Presta L, Gorman C, et al. Humanization of an anti- 114. Choi BS, Sondel PM, Hank JA, et al. Phase I trial of combined
p185Her2 antibody for human cancer. Proc Natl Acad Sci treatment with ch14.18 and R24 Mab and interleukin-2 for
1992;89:4285–9. patients with melanoma or sarcoma. Cancer Immunol Immunother
94. Carter PW. Monoclonal antibodies and the biological approach to 2006;55:761–74.
cancer. J Biol Response Modif 1985;4:325–39. 115. Chua YJ, Cunningham D. Panitumumab. Drugs Today 2006;
95. Cartron G, Dacheux L, Salles G, et al. Therapeutic activity of Nov;42:711–9.
humanized anti-CD20 monoclonal antibody and polymorphism in 116. Clamon G, Herndon J, Kern J, et al. Lack of trastuzumab activity
IgG Fc receptor FcgammaRIIIa gene. Blood 2002;99:754–8. in nonsmall cell lung carcinoma with overexpression of erb-B2:
96. Casali P, Inghirami G, Nakamura R, et al. Human monoclonals 39810: a phase II trial of Cancer and Leukemia Group B. Cancer
from antigen-specific selection of B-lymphocytes and transforma- 2005;103:1670–5.
tion by EBV. Science 1986; 234:476–8. 117. Clynes R. Antitumor antibodies in the treatment of cancer: Fc
97. Case DC Jr, Ebrahim K, Hedlund J, et al. Rituximab in relapsed receptors link opsonic antibody with cellular immunity. Hematol
follicular lymphoma. Blood 2002;100:292b [abstract #4704]. Oncol Clin North Am 2006;20:585–612.
Robert O. Dillman 389

118. Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational study of 137. Currie GA. Eighty years of immunotherapy: a review of immu-
the efficacy and safety of humanized anti-HER2 monoclonal nobiological methods used in the treatment of cancer. Int J Cancer
antibody in women who have HER2-overexperessing metastatic 1972;26:141–53.
breast cancer that has progressed after chemotehrapy for meta- 138. Curnow RT. Clinical experience with CD64-directed immunother-
static disease. J Clin Oncol 1999;17:2639–48. apy. An overview. Cancer Immunol Immunother 1997;45:210–5
119. Cohen A, Plliack A, Ben-Bassat I et al. Results of a Phase II study 139. Czuczman MS, Grillo-Lopez AJ, White CA, et al. Treatment of
employing a combination of fludarabine, cyclophosphamide, and patients with low-grade B-cell lymphoma with the combination
rituximab (FCR) as primary therapy for patients with advanced of chimeric anti-CD20 monoclonal antibody and CHOP chemo-
follicular lymphoma. Blood 2002;100:360a [abstract 1393]. therapy. J Clin Oncol 1999;17:268–76.
120. Cohenuram M, Saif MW. Panitumumab the first fully human 140. Czuczman MS, Weaver R, Alkuzweny B, et al. Prolonged clini-
monoclonal antibody: from the bench to the clinic. Anticancer cal and molecular remission in patients with low-grade or follicu-
Drugs 2007;18:7–15. lar non-Hodgkin’s lymphoma treated with rituximab plus CHOP
121. Coiffier B, Haioun C, Ketterer N, et al. Rituximab (anti-CD20 chemotherapy: 9-year follow-up. J Clin Oncol 2004;22:4711–6.
monoclonal antibody) for the treatment of patients with relapsing 141. Czuczman MS, Koryzna A, Mohr A, et al. Rituximab in combi-
or refractory aggressive lymphoma: a multicenter phase II study. nation with fludarabine chemotherapy in low-grade or follicular
Blood 1998;92:1927–32. lymphoma. J Clin Oncol 2005;23:694–704.
122. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus 142. Czuczman MS, Thall A, Witzig TE, et al. Phase I/II study of gal-
rituximab compared with CHOP alone in elderly patients with iximab, an anti-CD80 antibody, for relapsed or refractory follicu-
diffuse large-B-cell lymphoma. N Engl J Med 2002;346:235–42. lar lymphoma. J Clin Oncol 2005;23:4390–8.
123. Cole SP, Campling BG, Louwman IH, et al. A strategy for the 143. D’Adamo DR, Anderson SE, Albritton K, et al. Phase II study of
production of human monoclonal antibodies reactive with lung doxorubicin and bevacizumab for patients with metastatic soft-
tumor cells. Cancer Res 1984;44:2750–53. tissue sarcomas. J Clin Oncol 2005;23:7135–42.
124. Coloma MJ, Larrick JW, Ayala M, Gavilondo-Cowley JV. Primer 144. Davis ID, Wiseman GA, Lee FT, et al. A phase I multiple dose,
design for the cloning of immunoglobulin heavy-chain leader- dose escalation study of cG250 monoclonal antibody in patients
variable regions from mouse hybridoma cells using the PCR. with advanced renal cell carcinoma. Cancer Immun 2007;7:13.
Biotechniques 1991;11:152–4, 156 145. Davis T, White CA, Grillo-Lopez A, et al. Single-agent monoclo-
125. Colombat P, Salles G, Brousse, et al. Rituximab (anti-CD20 nal antibody efficacy in bulky non-Hodgkin’s lymphoma: results
monoclonal antibody) as single first-line therapy for patients with of a phase II trial of rituximab. J Clin Oncol 1999;17:1851–57.
follicular lymphoma with a low tumor burden: clinical and 146. Davis TA, Maloney DG, Caerwinski DK, et al. Anti-idiotype
molecular evaluation. Blood 2001;97:101–6. antibodies can induce long-term complete remissions in non-
126. Combs SE, Heeger S, Haselmann R, et al. Treatment of primary Hodgkin’s lymphoma without eradicating the malignant clone.
glioblastoma multiforme with cetuximab, radiotherapy and temo- Blood 1998;92:1184–90.
zolomide (GERT)-phase I/II trial: study protocol. BMC Cancer 147. Davis TA, Czerwinski DK, Levy R. Therapy of B-cell lymphoma
2006;6:133. with anti-CD20 antibodies can result in the loss of CD20 antigen
127. Conconi A, Martinelli G, Thieblemont C, et al. Clinical activity expression. Clin Cancer Res 1999;5:611–5.
of rituximab in extranodal marginal zone B-cell lymphoma of 148. Davis TA, Grillo-Lopez AJ, White CA, et al. Rituximab anti-CD20
MALT type. Blood. 2003;102:2741–5. monoclonal antibody therapy in non-Hodgkin’s lymphoma: safety
128. Cortelezzi A, Pasquini MC, Sarina B, et al. A pilot study of low- and eficacy of re-treatment. J Clin Oncol 2000;18:3135–43.
dose subcutaneous alemtuzumab therapy for patients with chemo- 149. Davis TA, Maloney DG, Grillo-Lopez AJ, et al. Combination
therapy-refractory chronic lymphocytic leukemia. Haematologica immunotherapy of relapsed or refractory low-grade or follicular
2005;90:410–2. non-Hodgkin’s lymphoma with rituximab and interferon-α-2a.
129. Cote RJ, Morrissey DM, Houghton AN, et al. Generation of Clin Cancer Res 2000;6:2644–52.
human monoclonal antibodies reactive with cellular antigens. 150. Davis TA, Kaminski MS, Leonard et al. The radioisotope con-
Proc Natl Acad Sci 1983;80:2026–30. tributes significantly to the activity of radioimmunotherapy. Clin
130. Coudert BP, Largillier R, Arnould L, et al. Multicenter phase II Cancer Res 2004;10 :7792–8.
trial of neoadjuvant therapy with trastuzumab, docetaxel, and 151. de Bono JS, Tolcher JS, Forero A, et al. ING-1, a monoclonal
carboplatin for human epidermal growth factor receptor-2-over- antibody targeting Ep-CAM in patients with advanced adenocar-
expressing stage II or III breast cancer: results of the GETN(A)-1 cinomas. Clin Cancer Res 2004;10:7555–65.
trial. J Clin Oncol 2007;25:2678–84. 152. de Bono JS, Rha SY, Stephenson J et al. Phase I trial of a murine
131. Courtenay-Luck NS, Epenetos AA, Moore R, et al. Development antibody to MUC1 in patients with metastatic cancer: evidence
of primary and secondary immune responses to mouse monoclo- for the activation of humoral and cellular antitumor immunity.
nal antibodies used in the diagnosis and therapy of malignant Ann Oncol 2004;15:1825–33.
neoplasms. Cancer Res 1986;46:6489–93. 153. de Bono JS, Bellmunt J, Attard G, et al. Open-label phase II study
132. Crane CH, Ellis LM, Abbruzzese JL, et al. Phase I trial evaluat- evaluating the efficacy and safety of two doses of pertuzumab in
ing the safety of bevacizumab with concurrent radiotherapy and castrate chemotherapy-naive patients with hormone-refractory
capecitabine in locally advanced pancreatic cancer. J Clin Oncol prostate cancer. J Clin Oncol 2007;25:257–62.
2006;24:1145–51. 154. Delbaldo C, Pierga JY, Dieras V, et al. Pharmacokinetic profile of
133. Cranmer LD, Hersch E. The role of the CTLA4 blockade in the cetuximab (Erbitux) alone and in combination with irinotecan in
treatment of malignant melanoma. Cancer Invest 2007;25:613–31. patients with advanced EGFR-positive adenocarcinoma. Eur J
134. Creekmore S, Urba W, Kopp W, et al. Phase IB/II trial of R24 Cancer 2005;41:1739–45.
antibody and interleukin-2 in melanoma. Proc Am Soc Clin 155. De Maio E, Pacilio C, Gravina A, et al. Vinorelbine plus 3-weekly
Oncol 1992;11:345. trastuzumab in metastatic breast cancer: a single-centre phase 2
135. Crombet T, Osorio M, Cruz T, et al. Use of the humanized anti- trial. BMC Cancer 2007;7:50.
epidermal growth factor receptor monoclonal antibody h-R3 in 156. Demiden A, Lam T, Alas S, et al. Chimeric anti-CD20 (IDEC
combination with radiotherapy in the treatment of locally advanced C2B8) monoclonal antibody sensitizes a B cell lymphoma cell
head and neck cancer patients. J Clin Oncol 2004;22:1646–54. line to cell killing by cytotoxic drugs. Cancer Biother Radiopharm
136. Cunningham D, Humblet Y, Siena S, et al. Cetuximab monother- 1997;12:177–186.
apy and cetuximab plus irinotecan in irinotecan-refractory meta- 157. DePinho RA, Feldman LB, Scharff MD. Tailor-made monoclonal
static colorectal cancer. N Engl J Med 2004;351:337–45. antibodies. Ann Intern Med 1986;104:225–33.
390 Monoclonal antibody therapy

158. Diamond BA, Yelton DE, Scharff MD. Monoclonal antibodies: a 183. Dillman RO, Beauregard J, Ryan KP, et al. Radioimmunodetection
new technology for producing serologic reagents. N Engl J Med of cancer using indium-labeled monoclonal antibodies. International
1981;304:1344–49. symposium on labeled and unlabeled antibodies in cancer diagnosis
159. Diaz A, Alfonso M, Alonso R et al. Immune responses in breast and therapy. NCI Monographs 1987;3:33–6.
cancer patients immunized with an anti-idiotype antibody 184. Dillman RO, Beauregard JC, Jamieson M, et al. Toxicities asso-
mimicking NeuGc-containing gangliosides. Clin Immunol ciated with monoclonal antibody infusions in cancer patients.
2003;107:80–9. Molec Biother 1988;1:81–5.
160. Di Bella N, Reynolds C, Faragher D, et al. An open-label pilot 185. Dillman RO, Shawler DL, McCallister TJ, et al. Human anti-
study of pentostatin, mitoxantrone, and rituximab in patients with mouse antibody response in cancer patients following single low-
previously untreated, Stage III or IV, low-grade non-Hodgkin dose injections of radiolabeled murine monoclonal antibodies.
lymphoma. Cancer 2005;103:978–84. Cancer Biother 1994;9:17–28.
161. Dillman RO. Monoclonal antibodies in the treatment of cancer. 186. Dillman RO, Schreeder MT, Hon JK, et al. Community-based phase
CRC Crit Rev Hematol/Oncol 1984;1:357–86. II tiral of pentostatin, cyclophosphamide, and rituximab (PCR) bio-
162. Dillman RO. Antibody Therapy, in Oldham RK (ed): Principles chemotherapy in chronic lymphocytic leukemia and small lympho-
of Cancer Biotherapy (ed1). New York, Raven Press, 1987. cytic lymphoma. Cancer Biother Radiopharm 2007;22:185.
163. Dillman RO. Monoclonal antibodies for treating cancer. Ann 187. Dimopoulos MA, Zervas C, Zomas A, et al. Treatment of
Intern Med 1989; 111:592–603. Waldenstrom’s macroglobulinemia with rituximab. J Clin Oncol
164. Dillman RO. The human antimouse and antiglobulin responses to 2002;20:2327–33.
monoclonal antibodies. Antibody, Immunocon & Radiopharm 188. Dono M, Cerruti G, Zupo S. The CD5 + B-cell. Int J Biochem
1990; 3:1–15. Cell Biol. 2004;36:2105–11.
165. Dillman RO. Antibody Therapy, in Oldham RK (ed): Principles of 189. Dorfman NA. The optimal technological approach to the devel-
Cancer Biotherapy (ed2). New York, Marcel Dekker, Inc, 1991. opment of human hybridomas. J Biol Response Modif 1985;
166. Dillman RO. Antibodies as cytotoxic therapy. J Clin Oncol 4:213–39.
1994;12:1497–515. 190. Downey SG, Klapper JA, Smith FO, et al. Prognostic factors
167. Dillman RO. Whither magic bullets and guided missiles: mono- related to clinical response in patients with metastatic melanoma
clonal antibodies 20 years later. Cancer Biother 1995;10:177–80. treated by CTL-associated antigen-4 blockade. Clin Cancer Res
168. Dillman RO. Magic bullets at last! Finally – approval of a mono- 2007;13:6681–8.
clonal antibody for the treatment of cancer! Cancer Biother 191. Dowsett M, Bartlett J, Ellis IO, et al. Correlation between immu-
1997;12:223–25. nohistochemistry (HercepTest) and fluorescence in situ hybrid-
169. Dillman RO. Antibody Therapy, in Oldham RK (ed): Principles of ization (FISH) for HER-2 in 426 breast carcinomas from 37
Cancer Biotherapy (ed3). Netherlands, Kluwer, 1998, 284–371. centres. J Pathol 2003;199:418–23.
170. Dillman RO. Infusion reactions associated with the therapeutic 192. Drapkin R, Di Bella NJ, Cuasay LC et al. Phase II multicenter
use of monoclonal antibodies in the treatment of malignancy. trial of pentostatin and rituximab in patients with previously
Cancer Metastasis Rev 1999;18:465–471. treated or untreated chronic lymphocytic leukemia. Blood
171. Dillman RO. Perceptions of Herceptin: a monoclonal antibody 2002;100:803a [abstract 3171].
for the treatment of breast cancer. Cancer Biother Radiopharm 193. Drapkin R, Di Bella NJ, Faragher DC, et al. Results of a phase II
1999;14:5–10. multicenter trial of pentostatin and rituximab in patients with low
172. Dillman RO. Monoclonal antibodies in the treatment of malig- grade B-cell non-Hodgkin’s lymphoma: an effective and mini-
nancy: basic concepts and recent developments. Cancer Invest mally toxic regimen. Clin Lymphoma 2003;4:169–75.
2001;19:833–41. 194. Drorovolskaia MA, Kozlov SV. Inflammation and cancer: when
173. Dillman RO. The history and rationale for monoclonal antibodies NF-kappaB amalgamates the perilous partnership. Curr Cancer
in the treatment of hematologic malignancy. Clinical Pharmaceut Drug Targets 2005;5:325–44.
Biotechnol 2001;2:293–300. 195. Dyer MJ. The role of CAMPATH-1 antibodies in the treatment of
174. Dillman RO. Radiolabeled anti-CD20 monoclonal antibodies for lymphoid malignancies. Semin Oncol 1999;26(5 suppl 14) 52–57.
the treatment of B-cell lymphoma. J Clin Oncol 2002;20:3545–57. 196. Dyer MJS, Hale G, Hayhoe FGJ, et al. Effects of CAMPATH-1
175. Dillman RO, Hendrix CS. Unique aspects of supportive care antibodies in vivo in patients with lymphoid malignancies: influ-
using monoclonal antibodies in cancer treatment. Supportive ence of antibody isotype. Blood 1989;73:1431–1439.
Cancer Therapy 2003;1:38–48. 197. Economopoulos T, Fountzilas G, Pavlidis N, et al. Rituximab in
176. Dillman RO, Royston I. Using monoclonal antibodies to treat combination with CNOP chemotherapy in patients with previ-
leukemia and lymphomas. Drug Ther 2003;8:62–74. ously untreated indolent non-Hodgkin’s lymphoma. Hematol J
177. Dillman RO, Shawler DL, Sobol RE, et al. Murine monoclonal 2003;4:110–5.
antibody therapy in two patients with chronic lymphocytic leuke- 198. Edelson RL, Raafat J, Berger CL, et al. Anti-thymocyte globulin
mia. Blood 1982;59:1036–45. in the management of cutaneous T-cell lymphoma. Cancer Treat
178. Dillman RO, Beauregard JC, Sobol RE, et al. Lack of radioim- Rep 1979; 63:675–80.
munodetection and complications associated with monoclonal 199. Editorial. Lancet 1994;344:1013.
antibody cross-reactivity with an antigen on circulating cells. 200. Ehlen TG, Hoskins PJ, Miller D, et al. A pilot phase 2 study of
Cancer Res 1984;44:2213–17. oregovomab murine monoclonal antibody to CA125 as an immu-
179. Dillman RO, Shawler DL, Dillman JB, et al. Therapy of chronic notherapeutic agent for recurrent ovarian cancer. Int J Gynecol
lymphocytic leukemia and cutaneous T-cell lymphoma with Cancer 2005;15:1023–34.
T101 monoclonal antibody. J Clin Oncol 1984; 2:881–91. 201. Ehrlich P. Uben den jetzigen stand der Karzinomforschung. In:
180. Dillman RO, Beauregard J, Shawler DL, et al. Continuous infusion of The collected papers of Paul Ehrlich. Vol. II. London: Pergamon
T101 monoclonal antibody in chronic lymphocytic leukemia and Press 1956:550–57.
cutaneous T-cell lymphoma. J Biol Response Mod 1986;5:394–410. 202. Eisenberg BI. The polymerase chain reaction. N Engl J Med
181. Dillman RO, Johnson DE, Shawler DL, et al. Immune interferon 1990;322:178–83
modulation of in vitro murine anti-human T-cell monoclonal 203. Ekstrand BC, Lucas JB, Horwitz SM, et al. Rituximab in lym-
antibody mediated cytotoxicity. J Immunol 1986; 136:728–31. phocyte-predominant Hodgkin disease: results of a phase 2 trial.
182. Dillman RO, Shawler DL, Johnson DE, et al. Preclinical trials Blood 2003;101:4285–9.
with combinations and conjugates of T101 and doxorubicin. 204. Elaraj DM, White DE, Steinberg SM, et al. A pilot study of anti-
Cancer Res 1986;46:4886–91. angiogenic therapy with bevacizumab and thalidomide in
Robert O. Dillman 391

patients with metastatic renal cell carcinoma. J Immunother 225. Fleming GF, Meropol NJ, Rosner GL, et al. A phase I trial of
2004;27:259–64. escalating doses of trastuzumab combined with daily subcutane-
205. Elias DJ, Hirschowitz L, Kline LE, et al. Phase I clinical com- ous interleukin 2: report of Cancer and Leukemia Group B 9661.
parative study of monoclonal antibody KS1/4 and KS1/4- Clin Cancer Res 2002;8:3718–27.
methotrexate immunoconjugate in patients with non-small cell 226. Folkman J. Tumor angiogenesis. Adv Cancer Res. 1985;43:175–203.
lung carcinoma. Cancer Res 1990;50:4154–9. 227. Foon KA, Bunn PA, Schroff RW, et al. Monoclonal antibody
206. Ellis JH, Barber KA, Tutt A, et al. Engineered anti-CD38 mono- therapy of chronic lymphocytic leukemia and cutaneous T-cell
clonal antibodies for immunotherapy of multiple myeloma. J lymphoma: preliminary observations. In: Langman RE,
Immunol 1995;155:925–37. Trowbridge IS, Dulbecco R, eds. Monoclonal antibody and
207. Elter T, Borchmann P, Schulz H, et al. Fludarabine in combina- Cancer. New York:Academic Press 1984; pp39–52.
tion with alemtuzumab is effective and feasible in patients with 228. Foon KA, Schroff RW, Bunn PA, et al. Effects of monoclonal
relapsed or refractory B-cell chronic lymphocytic leukemia: antibody therapy in patients with chronic lymphocytic leukemia.
results of a phase II trial. J Clin Oncol 2005;23:7024–31. Blood 1984;64:1085–1093.
208. Emmanouilides C, Sfakiotaki G, Androulakis N, et al. Front-line 229. Foon KA, John WJ, Chakraborty M, et al. Clinical and immune
bevacizumab in combination with oxaliplatin, leucovorin and responses in resected colon cancer patients treated with anti-idio-
5-fluorouracil (FOLFOX) in patients with metastatic colorectal type monoclonal antibody vaccine that mimics the carcinoem-
cancer: a multicenter phase II study. BMC Cancer 2007;7:91. bryonic antigen. J Clin Oncol 1999;17:2889–95.
209. Enblad G, Hagberg H, Erlanson M, et al. A pilot study of alemtu- 230. Foon KA, Lutzky J, Baral RN, et al. Clinical and immune
zumab (anti-CD52 monoclonal antibody) therapy for patients responses in advanced melanoma patients immunized with an
with relapsed or chemotherapy-refractory peripheral T-cell lym- anti-idiotype antibody mimicking disialoganglioside GD2. J Clin
phomas. Blood 2004;103:2920–4. Oncol 2000,18:376–84.
210. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus 231. Foran JM, Rohatiner AZ, Cunningham D, et al. European
interferon alfa-2a for treatment of metastatic renal cell carci- phase II study of rituximab (chimeric anti-CD20 monoclonal
noma: a randomised, double-blind phase III trial. Lancet antibody) for patients with newly diagnosed mantle-cell lym-
2007;370:2103–11. phoma and previously treated mantle-cell lymphoma, immuno-
211. Eskens FA, Verweij J. The clinical toxicity profile of vascular cytoma, and small B-cell lymphocytic lymphoma. J Clin Oncol
endothelial growth factor (VEGF) and vascular endothelial 2000;18:317–24.
growth factor receptor (VEGFR) targeting angiogenesis inhibi- 232. Forero A, Shah J, Carlisle R, et al. A phase I study of an anti-GD3
tors; a review. Eur J Cancer 2006;42:3127–39. monoclonal antibody, KW-2871, in patients with metastatic mel-
212. Esteva FJ, Valero V, Booser D, et al. Phase II study of weekly anoma. Cancer Biother Radiopharm 2006;21:561–8.
docetaxel and trastuzumab for patients with HER-2-overexpressing 233. Forstpointner R, Dreyling M, Repp R, et al. The addition of ritux-
metastatic breast cancer. J Clin Oncol 2002;20:1800–8. imab to a combination of fludarabine, cyclophosphamide, mitox-
213. Ettlinger DE, Mitterhauser M, Wadsak W, et al. In vivo disposi- antrone (FCM) significantly increases the response rate and
tion of irinotecan (CPT-11) and its metabolites in combination prolongs survival as compared with FCM alone in patients with
with the monoclonal antibody cetuximab. Anticancer Res relapsed and refractory follicular and mantle cell lymphomas:
2006;26(2B):1337–41. results of a prospective randomized study of the German Low-
214. Faderl S, Thomas DA, O’Brien S. Experience with alemtuzumab Grade Lymphoma Study Group. Blood. 2004;104:3064–71.
plus rituximab in patients with relapsed and refractory lymphoid 234. Forstpointner R, Unterhalt M, Dreyling M, et al. Maintenance
malignancies. Blood 2003;101:3413–5. therapy with rituximab leads to a significant prolongation of
215. Fagerberg J, Ragnhammar P, Liljefors M, et al: Humoral anti- response duration after salvage therapy with a combination of
idiotypic and anti-antiidiotypic immune response in cancer rituximab, fludarabine, cyclophosphamide, and mitoxantrone
patients treated with monoclonal antibody 17–1A. Cancer (R-FCM) in patients with recurring and refractory follicular and
Immunol Immunother 1996;42:81–87. mantle cell lymphomas: Results of a prospective randomized
216. Farag SS, Flinn IW, Modali R, et al. Fc gamma RIIIa and Fc study of the German Low Grade Lymphoma Study Group
gamma RIIa polymorphisms do not predict response to rituximab (GLSG). Blood 2006;108:4003–8.
in B-cell chronic lymphocytic leukemia. Blood 2004;103:1472–4. 235. Fountzilas G, Tsavdaridis D, Kalogera-Fountzila A, et al. Weekly
217. Feldman E, Kalaycio M, Weiner G, et al. Treatment of relapsed paclitaxel as first-line chemotherapy and trastuzumab in patients
or refractory acute myeloid leukemia with humanized anti-CD33 with advanced breast cancer. A Hellenic Cooperative Oncology
monoclonal antibody HuM195. Leukemia 2003;17:314–8. Group phase II study. Ann Oncol 2001;12:1545–51.
218. Ferrara N. Molecular and biological properties of vascular 236. Fountzilas G, Christodoulou C, Tsavardaridis D, et al. Paclitaxel
endothelial growth factor. J Mol Med 1999;77:527–43. and gemcitabine, as first-line chemotherapy, combined with tras-
219. Feugier P, Van Hoof A, Sebban C, et al. Long-term results of the tuzumab in patients with advanced breast cancer: a phase II study
R-CHOP study in the treatment of elderly patients with diffuse conducted by the Hellenic Cooperative Oncology Group
large B-cell lymphoma: a study by the Groupe d’Etude des (HeCOG). Cancer Invest 2004;22:655–62.
Lymphomes de l’Adulte. J Clin Oncol 2005;23:4117–26. 237. Fracasso PM, Burris H, 3rd, Arquette MA, et al. A phase 1 esca-
220. Fiddler IJ, Hart IR. Biological diversity in metastatic neoplasms: lating single-dose and weekly fixed-dose study of cetuximab:
origins and implications. Science 1982;217:998–1003. pharmacokinetic and pharmacodynamic rationale for dosing.
221. Fiedler W, Krüger W, Laack E, et al. A clinical trial of edreco- Clin Cancer Res 2007;13:986–93.
lomab, interleukin-2 and GM-CSF in patients with advanced col- 238. Frank MM. Complement in the pathophysiology of human dis-
orectal cancer. Oncol Rep 2001;8:225–31. ease. N Engl J Med 1987; 316:1525–30.
222. Fiegl M, Falkner A, Hopfinger G, et al. Routine clinical use of 239. Friedberg JW, Neuberg D, Gribben JG, et al. Combination immu-
alemtuzumab in patients with heavily pretreated B-cell chronic notherapy with rituximab and interleukin 2 in patients with
lymphocytic leukemia: a nation-wide retrospective study in relapsed or refractory follicular non-Hodgkin’s lymphoma. Br J
Austria. Cancer 2006;107:2408–16. Haematol 2002;117:828–34.
223. Fischer N, Leger O. Bispecific antibodies: molecules that enable 240. Frodin JE, Harmenberg U, Biberfeld P, et al. Clinical effects of
novel therapeutic strategies. Pathobiology 2007;74:3–14. monoclonal antibodies [MAb 17–1A] in patients with metastatic
224. Fisher RI, Kobota TT, Mandell GL, et al. Regression of a T-cell colorectal carcinomas. Hybridoma 1988;7:309–21.
lymphoma after administration of anti-thymocyte globulin. Ann 241. Furukawa K, Yamaguchi H, Oettgen HF, et al. Two human mono-
Intern Med 1978;88:799–800. clonal antibodies reacting with the major gangliosides of human
392 Monoclonal antibody therapy

melanomas and comparison with corresponding mouse monoclo- 262. Goodman GE, Hellstrom I, Brodzinsky L, et al. Phase I trial of
nal antibodies. Cancer Res 1989; 49:191–96. murine monoclonal antibody L6 in breast, colon, ovarian, and
242. Fury MG, Lipton A, Smith KM, et al. A phase-I trial of the epi- lung cancer. J Clin Oncol 1990; 8:1083–92.
dermal growth factor receptor directed bispecific antibody MDX- 263. Goodman GE, Hellstron I, Yelton DE, et al. Phase I trial of chi-
447 without and with recombinant human granulocyte-colony meric (human-mouse) monoclonal antibody L6 in patients with
stimulating factor in patients with advanced solid tumors. Cancer non-small cell lung, colon, and breast cancer. Cancer Immunol
Immunol Immunother 2008;57:155–63. Immunother 1993;36:267–73.
243. Galli SJ, Kalesnikoff J, Grimbaldeston MA et al. Mast cells as 264. Gordan LN, Grow WB, Pusateri A et al. Phase II trial of individu-
“tunable” effector and immunoregulatory cells: recent advances. alized rituximab dosing for patients with CD20-positive lym-
Annu Rev Immunol 2005;23:749–86. phoproliferative disorders. J Clin Oncol 2005;23:1096–102.
244. Gatzemeier U, Groth G, Butts C, et al. Randomized phase II trial 265. Gordon MS, Margolin K, Talpaz M, et al. Phase I safety and
of gemcitabine-cisplatin with or without trastuzumab in HER2- pharmacokinetic study of recombinant human anti-vascular
positive non-small-cell lung cancer. Ann Oncol 2004;15:19–27. endothelial growth factor in patients with advanced cancer. J Clin
245. Gebbia V, Del Prete S, Borsellino N, et al. Efficacy and safety of Oncol 2001;19:843–50.
cetuximab/irinotecan in chemotherapy-refractory metastatic col- 266. Gordon MS, Matei D, Aghajanian C, et al. Clinical activity of
orectal adenocarcinomas: a clinical practice setting, multicenter pertuzumab (rhuMAb 2C4), a HER dimerization inhibitor, in
experience.Clin Colorectal Cancer 2006;5:422–8. advanced ovarian cancer: potential predictive relationship
246. Geha RS. Regulation of the immune response by idiotype-anti- with tumor HER2 activation status. J Clin Oncol 2006;24:
idiotype interactions. N Engl J Med 1981; 305:25–8. 4324–32.
247. Geha RS. Idiotypic-anti-idiotypic interactions in humans. J Biol 267. Gori S, Colozza M, Mosconi AM, et al. Phase II study of weekly
Response Modif 1984; 3:573–9. paclitaxel and trastuzumab in anthracycline- and taxane-pre-
248. Gerber HP, Kowalski J, Sherman D, et al. Complete inhibition of treated patients with HER2-overexpressing metastatic breast can-
rhabdomyosarcoma xenograft growth and neovascularization cer. Br J Cancer 2004;90:36–40.
requires blockade of both tumor and host vascular endothelial 268. Gottlinger HG, Funke I, Hohnson JP, et al: The epithelial cell
growth factor. Cancer Res 2000;60:6253–58. surface antigen PANOREX, a target for antibody-mediated
249. Gertz MA, Rue M, Blood E, et al. Multicenter phase 2 trial of tumor therapy: its biochemical nature, tissue distribution and
rituximab for Waldenstrom macroglobulinemia (WM): an Eastern recognition by different monoclonal antibodies. Int J Cancer
Cooperative Oncology Group Study (E3A98). Leuk Lymphoma 1986;38:47–53.
2004;45:2047–55. 269. Goustin AS, Leof EB, Shipley GD, et al. Growth factors and can-
250. Gessner JE, Heiken H, Tamm A et al. The IgG Fc receptor family. cer. Cancer Res 1986; 46:1015–29.
Ann Hematol 1998;76:231–48. 270. Graeven U, Kremer B, Sudhoff T, et al. Phase I study of the
251. Ghielmini M, Schmitz SF, Cogliatti SB, et al. Prolonged treatment humanised anti-EGFR monoclonal antibody matuzumab (EMD
with rituximab in patients with follicular lymphoma significantly 72000) combined with gemcitabine in advanced pancreatic can-
increases event-free survival and response duration compared with cer. Br J Cancer 2006;94:1293–9.
the standard weekly x 4 schedule. Blood 2004;103:4416–23. 271. Green LL. Antibody engineering via genetic engineering of the
252. Giantonio BJ, Levy DE, O’dwyer PJ et al. A phase II study of mouse: XenoMouse strains are a vehicle for the facile generation
high-dose bevacizumab in combination with irinotecan, 5-fluo- of therapeutic human monoclonal antibodies. J Immunol Methods
rouracil, leucovorin, as initial therapy for advanced colorectal 1999;231:11–23.
cancer: results from the Eastern Cooperative Oncology Group 272. Greene WC, Leonard WJ, Depper JM, et al. The human interleu-
study E2200.Ann Oncol 2006;17:1399–403. kin-2 receptor: Normal and abnormal expression in T cells and in
253. Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in leukemias induced by the human T lymphotropic retroviruses.
combination with oxaliplatin, fluorouracil, and leucovorin Ann Intern Med 1986; 105:560–72.
(FOLFOX4) for previously treated metastatic colorectal cancer: 273. Gregory SA, Venugopal P, Adler S, et al. A phase II study of flu-
results from the Eastern Cooperative Oncology Group Study darabine phosphate and mitoxantrone followed by anti-CD20
E3200. J Clin Oncol 2007;25:1539–44. monoclonal antibody in the treatment of patients with newly
254. Gibson TB, Ranganathan A, Grothey A. Randomized phase III diagnosed advanced low-grade non-Hodgkin’s lymphoma:
trial results of panitumumab, a fully human anti-epidermal interim results. Blood 2003;102:412a [abstract 1499].
growth factor receptor monoclonal antibody, in metastatic col- 274. Greiner JW, Hand PH, Nugochi P, et al. Enhanced expression of
orectal cancer. Clin Colorectal Cancer 2006;6:29–31. surface tumor-associated antigens on human breast and colon
255. Giusti RM, Shastri KA, Cohen MH et al. FDA drug approval tumor cells after recombinant human leukocyte alpha-interferon
summary: panitumumab (Vectibix). Oncologist 2007;12:577–83. treatment. Cancer Res 1984; 44:3208–14.
256. Glassy MC, Handley HH, Hagiwara H, et al. UC729–6, a human 275. Greiner JW, Guadagni F, Goldstein D, et al. Intraperitoneal
lymphoblastoid B-cell line useful for generating antibody-secret- administration of interferon-gamma to carcinoma patients
ing human-human hybridomas. Proc Natl Acad Sci 1983; enhances expression of tumor-associated glycoprotein-72 and
80:6327–31. carcinoembryonic antigen on malignant ascites cells. J Clin
257. Glassy MC. Immortalization of human lymphocytes from a Oncol 1992; 10:735–46.
tumor-involved lymph node. Cancer Res 1987; 47:5181–8. 276. Habermann TM, Weller EA, Morrison VA, et al. Rituximab-
258. Glassy MC, Dillman RO. Molecular biotherapy with human CHOP versus CHOP alone or with maintenance rituximab in
monoclonal antibodies. Molec Biother 1988; 1:7–13. older patients with diffuse large B-cell lymphoma. J Clin Oncol
259. Gluck WL, Hurst D, Yuen A, et al. Phase I studies of interleukin 2006;24:3121–7.
(IL)-2 and rituximab in B-cell non-hodgkin’s lymphoma: IL-2 277. Hainsworth JD, Litchy S, Burris HA 3rd et al. Rituximab as first-
mediated natural killer cell expansion correlations with clinical line and maintenance therapy for patients with indolent non-
response. Clin Cancer Res 2004;10:2253–64. hodgkin’s lymphoma. J Clin Oncol. 2002;20:4261–7.
260. Golay J, Zaffaroni L, Vaccari T, et al. Biologic response of B 278. Hainsworth JD, Litchy S, Barton JH, et al. Single-agent ritux-
lymphoma cells to anti-CD20 monoclonal antibody rituximab in imab as first-line and maintenance treatment for patients with
vitro: CD55 and CD59 regulate complement-mediated cell lysis. chronic lymphocytic leukemia or small lymphocytic lymphoma:
Blood 2000;95:3900–8. a phase II trial of the Minnie Pearl Cancer Research Network.
261. Goodman GL, Beaumier P, Hellstrom I, et al. Pilot trial of murine J Clin Oncol 2003;21:1746–51.
monoclonal antibodies in patients with advanced melanoma. J 279. Hainsworth JD, Litchy S, Morrissey LH, et al. Rituximab plus
Clin Oncol 1985;3:340–52. short-duration chemotherapy as first-line treatment for follicular
Robert O. Dillman 393

non-Hodgkin’s lymphoma: a phase II trial of the Minnie Pearl 300. Heath JK, White SJ, Johnstone CN, et al. The human A33 antigen is
cancer research network. J Clin Oncol 2005a;23:1500–6. a transmembrane glycoprotein and a novel member of the immuno-
280. Hainsworth JD, Litchy S, Shaffer DW et al. Maximizing therapeu- globulin superfamily. Proc Natl Acad Sci U S A 1997;94:469–74.
tic benefit of rituximab: maintenance therapy versus re-treatment 301. Hebbar M, Wacrenier A, Desauw C, et al. Lack of usefulness of
at progression in patients with indolent non-Hodgkin’s lymphoma epidermal growth factor receptor expression determination for
– a randomized phase II trial of the Minnie Pearl Cancer Research cetuximab therapy in patients with colorectal cancer. Anticancer
Network. J Clin Oncol 2005b;23:1088–95. Drugs 2006;17:855–7.
281. Hainsworth JD, Sosman JA, Spigel DR, et al. Treatment of meta- 302. Hecht JR, Patnaik A, Berlin J, et al. Panitumumab monotherapy
static renal cell carcinoma with a combination of bevacizumab in patients with previously treated metastatic colorectal cancer.
and erlotinib. J Clin Oncol 2005c;23:7889–96. Cancer. 2007;110:980–8.
282. Haisma HJ, Pinedo HM, Kessel MAP, et al. Human IgM mono- 303. Hekman A, Honselaar A, Vuist WM, et al. Initial experience with
clonal antibody 16.88: Pharmacokinetics and immunogenicity in treatment of human B cell lymphoma with anti-CD19 monoclonal
colorectal cancer patients. J Natl Cancer Inst 1991; 83:1813–19. antibody. Cancer Immunol Immunother 1991;32:364–72.
283. Halaas JL, Moskowitz CH, Horwitz S, et al. R-CHOP-14 in 304. Hellstrom I, Garrigues U, Lavie E, et al. Antibody-mediated
patients with diffuse large B-cell lymphoma: feasibility and pre- killing of human tumor cells by attached effector cells. Cancer
liminary efficacy. Leuk Lymphoma 2005;46:541–7. Res 1988;48:624–7.
284. Hale G, Dyer M, Clark MR, et al. Remission induction in non- 305. Henney CS, Gillis S. Cell-mediated cytotoxicity. In: Paul WE,
Hodgkins lymphoma with reshaped human monoclonal antibody ed., Fundamental Immunology, pp New York, Raven Press, 1984,
CAMPATH-1H. Lancet 1988; 2:1394–99. pp669–684.
285. Hale G, Rebello P, Brettman LR, et al. Blood concentrations of 306. Hensel M, Villalobos M, Kornacker M, et al. Pentostatin/cyclo-
alemtuzumab and antiglobulin responses in patients with chronic phosphamide with or without rituximab: an effective regimen for
lymphocytic leukemia following intravenous or subcutaneous patients with Waldenstrom’s macroglobulinemia or lymphoplas-
routes of administration. Blood 2004;104:948–55. macytic lymphoma. Clin Lymphoma Myeloma 2005;6:131–5.
286. Halpern SE, Dillman RO. Radioimmunodetection with monoclo- 307. Heppner GH, Miller BE. Therapeutic implications of tumor het-
nal antibodies against prostatic acid phosphatase. In: Nuclear erogeneity. Semin Oncol 1989;16:91–105.
Medicine in Clinical Oncology. Winkler C, ed. Springer Verlag, 308. Herberman RB, Morgan AC, Reisfeld R, et al. Antibody-
Berlin, Heidelberg 1986; pp 164–70. dependent cellular cytotoxicity (ADCC) against human mela-
287. Halpern SE, Dillman RO, Witztum KF, et al. Radioimmunodetection noma by human effector cells in cooperation with mouse
of melanoma utilizing 111-In-96.5 monoclonal antibody: a pre- monoclonal antibodies. In: Monoclonal Antibodies and Cancer
liminary report. Radiology 1985; 155:493–9. Therapy. Reisfeld RA and Sell S, eds. A.R. Liss, Inc., New York.
288. Halpern SE, Haindl W, Beauregard J, et al. Scintigraphy with 1985; 27:193–203.
In-111-labeled monoclonal antitumor antibodies: kinetics, biodis- 309. Herbst RS, Arquette M, Shin DM, et al. Phase II multicenter study
tribution, and tumor detection. Radiology 1988; 168:529–36. of the epidermal growth factor receptor antibody cetuximab and
289. Halpern SE, Dillman RO, Amox D, et al. Detection of occult cisplatin for recurrent and refractory squamous cell carcinoma of
tumor using indium 111-labeled anticarcinoembryonic antigen the head and neck. J Clin Oncol 2005a;23:5578–87.
antibodies. Arch Surg 1992;127:1094–100. 310. Herbst RS, Johnson DH, Mininberg E, et al. Phase I/II trial evalu-
290. Haluska P, Shaw HM, Batzel GN, et al. Phase I dose escalation ating the anti-vascular endothelial growth factor monoclonal
study of the anti insulin-like growth factor-I receptor monoclonal antibody bevacizumab in combination with the HER-1/epidermal
antibody CP-751,871 in patients with refractory solid tumors. growth factor receptor tyrosine kinase inhibitor erlotinib for
Clin Cancer Res 2007;13:5834–40. patients with recurrent non-small-cell lung cancer. J Clin Oncol
291. Hamblin TJ, Cattan AR, Glennie MJ, et al. Initial experience in 2005b;23:2544–55.
treating human lymphoma with a chimeric univalent derivative 311. Herbst RS, Davies AM, Natale RB, et al. Efficacy and safety of
of monoclonal anti-idiotype antibody. Blood 1987; 69:790–7. single-agent pertuzumab, a human epidermal receptor dimeriza-
292. Han ES, Monk BJ. What is the risk of bowel perforation associ- tion inhibitor, in patients with non small cell lung cancer. Clin
ated with bevacizumab therapy in ovarian cancer? Gynecol Cancer Res 2007;13:6175–81.
Oncol 2007;105:3–6. 312. Herlyn D, Koprowski H. IgG2A monoclonal antibodies inhibit
293. Hanna N, Lilenbaum R, Ansari R, et al. Phase II trial of cetux- human tumor growth through interaction with effector cells. Proc
imab in patients with previously treated non-small-cell lung can- Natl Acad Sci USA 1982;79:4761–65.
cer. J Clin Oncol 2006;24:5253–8. 313. Herlyn D, Ross A, Koprowski H, et al. Anti-idiotypic antibodies
294. Harrison ML, Obermueller E, Maisey NR, et al. Tumor necrosis bear the internal image of a human tumor antigen. Science 1986;
factor alpha as a new target for renal cell carcinoma: two sequen- 232:100–2.
tial phase II trials of infliximab at standard and high dose. J Clin 314. Herlyn D, Wettendorff M, Schmoll E, et al. Anti-idiotype immu-
Oncol 2007;25:4542–9. nization of cancer patients: modulation of the immune response.
295. Hartmann F, Renner C, Jung W, et al. Treatment of refractory Proc Natl Acad Sci USA 1987;84:8055–59.
Hodgkin’s disease with an anti-CD16/CD30 bispecific antibody. 315. Herlyn DM, Steplewski Z, Herlyn MF, et al. Inhibition of growth
Blood 1998;91:2042–7. of colorectal carcinoma in nude mice by monoclonal antibody.
296. Hartmann F, Renner C, Jung W, et al. Anti-CD16/CD30 bispe- Cancer Res 1980;40:717–721.
cific antibody treatment for Hodgkin’s disease: role of infusion 316. Herlyn M, Steplewski Z, Herlyn D, et al: CO17–1A and related
schedule and costimulation with cytokines. Clin Cancer Res monoclonal antibodies: their production and characterization.
2001;7:1873–81. Hybridoma 1986,5:S3–S10.
297. Hartung G, Hofheinz RD, Dencausse Y, et al. Adjuvant therapy with 317. Herold M, Hass A, Srock S, et al. Rituximab added to first-line
edrecolomab versus observation in stage II colon cancer: a multi- mitoxantrone, chlorambucil, and prednisolone chemotherapy fol-
center randomized phase III study. Onkologie 2005;28:347–50. lowed by interferon maintenance prolongs survival in patients with
298. Haseman MK, Rosenthal SA, Polascik TJ. Capromab pendetide advanced follicular lymphoma: an East German Study Group
imaging of prostate cancer. Cancer Biother Radiopharm Hematology and Oncology Study. J Clin Oncol 2007;25:1986–92.
2000;15:131–40. 318. Hersey P, Schibeci SD, Townsend P, et al. Potentiation of lym-
299. Haspel MV, McCabe RP, Pomato N, et al. Generation of tumor phocyte responses by monoclonal antibodies to the ganglioside
cell-reactive human monoclonal antibodies using peripheral GD3. Cancer Res 1986;46:6083–90.
blood lymphocytes from actively immunized colorectal carci- 319. Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy
noma patients. Cancer Res 1985;45:3951–61. with rituximab added to the combination of cyclophosphamide,
394 Monoclonal antibody therapy

doxorubicin, vincristine, and prednisone (CHOP) significantly 339. Igarashi T, Ohtsu T, Fujii H, et al. Re-treatment of relapsed indo-
improves the outcome for patients with advanced-stage follicular lent B-cell lymphoma with rituximab. Int J Hematol
lymphoma compared with therapy with CHOP alone: results of a 2001;73:213–21.
prospective randomized study of the German Low-Grade 340. Imai K, Pellegrino MA, Wilson BS, et al. Higher cytolytic effi-
Lymphoma Study Group. Blood 2005;106:3725–32. ciency of an IgG2A than of an IgG1 monoclonal antibody with the
320. Hillmen P, Skotnicki AB, Robak T, et al. Alemtuzumab compared same (or spatially close) determinant on a human molecular-weight
with chlorambucil as first-line therapy for chronic lymphocytic melanoma-associated antigen. Cell Immunol 1982;72:239–47.
leukemia. J Clin Oncol 2007;25:5616–23. 341. Irie RF, Ollila DW, O’Day S, Morton DL. Phase I pilot clinical
321. Hjelm Skog A, Ragnhammar P, Fagerberg J, et al. Clinical effects trial of human IgM monoclonal antibody to ganglioside GM3 in
of monoclonal antibody 17–1A combined with granulocyte/mac- patients with metastatic melanoma. Cancer Immunol Immunother
rophage-colony-stimulating factor and interleukin-2 for treat- 2004;53:110–7.
ment of patients with advanced colorectal carcinoma. Cancer 342. Itala M, Geisler CH, Kimby E, et al: Standard-dose anti-CD20
Immunol Immunother 1999;48:463–70. antibody rituximab has efficacy in chronic lymphocytic leukae-
322. Hochster HS, Weller E, Ryan T, et al. Results of E1496: a pahse mia: results from a Nordic multicentre study. Eur J Haematol
III trial of CVP with or without maintenance rituximab in 2002;69:129–34.
advanced indolent lymphoma. Proc Am Soc Clin Oncol 343. Jacot W, Bessis D, Jorda E, et al. Acneiform eruption induced by
2004;23:556 [abstract 6502] epidermal growth factor receptor inhibitors inpatients with solid
323. Hofheinz RD, al-Batran SE, Hartmann F, et al. Stromal antigen tumours Br J Dermatol 2004;151:238–41.
targeting by a humanised monoclonal antibody: an early phase II 344. Jahanzeb M, Mortimer JE, Yunus F, et al. Phase II trial of weekly
trial of sibrotuzumab in patients with metastatic colorectal can- vinorelbine and trastuzumab as first-line therapy in patients with
cer. Onkologie 2003;26:44–8. HER2(+) metastatic breast cancer. Oncologist 2002;7:410–7.
324. Hofheinz RD, Horisberger K, Woernle C, et al. Phase I trial of 345. Jain RK. Physiological barriers to delivery of monoclonal anti-
cetuximab in combination with capecitabine, weekly irinotecan, bodies and other macromolecules in tumors. Cancer Res
and radiotherapy as neoadjuvant therapy for rectal cancer. Int J 1990;50:814s-9s.
Radiat Oncol Biol Phys 2006;66:1384–90. 346. James ND, Atherton PJ, Jones J, et al. A phase II study of the
325. Holliger P, Hudson PJ. Engineered antibody fragments and the bispecific antibody MDX-H210 (anti-HER2 x CD64) with
rise of single domains. Nat biotechnol 2005;23:1126–36. GM-CSF in HER2 + advanced prostate cancer. Br J Cancer
326. Hoogenboom HR, Chames P: Natural and designer binding sites 2001;85:152–6.
made by phage display technology. Immunol Today 2000; 21:371–8. 347. Janson D, Hoffman M, Fuchs A, et al. Campath 1-H therapy
327. Horning SJ, Rosenberg SA. The natural history of initially causes clearing of lymphocytic infiltration of bone marrow in
untreated low grade non-Hodgkin’s lymphoma. N Engl J Med advanced refractory chronic lymphocytic leukemia. Blood
1984;311:1471–5. 84(suppl 100):1994; 526a.
328. Houghton AN, Mintzer D, Cordon-Cardo C, et al. Mouse mono- 348. Jayson GC, Zweit J, Jackson A, et al. Molecular imaging and
clonal IgG3 antibody detecting GD3 ganglioside: a phase I trial biological evaluation of HuMV833 anti-VEGF antibody: impli-
in patients with malignant melanoma. Proc Natl Acad Sci USA cations for trial design of antiangiogenic antibodies. J Natl
1985;82:1242–6. Cancer Inst 2002;94:1484–93.
329. Howard OM, Gribben JG, Neuberg DS, et al. Rituximab and 349. Jensen AD, Munter MW, Bischoff H, et al. Treatment of non-small
CHOP induction therapy for newly diagnosed mantle-cell lym- cell lung cancer with intensity-modulated radiation therapy in
phoma: molecular complete responses are not predictive of combination with cetuximab: the NEAR protocol (NCT00115518).
progression-free survival. J Clin Oncol 2002;20:1288–94. BMC Cancer 20068;6:122.
330. Hu F, Epstein AL, Naeve GS, et al. A phase 1a clinical trial of 350. Jermann M, Jost LM, Taverna CH. Rituximab-EPOCH, an effec-
LYM-1 monoclonal antibody serotherapy in patients with refrac- tive salvage therapy for relapsed, refractory or transformed B-cell
tory B cell malignancies. Hematol Oncol 1989;7:155–166. lymphomas: results of a phase II study. Ann Oncol 2004;15 511–6.
331. Hudis CA. Trastuzumab – mechanisms of action and use in 351. Jerne NK. Idiotypic networks and other preconceived ideas.
clinical practice. N Engl J Med 2007;357–39–51. Immunol Rev 1984;79:5–24.
332. Huen J, Holen K. Treatment with panitumumab after a severe infu- 352. Ji SQ, Chen HR, Yan HM, et al. Anti-CD25 monoclonal antibody
sion reaction to cetuximab in a patient with metastatic colorectal (basiliximab) for prevention of graft-versus-host disease after
cancer: a case report. Clin Colorectal Cancer. 2007;6:529–31. haploidentical bone marrow transplantation for hematological
333. Huhn D, von Schilling C, Wilhelm M, et al. Rituximab therapy of malignancies. Bone Marrow Transplant 2005;36:349–54.
patients with B-cell chronic lymphocytic leukemia. Blood 353. Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant
2001;98:1326–31. docetaxel or vinorelbine with or without trastuzumab for breast
334. Hurley J, Doliny P, Reis I, et al. Docetaxel, cisplatin, and trastu- cancer. N Engl J Med 2006;354:809–20.
zumab as primary systemic therapy for human epidermal growth 354. Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized
factor receptor 2-positive locally advanced breast cancer. J Clin phase II trial comparing bevacizumab plus carboplatin and pacli-
Oncol 2006;24:1831–8. taxel with carboplatin and paclitaxel alone in previously untreated
335. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus locally advanced or metastatic non-small-cell lung cancer. J Clin
irinotecan, fluorouracil, and leucovorin for metastatic colorectal Oncol 2004;22:2184–91.
cancer. N Engl J Med 2004;350:2335–42. 355. Jonker DJ, O’Callaghan CJ, Karapetis et al. Cetuximab for the
336. Hurwitz HI, Fehrenbacher L, Hainsworth JD, et al. Bevacizumab treatment of colorectal cancer. N Engl J Med 2007;357:2040–8.
in combination with fluorouracil and leucovorin: an active regi- 356. Jubb AM, Hurwitz HI, Bai W, et al. Impact of vascular endothe-
men for first-line metastatic colorectal cancer. J Clin Oncol lial growth factor-A expression, thrombospondin-2 expression,
2005;23:3502–8. and microvessel density on the treatment effect of bevacizumab
337. Hussain MH, MacVicar GR, Petrylak DP, et al. Trastuzumab, in metastatic colorectal cancer. J Clin Oncol 2006;24:217–27.
paclitaxel, carboplatin, and gemcitabine in advanced human epi- 357. Junghans RP. Finally! The Bframbell receptor (FcRB). Mediator
dermal growth factor receptor-2/neu-positive urothelial carci- of transmission of immunity and protection from catabolism for
noma: results of a multicenter phase II National Cancer Institute IgG. Immunol Res 1997;16:27–57.
trial. J Clin Oncol 2007;25:2218–24. 358. Junghans RP, Waldmann TA, Landolfi NF, et al. Anti-tac-H, a
338. Hussein MA, Karam MA, McLain DA, et al. Biologic and clini- humanized antibody to the interleukin 2 receptor with new fea-
cal evaluation of rituxan in the management of newly diagnosed tures for immunotherapy in malignant and immune disorders.
multiple myeloma patients. Blood 1999;94:313a [abstract 1400]. Cancer Res 1990;50:1495–502.
Robert O. Dillman 395

359. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. Phase II, ran- 379. Kewalramani T, Zelenetz AD, Nimer SD et al. Rituximab and
domized trial comparing bevacizumab plus fluorouracil (FU)/ ICE as second-line therapy before autologous stem cell trans-
leucovorin (LV) with FU/LV alone in patients with metastatic plantation for relapsed or primary refractory diffuse large B-cell
colorectal cancer. J Clin Oncol 2003;21:60–5. lymphoma. Blood 2004;103:3684–8.
360. Kabbinavar FF, Hambleton J, Mass RD, et al. Combined analysis 380. Khan KD, Emmanouilides C, Benson DM Jr, et al. A phase 2
of efficacy: the addition of bevacizumab to fluorouracil/leuco- study of rituximab in combination with recombinant interleu-
vorin improves survival for patients with metastatic colorectal kin-2 for rituximab-refractory indolent non-Hodgkin’s lym-
cancer. J Clin Oncol 2005a;23:3706–12. phoma. Clin Cancer Res 2006;12:7046–53.
361. Kabbinavar FF, Schulz J, McCleod M, et al. Addition of bevaci- 381. Khazaeli MB, Saleh MN, Liu TP, et al. Pharmacokinetics and
zumab to bolus fluorouracil and leucovorin in first-line metastatic immune response of 131I-chimeric mouse/human B72.3 (human γ)
colorectal cancer: results of a randomized phase II trial. J Clin monoclonal antibody in humans. Cancer Res 1991;51:5461–6.
Oncol. 2005b;23:3697–705. 382. Khazaeli MB, Conry RM, LoBuglio AF. Human immune response
362. Kaminski MS, Tuck M, Estes J, et al. 1311-tositumomab therapy to monoclonal antibodies. J Immunother 1994;15:42–52.
as initial treatment for follicular lymphoma. N Engl J Med 383. Khorana A, Bunn P, McLaughlin P, et al. A phase II multicenter
2005;352:441–9. study of CAMPATH-1H antibody in previously treated patients
363. Kanelli S Ansell SM, Habermannn TM, et al. Rituximab toxicity with nonbulky non-Hodgkin’s lymphoma. Leuk Lymphoma
in patients with peripheral blood malignant B-cell lymphocyto- 2001;41:77–87.
sis. Leuk Lymphoma 2001;42:1329–37. 384. Kiewe P, Hasmüller S, Kahlert S, et al. Phase I trial of the trifunc-
364. Kan-Mitchell J, Iman A, Kempf RA, et al. Human monoclonal tional anti-HER2 x anti-CD3 antibody ertumaxomab in meta-
antibodies directed against melanoma tumor-associated antigens. static breast cancer. Clin Cancer Res 2006;12:3085–91.
Cancer Res 1986;46:2490–96. 385. Kim DH, Jung HD, Kim JG, et al. FCGR3A gene polymorphisms
365. Kaplan LD, Lee JY, Ambinder RF et al. Rituximab does not may correlate with response to frontline R-CHOP therapy for dif-
improve clinical outcome in a randomized phase 3 trial of CHOP fuse large B-cell lymphoma. Blood 2006;108:2720–5.
with or without rituximab in patients with HIV-associated non- 386. Kim SJ, Park Y, Hong HJ. Antibody engineering for the develop-
Hodgkin lymphoma: AIDS-Malignancies Consortium Trial 010. ment of therapeutic antibodies. Mol Cell 2005;20:17–29.
Blood 2005;106:1538–43. 387. Kim YH, Duvic M, Obitz E, et al. Clinical efficacy of zanoli-
366. Karagiannis SN, Bracher MG, Hung J, et al. IgE-antibody- mumab (HuMax-CD4): two phase 2 studies in refractory cutane-
dependent immunotherapy of solid tumors: cytotoxic and phago- ous T-cell lymphoma. Blood 2007;109:4655–62.
cytic mechanisms of eradication of ovarian cancer cells. J 388. Kim YS, Maslinski W, Zheng XX, Schachter AD, Strom TB.
Immunol 2007;179:2832–43. Immunoglobulin-cytokine fusion molecules: the new generation of
367. Katzenwadel A, Schleer H, Gierschner D, et al. Construction and immunomodulating agents. Transplant Proc 1998;30:4031–4036.
in vivo evaluation of an anti-PSA x anti-CD3 bispecific antibody 389. Kimby E, Geisler C, Hagbert H, et al. Rituximab (Mabthera®) as
for the immunotherapy of prostate cancer. Anticancer Res single agent and in combination with interferon-α-2a as treatment
2000;20:1551–5. of untreated and first relapse follicular or other low-grade lym-
368. Kaufmann H, Raderer M, Wohrer S, et al. Antitumor activity of phomas. A randomized phase II study M 39035. Blood
rituximab plus thalidomide in patients with relapsed/refractory 2000;96:577a [abstract 2479].
mantle cell lymphoma. Blood 2004;104:2269–71. 390. Kindler HL, Friberg G, Singh DA, et al. Phase II trial of bevaci-
369. Kawai S, Yoshimura Y, Iida S, et al. Antitumor activity of human- zumab plus gemcitabine in patients with advanced pancreatic
ized monoclonal antibody against HM1.24 antigen in human cancer. J Clin Oncol 2005;23:8033–40.
myeloma xenograft models. Oncol Rep 2006 15:361–7. 391. Kindler HL, Niedzwiecki D, Hollis E, et al. A double-blind, pla-
370. Kay NE, Geyer SM, Call TG, et al. Combination chemother- cebo-controlled, randomized phase III trial of gemcitabine plus
apy with pentostatin, cyclophosphamide, and rituximab shows bevacizumab versus gemcitabine plus placebo in patients with
significant activity with low accompanying toxicity in previ- advanced pancreatic cancer: a preliminary analysis of Cancer and
ously untreated B chronic lymphocytic leukemia. Blood Leukemia Group B. Proc Am Soc Clin Oncol 2007;25:1992
109:405–11, 2007. [abstract 4508].
371. Keating MJ, Cazin B, Coutre S, et al. Campath-1H treatment of 392. Kirkwood JM, Neumann RD, Zoghbi SS, et al. Scintigraphic detec-
T-cell prolymphocytic leukemia in patients for whom at least one tion of metastatic melanoma using Indium-111/DTPA conjugated
prior chemotherapy regimen has failed. J Clin Oncol anti-gp240 antibody [ZME018]. J Clin Oncol 1987; 5:1247–1255.
2002a;20:205–13. 393. Kirkwood JM, Mascari RA, Edington HD, et al. Analysis of
372. Keating MJ, Flinn I, Jain V, et al. Therapeutic role of alemtu- therapeutic and immunologic effects of R(24) anti-GD3 mono-
zumab (Campath-1H) in patients who have failed fludarabine: clonal antibody in 37 patients with metastatic melanoma. Cancer
results of a large international study. Blood 2002b;99:3554–61. 2000;88:2693–702.
373. Keating MJ, O’Brien S, Albitar M, et al. Early results of a 394. Kiyotaki M, Cooper MD, Bertoli LF, et al. Monoclonal anti-Id
chemoimmunotherapy regimen of fludarabine, cyclophosph- antibodies react with varying proportions of human B lineage
amide, and rituximab as initial therapy for chronic lymphocytic cells. J Immunol 1987;138:4150–4158.
leukemia. J Clin Oncol 2005;23:4079–88. 395. Kjeldson TB, Rasmussen BB, Rose C, Zeuthen J. Human-human
374. Keefe DL. Trastuzumab-associated cardiotoxicity. Cancer hybridomas and human monoclonal antibodies obtained by
2002;95:1592–600. fusion of lymph node lymphocytes from breast cancer patients.
375. Kelley MJ, Linnoila RI, Avis IL, et al. Antitumor activity of a Cancer Res 1988;48:3208–14.
monoclonal antibody directed against gastrin-releasing peptide in 396. Klein B, Wijdenes J, Zhang XG, et al. Murine anti-interleukin 6
patients with small cell lung cancer. Chest 1997;112:256–61. monoclonal antibody therapy for a patient with plasma cell leu-
376. Kelly H, Kimmick G, Dees EC, et al. Response and cardiac toxic- kemia. Blood 1991;78:1198–204.
ity of trastuzumab given in conjunction with weekly paclitaxel 397. Knox SJ, Levy R, Hodgkinson S, et al. Observations on the effect
after doxorubicin/cyclophosphamide. Clin Breast Cancer of chimeric anti-CD4 monoclonal antibody in patients with
2006;7:237–43. mycosis fungoides. Blood 1991;77:20–30.
377. Kennedy GA, Seymour JF, Wolf M, et al. Treatment of patients 398. Koda K, Nakajima N, Saito N, et al. A human natural antibody to
with advanced mycosis fungoides and Sezary syndrome with adenocarcinoma that inhibits tumour cell migration. Br J Cancer.
alemtuzumab. Eur J Haematol 2003;71:250–6. 1998;78:1313–22.
378. Kennedy RC, Zhou EM, Lanford RE, et al. Possible role of anti- 399. Koda K, Glassy MC, McKnight ME, et al. Immunotherapy for
idiotypic antibodies in the induction of tumor immunity. J Clin recurrent colorelctal cancers with human monoclonal antibody
Invest 1987;80:1217–24. SK-1. Anticancer Res 2001;21:621–7.
396 Monoclonal antibody therapy

400. Kohler G, Milstein C. Continuous cultures of fused cells secreting 422. Lencer WI, Blumberg RS. A passionate kiss, then run: exocytosis
antibody of predetermined specificity. Nature 1975;256:495–597. and recycling of IgG by FcRn. Trends Cell Biol. 2005;15:5–9.
401. Kollmannsberger C, Schittenhelm M, Honecker F,et al.A phase I 423. Lenz G, Dreyling M, Hoster E, et al. Immunochemotherapy with
study of the humanized monoclonal anti-epidermal growth factor rituximab and cyclophosphamide, doxorubicin, vincristine, and
receptor (EGFR) antibody EMD 72000 (matuzumab) in combina- prednisone significantly improves response and time to treatment
tion with paclitaxel in patients with EGFR-positive advanced non- failure, but not long-term outcome in patients with previously
small-cell lung cancer (NSCLC). Ann Oncol 2006;17:1007–13. untreated mantle cell lymphoma: results of a prospective ran-
402. Koon HB, Jurghans RP. Anti-CD30 antibody-based therapy. Curr domized trial of the German Low Grade Lymphoma Study Group
Opin Oncol 2000;12:588–93. (GLSG). J Clin Oncol 2005;23:1984–92.
403. Koon HB, Severy P, Hagg DS, et al. Antileukemic effect of dacli- 424. Lenz HJ, Van Cutsem E, Khambata-Ford S, et al. Multicenter
zumab in CD25 high-expressing leukemias and impact of tumor phase II and translational study of cetuximab in metastatic col-
burden on antibody dosing. Leuk Res 2006;30:190–203. orectal carcinoma refractory to irinotecan, oxaliplatin, and fluo-
404. Koprowski H, Herlyn D, Lubeck M, et al. Human anti-idiotype ropyrimidines. J Clin Oncol 2006;24:4914–21.
antibodies in cancer patients: is the modulation of the immune 425. Leonard JP, Coleman M, Ketas JC, et al. Phase I/II trial of epratu-
response beneficial for the patient? Proc Natl Acad Sci USA zumab (humanized anti-CD22 antibody) in indolent non-Hodg-
1984;81:216–9. kin’s lymphoma. J Clin Oncol 2003;21:3051–9.
405. Kosmos C, Epenetos AE, Colurtenay-Luck NS. Activation of 426. Leonard JP, Coleman M, Ketas J, et al. Combination antibody
cellular immunity after intracavitary monoclonal antibody ther- therapy with epratuzumab and rituximab in relapsed or refractory
apy of ovarian cancer. Cancer 1994;73:3000–10. non-Hodgkin’s lymphoma. J Clin Oncol 2005;23:5044–51.
406. Kossman SE, Scheinberg DA, Jurcic JG, et al. A phase I trial of 427. Leroy M. Teillac P, Rain JD, et al. Radioimmunodetection of
humanized monoclonal antibody HuM195 (anti-CD33) with lymph node invasion in prostatic cancer. Cancer 1989; 64:1–5.
low-dose interleukin-2 in acute myelogenous leukemia. Clin 428. Levy R. Will monoclonal antibodies find a place in our therapeu-
Cancer Res 1999;5:2748–2755. tic armamentarium? J Clin Oncol 1987;5:527–9.
407. Krance RA, Kuehnle I, Rill DR, et al. Hematopoietic and immuno- 429. Levy R, Miller RA. Biological and clinical indications of lym-
modulatory effects of lytic CD45 Mab in patients with hematologic phocyte hybridomas: tumor therapy with monoclonal antibodies.
malignancy. Biol Blood Marrow Transplant 2003;9:273–81. Ann Rev Med 1983;34:107–16.
408. Krempien R, Muenter MW, Huber PE, et al. Randomized phase 430. Lewiecki EM. RANK ligand inhibition with denosumab
II-study evaluating EGFR targeting therapy with cetuximab in for the management of osteoporosis. Expert Opin Biol Ther
combination with radiotherapy and chemotherapy for patients 2006;6:1041–50.
with locally advanced pancreatic cancer – PARC: study protocol. 431. Leyland-Jones B, Gelmon K, Ayoub JP, et al. Pharmacokinetics,
BMC Cancer 2005;5:131. safety, and efficacy of trastuzumab administered every three weeks
409. Krug LM, Miller VA, Patel J, et al. Randomized phase II study of in combination with paclitaxel. J Clin Oncol 2003;21:3965–71.
weekly docetaxel plus trastuzumab versus weekly paclitaxel plus 432. Liao S, Kwong PC, Khosravi M, Dent PB. Enhanced expression
trastuzumab in patients with previously untreated advanced of melanoma-associated antigens and B2-microglobulin on cul-
nonsmall cell lung carcinoma. Cancer 2005;104:2149–55. tured human melanoma cells by interferon. J Natl Cancer Inst
410. Krug LM, Milton DT, Jungbluth AA, et al. Targeting Lewis Y 1982;68:19–25.
(Le(y) ) in small cell lung cancer with a humanized monoclonal 433. Liao SK, Meranda C, Avner BP, et al. Immunohistochemical phe-
antibody, hu3S193: a pilot trial testing two dose levels. J Thorac notyping of human solid tumors with monoclonal antibodies in
Oncol 2007;2:947–52. devising biotherapeutic strategies. Cancer Immunol Immunother
411. Kushner BH, Kramer K, Cheung NK. Phase II trial of the anti- 1989; 28:77–86.
G(D2) monoclonal antibody 3F8 and granulocyte-macrophage 434. Liljefors M, Ragnhammar P, Nilsson B, et al. Anti-EpCAM
colony-stimulating factor for neuroblastoma. J Clin Oncol monoclonal antibody (MAb17–1A) based treatment combined
2001;19:4189–94. with alpha-interferon, 5-fluorouracil and granulocyte-mac-
412. Lam JS, Pantuck AJ, Belldegrun AS, Figlin RA. G250: a car- rophage colony-stimulating factor in patients with metastatic
bonic anhydrase IX monoclonal antibody. Curr Oncol Rep colorectal carcinoma. Int J Oncol 2004;25:703–11.
2005;7:109–15. 435. Lim LC, Koh LP, Tan P. Fatal cytokine release syndrome with
413. Lamanna N, Kalaycio M, Maslak P, et al. Pentostatin, cyclophos- chimeric anti-CD20 monoclonal antibody rituximab in a 71-year
phamide, and rituximab is an active, well-tolerated regimen for old patient with chronic lymphocytic leukemia. J Clin Oncol
patients with previously treated chronic lymphocytic leukemia. 1999;17:1962–3.
J Clin Oncol 2006;24:1575. 436. Lin TS, Flinn IW, Lucas MS, et al. Filgrastim and alemtuzumab
414. Langer LF, Clay TM, Morse MA. Update on anti-CTLA-4 anti- (Campath-1H) for refractory chronic lymphocytic leukemia.
bodies in clinical trials. Expert Opin Biol Ther 2007;7:1245–56. Leukemia 2005a;19:1207–10.
415. Lara PN Jr, Laptalo L, Longmate J, et al. Trastuzumab plus doc- 437. Lin TS, Flinn IW, Modali R, et al. FCGR3A and FCGR2A poly-
etaxel in HER2/neu-positive non-small-cell lung cancer: a morphisms may not correlate with response to alemtuzumab in
California Cancer Consortium screening and phase II trial. Clin chronic lymphocytic leukemia. Blood 2005b;105:289–91.
Lung Cancer 2004;5:231–6. 438. Lindstrom BA. An experimental study of myelotoxic sera.
416. Larrick JW, Bourla JM. Prospects for therapeutic use of human Therapeutic attempts in myeloid leukaemia. Acta Med Scand
monoclonal antibodies. J Biol Response Mod 1986;5:379–93. Suppl 1927;22:1769–75.
417. Larson SM, Brown, JP, Wright PW, et al. Imaging of melanoma with 439. Lipton A, Steger GG, Figueroa J, et al. Randomized active-con-
I-131-labeled monoclonal antibodies. J Nucl Med 1982; 24:123–9. trolled phase II study of denosumab efficacy and safety in patients
418. Lauria F, Lenoci M, Annino L, et al. Efficacy of Mabthera (anti- with breast cancer-related bone metastases. J Clin Oncol
CD20 Ab) in patients with relapsed/progressed hairy cell leuke- 2007;25:4431–7.
mia. Blood 2000:96:138a [abstract 592] 440. Liu AY, Robinson RR, Hellstrom AE, et al. Chimeric-mouse-
419. Leder P. The genetics of antibody diversity. Sci Amer 1980; human IgG1 antibody that can mediate lysis of cancer cells. Proc
243:102–15. Natl Acad Sci USA 1987a; 84:3439–3443.
420. Lee CG, Heijn M, di Tomaso E, et al. Anti-vascular endothelial 441. Liu AY, Robinson RR, Murray Ed, et al. Production of a mouse-
growth factor treatment augments tumor radiation response under human chimeric monoclonal antibody to CD20 with potent
normoxic or hypoxic conditions. Cancer Res 2000;60:5565–70. Fc-dependent biologic activity. J Immunol 1987b;139:3521–6.
421. Lee KF, Simon H, Chen H, et al. Requirement for neuregulin receptor 442. Lobo Ed, Hansen RJ, Balthasar JP. Antibody pharmacokinetics
erbB2 in neural and cardiac development. Nature 1995;378:394–8. and pharmacodynamics. J Pharm Sci 2004;93:2645–68.
Robert O. Dillman 397

443. LoBuglio AF, Saleh MN, Lee J, et al. Phase I trial of multiple with advanced or recurrent non-squamous non-small cell lung
large doses of murine monoclonal antibody CO17–1A. Clinic cancer. J Clin Oncol 2007;25 (18s part II) [abstract LBA7514].
aspects. J Natl Cancer Inst 1988; 17:932–6. 464. Marcom PK, Isaacs C, Harris L, et al. The combination of letro-
444. LoBuglio AF, Wheeler RH, Trang J, et al. Mouse/human chime- zole and trastuzumab as first or second-line biological therapy
ric monoclonal antibody in man: kinetics and immune response. produces durable responses in a subset of HER2 positive and ER
Proc Natl Acad Sci USA 1989;86:4220–4. positive advanced breast cancers. Breast Cancer Res Treat
445. Lofgren JA, Dhandapani S, Pennuccii JJ, et al. Comparing ELISA 2007;102:43–9.
and surface plasmon resonance for assessing clinical immunoge- 465. Marcus R, Imrie K, Belch A, et al. CVP chemotherapy plus ritux-
nicity of panitumumab. J Immunol 2007;178:7467–72. imab compared with CVP as first-line treatment for advanced
446. Lowder JN, Meeker TC, Campbell M, et al. Studies on B lym- follicular lymphoma. Blood 2005;105:1417–23.
phoid tumors treated with monoclonal anti-idiotype antibodies: 466. Margolin K, Gordon MS, Holmgren E, et al. Phase Ib trial of
Correlation with clinical responses. Blood 1987; 69:199–210. intravenous recombinant humanized monoclonal antibody to
447. Lubeck MD, Steplewski Z, Baglia F, et al. The interaction of vascular endothelial growth factor in combination with chemo-
murine IgG subclass proteins with human monocyte Fc recep- therapy inpatients with advanced cancer: pharmacologic and
tors. J Immunol 1985; 135:1299–304. long-term safety data. J Clin Oncol 2001;19:851–6.
448. Lundin J, Osterborg A, Brittinger G, et al. CAMPATH1-H mono- 467. Marini P. Drug evaluation: lexatumumab, an intravenous human
clonal antibody in therapy for previously treated low-grade non- agonistic mAb targeting TRAIL receptor 2. Curr Opin Mol Ther
Hodgkin’s lymphomas: a phase II multicenter study. J Clin Oncol 2006;8:539–46.
1998;16:3257–63. 468. Martinelli G, Laszlo D, Bertolini F, et al. Chlorambucil in combi-
449. Lundin J, Kimby E, Bjorkholm M, et al. Phase II trial of subcuta- nation with induction and maintenance rituximab is feasible and
neous anti-CD52 monoclonal antibody alemtuzumab (Campath-1H) active in indolent non-Hodgkin’s lymphoma. Br J Haematol
as first-line treatment for patients with B-cell chronic lymphocytic 2003;123:271–7.
leukemia (B-CLL). Blood 2002;100:768–73. 469. Martinelli G, Laszlo D, Ferreri AJ, et al. Clinical activity of ritux-
450. Lundin J, Hagberg H, Repp R, et al. Phase 2 study of alemtuzumab imab in gastric marginal zone non-Hodgkin’s lymphoma resis-
(anti-CD52 monoclonal antibody) in patients with advanced myco- tant to or not eligible for anti-Helicobacter pylori therapy. J Clin
sis fungoides/Sezary syndrome. Blood 2003;101:4267–72. Oncol 2005;23:1979–83.
451. Mach J-P, Chatal J-F, Lumbroso J-D, et al. Tumor localization in 470. Marty M, Cognetti F, Maraninchi D, et al. Randomized phase II
patients by radiolabeled monoclonal antibodies against colon trial of the efficacy and safety of trastuzumab combined with
carcinoma. Cancer Res 1983; 43:5593–600. docetaxel in patients with human epidermal growth factor
452. Machiels JP, Sempoux C, Scalliet P, et al. Phase I/II study of pre- receptor 2-positive metastatic breast cancer administered as
operative cetuximab, capecitabine, and external beam radiother- first-line treatment: the M77001 study group. J Clin Oncol
apy in patients with rectal cancer. Ann Oncol 2007;18:738–44. 2005;23:4265–74.
453. Maguire HC Jr, Berd D, Lattime EC, et al. Phase I study of R24 in 471. Masui H, Kawamoto T, Sato JD, et al. Growth inhibition of
patients with metastatic melanoma including evaluation of immu- human tumor cells in athymic mice by anti-epidermal growth
nologic parameters. Cancer Biother Radiopharm 1998;13:13–23. factor receptor monoclonal antibodies. Cancer Res 1984;
454. Maguire RT, VanNostrand D. Diagnosis of colorectal and ovarian 44:1002–1007.
carcinoma. New York: Marcel Dekker Inc, 1992. 472. Masui H, Kamrath H, Apell G, et al. Cytotoxicity against human
455. Maker AV, Phan GQ, Attia P, et al. Tumor regression and autoim- tumor cells mediated by the conjugate of anti-epidermal growth
munity in patients treated with cytotoxic T lymphocyte-associ- factor receptor monoclonal antibody to recombinant ricin A
ated antigen 4 blockade and interleukin 2: a phase I/II study. Ann chain. Cancer Res 1989;49:3482–8.
Surg Oncol 2005;12:1005–16. 473. Mathas S, Rickers A, Bommert K, et al. Anti-CD20- and B-cell
456. Maker AV, Yang JC, Sherry RM, et al. Intrapatient dose escala- receptor –mediated apoptosis: evidence for shared intracellular
tion of anti-CTLA-4 antibody in patients with metastatic mela- signaling pathways. Cancer Res 2000;60:7170–6.
noma. J Immunother 2006;29:455–63. 474. McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chi-
457. Makower D, Sparano JA, Wadler S, et al. A pilot study of edreco- meric anti-CD20 monoclonal antibody therapy for relapsed indo-
lomab (Panorex, 17–1A antibody) and capecitabine in patients lent lymphoma: half of patients respond to a four-dose treatment
with advanced or metastatic adenocarcinoma. Cancer Invest program. J Clin Oncol 1998;16:2825–33.
2003;21:177–84. 475. McLaughlin P, Rodriguez MA, Reuben J, et al. Rituximab in
458. Maloney DG, Levy R, Miller RA. Monoclonal antiidiotype ther- combination with GM-CSF (Leukine) for patients with recurrent
apy of B cell lymphoma. Biological Therapy of Cancer Updates indolent lymphoma. Blood 2001;98:605a, [abstract #2536].
1992;6:1–10. 476. McLaughlin P, Hagemeister FB, Romaguera J, et al. Stage IV
459. Maloney DG, Liles TM, Czerwinski DK, et al. Phase I clinical indolent lymphoma: a randomized study of concurrent vs.
trial using escalating single dose infusion of chimeric monoclo- sequential use of FND chemotherapy (fludarabine, mitoxantrone,
nal antibody (IDEC C2B8) in patients with recurrent B-cell lym- dexamethasone) and rituximab monoclonal antibody therapy,
phoma. Blood 1994;84:2457–66 with interferon maintenance. Proc Am Soc Clin Oncol
460. Maloney DG, Grillo-Lopez AJ, Bodkin DJ, et al. IDEC-C2B8: 2003;22:564 [abstract 2269].
results of a phase I multiple-dose trial in patients with relapsed 477. McNeel DG, Eickhoff J, Lee FT, et al. Phase I trial of a monoclo-
non-Hodgkin’s lymphoma. J Clin Oncol 1997a;15:3266–74. nal antibody specific for alphavbeta3 integrin (MEDI-522) in
461. Maloney DG, Grillo-Lopez AJ, White CA, et al. IDEC-C2B8 patients with advanced malignancies, including an assessment of
(Rituximab) anti-CD20 monoclonal antibody therapy in patients effect on tumor perfusion. Clin Cancer Res 2005;11:7851–60.
with relapsed low-grade non-Hodgkin’s lymphoma. Blood 478. Meeker T, Lowder J, Cleary ML, et al. Emergence of idiotype
1997b;90:2188–95. variants during treatment of B-cell lymphoma with anti-idiotype
462. Maloney DG, Press OW, Braziel RM, et al. A phase II trial of antibodies. Blood 1985; 65:1373–1381.
CHOP followed by rituximab chimeric monoclonal anti-CD20 479. Meeker TC, Lowder J, Maloney DG, et al. A clinical trial of anti-
antibody for treatment of newly diagnosed follicular non-Hodg- idiotype therapy for B cell malignancy. Blood 1985;65:1349–72.
kin’s lymphoma: SWOG 9800. Blood 2001;98:843a [abstract 480. Mendelsohn J. Blockade of receptors for growth factors: an anti-
3502]. cancer therapy. Clin Cancer Res 2000;6:747–753.
463. Manegold C, von Pawel J, Zatoukal P et al. Randomised, double- 481. Mersmann M, Schmidt A, Rippman JF, et al. Human antibody
blind multicenter phase III study of bevacizumab in combination derivatives against the fibroblast activation protein for tumor
with cisplatin and gemcitabine in chemotherapy-naïve patients stroma targeting of carcinomas. Int J Cancer 2001;92:240–8.
398 Monoclonal antibody therapy

482. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bavacizumab and high-dose melphalan induces high complete response reates
versus paclitaxel alone for metastatic breast cancer. N Engl J Med in advanced multiple myeloma. Br J Haematol 2000;109:661–4.
2007;357:2666–76. 502. Moreno M, Bontkes HJ, Scheper RJ et al. High level of MUC1 in
483. Miller KD, Sisk J, Ansari R, et al. Gemcitabine, paclitaxel, and serum of ovarian and breast cancer patients inhibits huHMFG-1
trastuzumab in metastatic breast cancer. Oncology 2001;15(2 dependent cell-mediated cytotoxicity (ADCC). Cancer Lett
Suppl 3):38–40. 2007;257:47–55.
484. Miller KD, Chap LI, Holmes FA, et al. Randomized phase III 503. Moreton P, Kennedy B, Lucas G, et al. Eradication of minimal
trial of capecitabine compared with bevacizumab plus capecit- residual disease in B-cell chronic lymphocytic leukemia after
abine in patients with previously treated metastatic breast cancer. alemtuzumab therapy is associated with prolonged survival. J
J Clin Oncol 2005;23:792–9. Clin Oncol 2005;23:2971–9.
485. Miller RA, Levy R. Response of cutaneous T cell lymphoma to ther- 504. Moroni M, Veronese S, Benvenuti S, et al. Gene copy number for
apy with hybridoma monoclonal antibody. Lancet 1981; 2:226–30. epidermal growth factor receptor (EGFR) and clinical response
486. Miller RA, Maloney DG, McKillop J, Levy R, et al. In vivo to antiEGFR treatment in colorectal cancer: a cohort study.
effects of murine hybridoma monoclonal antibody in a patient Lancet Oncol 2005;6:279–86.
with T-cell leukemia. Blood 1981; 58:78–86. 505. Morris JC, Janik JE, White JD, et al. Preclinical and phase I clini-
487. Miller RA, Maloney DG, Warnke R, et al. Treatment of B-cell cal trial of blockade of IL-15 using Mikbeta1 monoclonal anti-
lymphoma with monoclonal anti-idiotype antibody. N Engl J body in T cell large granular lymphocyte leukemia. Proc Natl
Med 1982;306:517–22. Acad Sci U S A 2006;103:401–6.
488. Miller RA, Oseroff AR, Stratte PT, et al. Monoclonal antibody 506. Morris MJ, Divgi Cr, Pandit-Taskar N et al. Pilot trial of unla-
therapeutic trials in seven patients with T-cell lymphoma. Blood beled and indium-111-labeled anti-prostate-specific membrane
1983;62:988–95. antigen antibody J591 for castrate metastatic prostate cancer.
489. Miller RA, Hart S, Samoszuk M, et al. Shared idiotypes expressed Clin Cancer Res 2005;11:7454–61.
by human B-cell lymphomas. N Engl J Med 1989; 321:851–7. 507. Morris MJ, Pandit-Taskar N, Divgi CR, et al. Phase I evaluation
490. Milowsky MI, Nanus DM, Kostakoglu L, et al. Vascular targeted of J591 as a vascular targeting agent in progressive solid tumors.
therapy with anti-prostate-specific membrane antigen monoclonal Clin Cancer Res 2007;13:2707–13.
antibody J591 in advanced solid tumors. J Clin Oncol 508. Morrison SL, Oi T. Genetically engineered antibody molecules.
2007;25:540–7. Adv Immunol 1989; 44:65–92.
491. Minasian LM, Szatrowski TP, Rosenblum M, et al. Hemorrhagic 509. Motzer RJ, Amato R, Todd M, et al. Phase II trial of antiepidermal
tumor necrosis during a pilot trial of tumor necrosis factor-a and growth factor receptor antibody C225 in patients with advanced
anti-GD3 ganglioside monoclonal antibody in patients with met- renal cell carcinoma. Invest New Drugs 2003;21:99–101.
astatic melanoma. Blood 1994; 83:56–64. 510. Mountain A, Adair JR: Engineering antibodies for therapy.
492. Minasian LM, Yao TJ, Steffens TA, et al. A phase I study of anti- Biotechnol Genet Eng Rev 1992;10:1–142.
GD3 ganglioside monoclonal antibody R24 and recombinant 511. Mujoo K, Kipps TJ, Yang HM, et al. Functional properties and
human macrophage-colony stimulating factor inpatients with effect on growth suppression of human neuroblastoma tumors by
metastatic melanoma. Cancer 1995;75:2251–7. isotype switch variants of monoclonal antiganglioside GD2 anti-
493. Mittelman A, Chen ZJ, Kageshita T, et al. Active specific immu- body 14.18. Cancer Res 1989;49:2857–61.
notherapy in patients with melanoma: A clinical trial with mouse 512. Mullamitha SA, Ton NC, Parker GJ, et al. Phase I evaluation of a
antiidiotypic monoclonal antibodies elicited with synergeneic fully human anti-alphav integrin monoclonal antibody (CNTO
anti-high-molecular-weight melanoma-associated antigen mono- 95) in patients with advanced solid tumors. Clin Cancer Res
clonal antibodies. J Clin Invest 1990;86:2136–44. 2007;13:2128–35.
494. Mittelman A, Chen ZJ, Yang H, et al. Human high molecular 513. Mulshine JL, Avis I, Treston AM, et al. Clinical use of a mono-
weight melanoma-associated antigen (HMW-MAA) mimicry by clonal antibody to bombesin-like peptide in patients with lung
mouse anti-idiotypic monoclonal antibody MK2–23: Induction cancer. Ann NY Acad Sci 1988;547:360–72.
of humoral anti-HMW-MAA immunity and prolongation of sur- 514. Murray JL, Rosenblum MG, Sobol RE, et al. Radioimmunoimaging
vival in patients with stage IV melanoma. Proc Natl Acad Sci in malignant melanoma with 111-In-labeled monoclonal anti-
USA 1992;89:466–70. body 96.5. Cancer Res 1985;45:2376–81.
495. Mohsin Sk, Weiss HL, Gutierrez MC, et al. Neoadjuvant trastu- 515. Murray JL, Rosenblum MG, Lamki K, et al. Clinical parameters
zumab induces apoptosis in primary breast cancers. J Clin Oncol related to optimal tumor localization of Indium-111-labeled
2005;23:2460–8. mouse antimelanoma monoclonal antibody ZME018. J Nucl
496. Molodofsky PJ, Sears HF, Mulhearn Jr CB, et al. Detection of Med 1987;28:25–33.
metastatic tumor in normal sized retroperitoneal lymph nodes by 516. Murray JL, Cunningham JE, Brewer H, et al. Phase I trial of
monoclonal antibody imaging. N Engl J Med 1984;311:106–7. murine monoclonal antibody 14G12 administered by prolonged
497. Molthoff CF, Prinssen HM, Kenemans P, et al. Escalating protein intravenous infusion in patients with neuroectodermal tumor. J
doses of chimeric monoclonal antibody Mov 18 immunoglobulin Clin Oncol 1994;12:184–93.
G in ovarian carcinoma patients: a phase 1 study. Cancer 517. Nabhan C, Patton D, Gordon LI, et al. A pilot trial of rituximab
1997;80:2712–20. and alemtuzumab combination therapy in patients with relapsed
498. Montemurro F, Choa G, Faggiuolo R, et al. Safety and activity and/or refractory chronic lymphocytic leukemia (CLL). Leuk
of docetaxel and trastuzumab in HER2 overexpressing meta- Lymphoma 2004;45:2269–73.
static breast cancer: a pilot phase II study. Am J Clin Oncol 518. Nadler LM, Stashenko P, Hardy R, et al. Serotherapy of a patient
2003;26:95–7. with a monoclonal antibody directed against a human lympho-
499. Montillo M, Tedeschi A, Miqueleiz S, et al. Alemtuzumab as ma-associated antigen. Cancer Res 1980; 40:3147–54.
consolidation after a response to fludarabine is effective in purg- 519. Nayak SK, Schiltz PM, Dillman RO. Modulation of renal carci-
ing residual disease in patients with chronic lymphocytic leuke- noma cells in vitro: comparison after transduction with retroviral
mia. J Clin Oncol 2006;24:2337–42. vector containing a human γ-interferon gene versus incubation with
500. Morabito A, Longo R, Gattuso D, et al. Trastuzumab in combina- soluble γ-interferon. J Interferon Cytokine Res 1999;19:49–58.
tion with gemcitabine and vinorelbine as second-line therapy for 520. Nemeth JA, Nakada MT, Trikha M, et al. Alpha-v integrins as
HER-2/neu overexpressing metastatic breast cancer. Oncol Rep therapeutic targets in oncology. Cancer Invest 2007;25:632–46.
2006;16:393–8. 521. Neninger E, Diaz RM, de la Torre A et al. Active immunotherapy
501. Moreau P, Harousseau JL, Wijdenes J, et al. A combination of with 1E10 anti-idiotype vaccine in patients with small cell lung
anti-interleukin 6 murine monoclonal antibody with dexamethasone cancer: report of a phase I trial. Cancer Biol Ther 2007;6:145–50.
Robert O. Dillman 399

522. Nicholson GL. Tumor cell instability, diversification, and pro- without trastuzumab according to HER2 expression: changing
gression to the metastatic phenotype: from oncogene to oncofetal the natural history of HER2-positive disease. Ann Oncol
expression. Cancer Res 1987; 47:1473–87. 2006;17:630–6.
523. Niitsu N, Kohuri M, Higashihara M et al. Phase II study of the 544. Pauletti G, Dandekar S, Rong HM, et al. Assessment of methods for
CPT-11, mitoxantrone and dexamethasone regimen in combina- tissue-based detection of the HER-2/neu alteration in human breast
tion with rituximab in elderly patients with relapsed diffuse large cancer: a direct comparison of fluorescence in situ hybridization
B-cell lymphoma. Cancer Sci 2006;97:933–7. and immunohistochemistry. J Clin Oncol 2000;18:3651–64.
524. Nimmerjahn F, Ravetch JV. Antibodies, Fc receptors and cancer. 545. Pawson R, Dyer JMS, Barge R, et al. Treatment of T-cell prolym-
Curr Opin Immunol 2007;19:239–45. phocytic leukemia with human CD52 antibody. J Clin Oncol
525. Norman DJ, Vincenti F, de Mattos AM, et al. Phase I trial of 1997;15:2667–72.
HuM291, a humanized anti-CD3 antibody, in patients 546. Pegram MD, Lipton A, Hayes DF, et al. Phase II study of receptor-
receiving renal allografts from living donors.Transplantation enhanced chemosensitivity using recombinant humanized anti-
2000;70:1707–12. p185HER2/neu monoclonal antibody plus cisplatin in patients
526. Noujaim AA, Schultes BC, Baum RP, Madiyalakan R. Induction with HER2/neu-overexpressing metastatic breast cancer refrac-
of CA125-specific B and T cell responses in patients injected tory to chemotherapy treatment. J Clin Oncol 1998;16:2659–71.
with MAb-B43.13 – evidence for antibody-mediated antigen- 547. Pegram MD, Pienkowski T, Northfelt DW, et al. Results of two
processing and presentation of CA125 in vivo. Cancer Biother open-label, multicenter phase II studies of docetaxel, platinum
Radiopharm 2001;16:187–203. salts, and trastuzumab in HER2-positive advanced breast cancer.
527. Oaber RJ, Radu CG, Ghetie V et al. Difference in promiscuity for J Natl Cancer Inst 2004;96:759–69.
antibody-FcRn, interactions across species: implications for ther- 548. Perez EA, Suman VJ, Davidson NE, et al. Effect of doxorubicin plus
apeutic antibodies. Int Immunol 2001;13:1551–9. cyclophosphamide on left ventricular ejection fraction in patients
528. Oberneder R, Weckermann D, Ebner B, et al. A phase I study with breast cancer in the North Central Cancer Treatment Group
with adecatumumab, a human antibody directed against epithelial N9831 Intergroup Adjuvant Trial. J Clin Oncol 2004;22:3700–4.
cell adhesion molecule, in hormone refractory prostate cancer 549. Perez EA, Suman VJ, Rowland KM, et al. Two concurrent phase
patients. Eur J Cancer 2006;42:2530–8. II trials of paclitaxel/carboplatin/trastuzumab (weekly or every-
529. O’Brien SM, Kantarjian H, Thomas DA, et al. Rituximab dose- 3-week schedule) as first-line therapy in women with HER2-
escalation trial in chronic lymphocytic leukemia. J Clin Oncol overexpressing metastatic breast cancer: NCCTG study 983252.
2001;19:2165–70. Clin Breast Cancer 2005;6:425–32.
530. O’Brien SM, Kantarjian HM, Thomas DA, et al. Alemtuzumab 550. Perez EA, Suman VJ, Davidson NE, et al. HER2 testing by local,
as treatment for residual disease after chemotherapy in patients central, and reference laboratories in specimens from the North
with chronic lymphocytic leukemia. Cancer 2003;98:2657–63. Central Cancer Treatment Group N9831 intergroup adjuvant
531. O’Day SJ, Hamid O, Urba WJ. Targeting cytotoxic T-lymphocyte trial. J Clin Oncol 2006;24:3032–8.
antigen-4 (CTLA-4): a novel strategy for the treatment of mela- 551. Perez-Soler R, Delord JP, Halpern A, et al. HER1/EGFR inhibi-
noma and other malignancies. Cancer 2007;110:2614–27. tor-associated rash: future directions for managment and investi-
532. Ogura M, Morishima Y, Kagami Y, et al. Randomized phase II gation outcomes for the HER1/EGFR inhibitor rash management
study of concurrent and sequential rituximab and CHOP chemo- forum. Oncologist 2005;10:345–56.
therapy in untreated indolent B-cell lymphoma. Cancer Sci 2006 552. Pericleous LM, Richards J, Epenetos AA et al. Characterisation
97:305–12. and internalisation of recombinant humanised HMFG-1 antibod-
533. Oldham RK. Monoclonal antibodies in cancer therapy. J Clin ies against MUC1. Br J Cancer 2005;93:1257–66.
Oncol 1983;1:582–90. 553. Pfister DG, Su YB, Kraus DH, et al. Concurrent cetuximab, cispla-
534. Oldham RK. Monoclonal antibodies: does sufficient selectivity tin, and concomitant boost radiotherapy for locoregionally advanced,
to cancer cells exist for therapeutic application? J Biol Response squamous cell head and neck cancer: a pilot phase II study of a new
Med 1987;6:227–34. combined-modality paradigm. J Clin Oncol 2006;24:1072–8.
535. Oldham RK, Foon KA, Morgan C, et al. Monoclonal antibody 554. Pfisterer J, du Bois A, Sehouli J, et al. The anti-idiotypic antibody
therapy of malignant melanoma: In vivo localization in cutane- abagovomab in patients with recurrent ovarian cancer. A phase I
ous metastases after intravenous administration. J Clin Oncol trial of the AGO-OVAR. Ann Oncol 2006;17:1568–77.
1984;2:1235–44. 555. Pfreundschuh M, Trumper L, Kloess M, et al. Two-weekly or
536. Oosterwijk E, Bander NH, Divgi CR, et al. Antibody localization 3-weekly CHOP chemotherapy with or without etoposide for the
in human renal cell carcinoma: a phase I study of monoclonal treatment of elderly patients with aggressive lymphomas: results
antibody G250. J Clin Oncol 1993;11:738–50. of the NHL-B2 trial of the DSHNHL. Blood 2004;104:634–41.
537. O’Shaughnessy JA, Vukelja S, Marsland T, et al. Phase II study 556. Pfreundschuh M, Trumper L, Osterborg A, et al. CHOP-like che-
of trastuzumab plus gemcitabine in chemotherapy-pretreated motherapy plus rituximab versus CHOP-like chemotherapy alone
patients with metastatic breast cancer. Clin Breast Cancer in young patients with good-prognosis diffuse large-B-cell lym-
2004;5:142–7. phoma: a randomised controlled trial by the MabThera
538. Ostendorf T, Kunter U, Eitner F, et al. VEGF(165) mediates International Trial (MInT) Group. Lancet Oncol 2006;7:379–91.
glomerular endothelial repair. J Clin Invest. 1999;104:913–23. 557. Pfreundschuh M, Schubert J, Ziepert M, et al. Six versus eight
539. Osterborg A, Dyer MJS, Bunjes D, et al. Phase II multicenter cycles of bi-weekly CHOP-14 with or without rituximab in elderly
study of human CD52 antibody in previously untreated chronic patients with aggressive CD20 + B-cell lymphomas: a randomised
lymphocytic leukemia. J Clin Oncol 1997;15:1567–74. controlled trial (RICOVER-60). Lancet Oncol 2008;9:105–16.
540. Ozaki S, Kosaka M, Wakahara Y, et al. Humanized anti-HM1.24 558. Phan GQ, Yang JC, Sherry RM, et al. Cancer regression and auto-
antibody mediates myeloma cell cytoxicity that is enhanced by immunity induced by cytotoxic T lymphocyte-associated antigen
cytokine stimulation of effector cells. Blood 1999;93:3922–30. 4 blockade in patients with metastatic melanoma. Proc Natl Acad
541. Pai LH, Bookman MA, Ozols RF, et al: Clinical evaluation of intra- Sci USA 2003;100:8372–7.
peritoneal Pseudomonas exotoxin immunoconjugate OBV3-PE in 559. Philip PA, Benedetti J, Fenoglio-Preiser C, et al. Phase III study
patients with ovarian cancer. J Clin Oncol 1991;9:2095–103. of gemcitabine plus cetuximab versus gemcitabine in patients
542. Paik S, Bryant J, Tan-Chiu E, et al. Real-world performance of with locally advanced or metastatic pancreatic adenocarcinoma.
HER2 testing-National Surgical Adjuvant Breast and Bowel Proc Am Soc Clin Oncol 2007;25:199s [abstract 4509].
Project experience. J Natl Cancer Inst 2002;94:852–4. 560. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al.
543. Papaldo P, Fabi A, Ferretti G, et al. A phase II study on metastatic Trastuzumab after adjuvant chemotherapy in HER2-positive
breast cancer patients treated with weekly vinorelbine with or breast cancer N Engl J Med 2005;353:1659–72.
400 Monoclonal antibody therapy

561. Pinto C, Di Fabio F, Siena, et al. Phase II study of cetuximab 581. Rankin EM, Hekman A, Somers R, et al. Treatment of two
in combination with FOLFIRI in patients with untreated patients with B cell lymphoma with monoclonal anti-idiotype
advanced gastric or gastroesophageal junction adenocarci- antibodies. Blood 1985; 65:1373–81.
noma (FOLCETUX study). Ann Oncol 2007;18:510–7. 582. Rech J, Repp R, Rech D, et al. A humanized HLA-DR antibody
562. Piro LD, White CA, Grillo-Lopez AJ, et al. Extended rituximab (hu1D10, apolizumab) in combination with granulocyte colony-
(anti-CD20 monoclonal antibody therapy in patients with stimulating factor (filgrastim) for the treatment of non-Hodgkin’s
relapsed low-grade or follicular non-Hodgkins lymphoma. Ann lymphoma: a pilot study. Leuk Lymphoma 2006;47:2147–54.
Oncol 1999;10:655–61. 583. Reece DE, Foon KA, Bhattacharya-Chatterjee M et al. Use of the
563. Plevy S, Salzberg B, Van Assche G, et al. A phase I study of anti-idiotype breast cancer Vaccine 11D10 in conjunction with
visilizumab, a humanized anti-CD3 monoclonal antibody, in autologous stem cell transplantation in patients with metastatic
severe steroid-refractory ulcerative colitis.Gastroenterology. breast cancer. Clin Breast Cancer 2003;3 Suppl 4:S152–7.
2007;133:1414–22. 584. Reff ME, Heard C. A review of modifications to recombinant
564. Plummer R, Attard G, Pacey S, et al. Phase 1 and pharmacoki- antibodies: attempt to increase efficacy in oncology applications.
netic study of lexatumumab in patients with advanced cancers. Crit Rev Oncol Hematol 2001;40:25–35.
Clin Cancer Res 2007;13:6187–94. 585. Regazzi MB, Iacona I, Avanzini MA, et al. Pharmacokinetic
565. Posey JA, Khazaeli MB, DelGrosso A, et al. A pilot trial of behavior of rituximab: a study of different schedules of adminis-
Vitaxin, a humanized anti-vitronectin receptor (anti alpha v beta 3) tration for heterogeneous clinical settings. Ther Drug Monit
antibody in patients with metastatic cancer. Cancer Biother 2005;27:785–92.
Radiopharm 2001;16:125–32. 586. Rehwald U, Schulz H, Reiser M, et al. Treatment of relapsed CD20
566. Posey JA, Ng TC, Yang B, et al. A phase 1 study of anti-kinase + Hodgkin lymphoma with the monoclonal antibody rituximab is
insert domain-containing receptor antibody, IMC-1C11, in effective and well tolerated: results of a phase 2 trial of the German
patients with liver metastases from colorectal carcinoma. Clin Hodgkin Lymphoma Study Group. Blood 2003;101:420–4
Cancer Res 2003;9:1323–32. 587. Reichert JM. Technology evaluation: lumiliximab, Biogen Idec.
567. Posner MC, Niedzwiecki D, Venook AP, et al. A phase II pro- Curr Opin Mol Ther 2004;6:675–83.
spective multi-institutional trial of adjuvant active specific immu- 588. Reinartz S, Kohler S, Schlebusch H, et al. Vaccination of patients
notherapy following curative resection of colorectal cancer with advanced ovarian carcinoma with the anti-idiotype ACA125:
hepatic metastases: Cancer and Leukemia Group B study 89903. immunological response and survival (phase Ib/II). Clin Cancer
Ann Surg Oncol 2008;15:158–64. Res 2004;10:1580–7.
568. Prehn RT. Tumor-specific antigens as altered growth factor recep- 589. Repka T, Chiorean EG, Gay J, et al. Trastuzumab and interleu-
tors. Cancer Res 1989;49:2823–26. kin-2 in HER2-positive metastatic breast cancer: a pilot study.
569. Press OW, Appelbaum F, Ledbetter JA, et al. Monoclonal anti- Clin Cancer Res 2003;9:2440–6.
body 1F5 [anti-CD-20] serotherapy of human B cell lymphomas. 590. Reuben JM, Lee BN, Li C, et al. Biologic and immunomodula-
Blood 1987; 69:584–591. tory events after CTLA-4 blockade with ticilimumab in patients
570. Presta LG, Chen H, O’Connor SJ, et al. Humanization of an anti- with advanced malignant melanoma. Cancer 2006;106:
vascular endotherlial growth factor monoclonal antibody for the 2437–44.
therapy of solid tumors and other disorders. Cancer Res 591. Ribas A, Camacho LH, Lopez-Berestein G, et al. Antitumor
1997;57:4593–9. activity in melanoma and anti-self responses in a phase I trial
571. Prewett M, Rolthman M, Walsal H, et al. Mouse-human chimeric with the anti-cytotoxic T lymphocyte-associated antigen 4 mono-
anti-epidermal growth factor receptor antibody C225 inhibits the clonal antibody CP-675,206. J Clin Oncol 2005;23:8968–77.
growth of human renal cell carcinoma xenografts in nude mice. 592. Richards J, Auger J, Peace D, et al. Phase I evaluation of human-
Clin Cancer Res 1998;4:2957–66. ized OKT3: toxicity and immunomodulatory effects of hOKT3-
572. Przepiorka D, Kernan NA, Ippoliti C, et al. Daclizumab, a gamma4. Cancer Res 1999;59:2096–101.
humanized anti-interleukin-2 receptor alpha chain antibody, for 593. Richards JM, Vogelzang NJ, Bluestone JA. Neurotoxicity after
treatment of acute graft-vs-host disease. Blood 2000;95:83–89. treatment with muromonab-CD3. New Engl J Med 1990;323:
573. Pukel CS, Lloyd KO, Travassos LR, et al. GD3, a prominent gan- 487–8.
glioside of human melanoma. J Exp Med 1982; 155:1133–47. 594. Rider DA, Havenith CE, de Ridder R, et al. A human CD4 mono-
574. Punt CJ, Nagy A, Douillard JY, et al. Edrecolomab alone or in clonal antibody for the treatment of T-cell lymphoma combines
combination with fluorouracil and folinic acid in the adjuvant inhibition of T-cell signaling by a dual mechanism with potent
treatment of stage III colon cancer: a randomised study. Lancet Fc-dependent effector activity. Cancer Res 2007;67:9945–53.
2002;360:671–7. 595. Riethmuller G, Holz E, Schlimok G, et al. Monoclonal antibody
575. Ragnhammar P, Pagerberg J, Frodin JE, et al: Effect of monoclo- therapy for resected Dukes’ C colorectal cancer: seven-year out-
nal antibody 17–1A and GM-CSF in patients with advanced col- come of a multicenter randomized trial J Clin Oncol
orectal carcinoma: long-lasting complete remissions can be 1998;16:1788–94.
induced. Int J Cancer 1993;53:751–8. 596. Rigacci L, Nassi L, Alterini R, et al. Dose-dense CHOP plus
576. Rai KR, Peterson BL, Appelbaum FR, et al. Fludarabine com- rituximab (R-CHOP14) for the treatment of elderly patients with
pared with chlorambucil as primary therapy for chronic lympho- high-risk diffuse large B cell lymphoma: a pilot study. Acta
cytic leukemia. N Engl J Med. 2000; 343:1750–7. Haematol 2006;115:22–7.
577. Rai K, Mercier RJ, Cooper MR, et al. Campath-1H is an effective 597. Rini BI, Halabi S, Taylor J, et al. Cancer and Leukemia Group B
salvage therapy for fludarabine failing CLL patients: results of a 90206: A randomized phase III trial of interferon-alpha or inter-
phase II trial. Blood 2001;96:163a [abstract 703]. feron-alpha plus anti-vascular endothelial growth factor antibody
578. Rai KR, Freter CE, Mercier RJ, et al. Alemtuzumab in previously (bevacizumab) in metastatic renal cell carcinoma. Clin Cancer
treated chronic lymphocytic leukemia patients who also had Res 2004;10:2584–6.
received fludarabine. J Clin Oncol 2002;20:3891–7. 598. Rini BI, Weinberg V, Fong L, et al. Combination immunotherapy
579. Ramaswamy B, Elias AD, Kelbick NT, et al. Phase II trial of with prostatic acid phosphatase pulsed antigen-presenting cells
bevacizumab in combination with weekly docetaxel in metastatic (Provenge) plus bevacizumab in patients with serologic progres-
breast cancer patients. Clin Cancer Res. 2006;12:3124–9. sion of prostate cancer after definitive local therapy. Cancer
580. Ramos TC, Figueredo J, Catala M, et al. Treatment of high-grade 2006;107:67–74.
glioma patients with the humanized anti-epidermal growth factor 599. Ritz J, Pesando JM, Notis-McConarty J, et al. Modulation of
receptor (EGFR) antibody h-R3: report from a phase I/II trial. human acute lymphoblastic leukemia antigen induced by mono-
Cancer Biol Ther 2006;5:375–9. clonal antibody in vitro. J Immunol 1980; 125:1506–14.
Robert O. Dillman 401

600. Ritz J, Pesando JM, Sallan SE, et al. Serotherapy of acute lym- 620. Safran H, Iannitti D, Ramanathan R, et al. Herceptin and gemcit-
phoblastic leukemia with monoclonal antibody. Blood 1981; abine for metastatic pancreatic cancers that overexpress HER-2/
58:141–52. neu. Cancer Invest. 2004;22:706–12.
601. Robak T, Smolewski P, Cebula B, et al. Rituximab combined 621. Safran H, Diepetrillo T, Akerman P, et al. Phase I/II study of tras-
with cladribine or with cladribine and cyclophosphamide in tuzumab, paclitaxel, cisplatin and radiation for locally advanced,
heavily pretreated patients with indolent lymphoproliferative dis- HER2 overexpressing, esophageal adenocarcinoma. Int J Radiat
orders and mantle cell lymphoma.Cancer 2006;107:1542–50. Oncol Biol Phys 2007;67:405–9.
602. Robert F, Ezekiel MP, Spencer SA, et al. Phase I study of anti – 622. Saleh MN, LoBuglio AF, Wheeler RH, et al. A phase II trial of
epidermal growth factor receptor antibody cetuximab in combi- murine monoclonal antibody 17–1A and interferon-gamma: Clinical
nation with radiation therapy in patients with advanced head and and immunological data. Cancer Immunol 1990;32:185–90.
neck cancer. J Clin Oncol 2001;19:3234–43. 623. Saleh MN, Khazaeli MB, Wheeler RH, et al. Phase I trial of the
603. Robert F, Blumenschein G, Herbst RS, et al. Phase I/IIa study of murine monoclonal anti-GD2 antibody 14G2a in metastatic mela-
cetuximab with gemcitabine plus carboplatin in patients with noma. Cancer Res 1992a;52:4342–7.
chemotherapy-naive advanced non-small-cell lung cancer. J Clin 624. Saleh MN, Khazaeli MD, Wheeler RH, et al. Phase I trial of the
Oncol 2005;23:9089–96. chimeric anti-GD2 monoclonal antibody ch14.18 in patients
604. Robert N, Leyland-Jones B, Asmar L, et al. Randomized with malignant melanoma. Hum Antibod Hybridomas 1992b:3:
phase III study of trastuzumab, paclitaxel, and carboplatin 19–23.
compared with trastuzumab and paclitaxel in women with 625. Saleh MN, Khazaeli MB, Grizzle WE, et al. A phase I clinical
HER-2-overexpressing metastatic breast cancer. J Clin Oncol trial of murine monoclonal antibody D612 in patients with meta-
2006;24:2786–92. static gastrointestinal cancer. Cancer Res 1993;53:4555–62.
605. Romaguera JE, Fayad L, Rodriguez MA et al. High rate of dura- 626. Saleh MN, Khazaeli MB, Wheeler RH et al. Phase II trial of
ble remissions after treatment of newly diagnosed aggressive murine monoclonal antibody D612 combined with recombi-
mantle-cell lymphoma with rituximab plus hyper-CVAD alter- nant human monocyte colony-stimulating factor (rhM-CSF) in
nating with rituximab plus high-dose methotrexate and cytara- patients with metastatic gastrointestinal cancer. Cancer Res
bine. J Clin Oncol 2005;23:7013–23. 1995;55:4339–46.
606. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adju- 627. Saleh MN, LeMaistre CF, Kuzel TM, et al: Antitumor activity of
vant chemotherapy for operable HER2-positive breast cancer. N DAB389IL-2 fusion toxin in mycosis fungoides. J Am Acad
Engl J Med 2005;353:1673–84. Dermatol 1998; 39:63–73.
607. Rosenberg SA, Terry WD. Passive immunotherapy of cancer in 628. Saltz LB, Meropol NJ, Loehrer PJ Sr, et al. Phase II trial of cetux-
animals and man. Adv Cancer Res 1977;25:323–88. imab in patients with refractory colorectal cancer that expresses the
608. Roskoski R Jr. The ErbB/HER receptor protein-tyrosine kinases epidermal growth factor receptor. J Clin Oncol 2004;22:1201–8.
and cancer. Biochem Biophys Res Commun. 2004;319:1–11. 629. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone
609. Rossi J-F, Yang LZ, Quittet P, et al. Rituximab and GM-CSF, an or with bevacizumab for non-small-cell lung cancer. N Engl J
effective therapy for relapsed refractory patients with low-grade Med 2006;355:2542–50.
B-cell lymphoma: correlation between response and dendritic cell 630. Sato N, Sano M, Tabei T, et al. Combination docetaxel and tras-
subpopulation mobilized. Blood 2001;98:607a [abstract 2542]. tuzumab treatment for patients with HER-2-overexpressing met-
610. Rossi JF, Feguex N, Lu Zy, et al. Optimizing the use of anti-inter- astatic breast cancer: a multicenter, phase-II study. Breast Cancer
leukin-6 monoclonal antibody with dexamethasone and 140 mg/m2 2006;13:166–71.
of melphalan in multiple myeloma: results of a pilot study includ- 631. Scallon BJ, Snyder LA, Anderson GM, et al. A review of anti-
ing biological aspects. Bone Marrow Transplant 2005;36:771–9. body therapeutics and antibody-related technologies for oncol-
611. Rowinsky EK. The erbB family: targets for therapeutic devel- ogy. J Immunother 2006;29:351–364.
opment against cancer and therapeutic strategies using mono- 632. Schecter AL, Stern DF, Vaidanathan L, et al. The neu oncogene:
clonal antibodies and tyrosine kinase inhibitors. Annu Rev Med an erb-B-related gene encoding a 185,000-Mr tumor antigen.
2004;55:433–57. Nature 1984;312:513–516.
612. Rowinsky EK, Schwartz GH, Gollob JA, et al. Safety, pharma- 633. Scheinberg DA, Lovett D, Divgi CR, et al. A Phase I trial of
cokinetics, and activity of ABX-EGF, a fully human anti-epider- monoclonal antibody M195 in acute myelogenous leukemia:
mal growth factor receptor monoclonal antibody in patients with Specific bone marrow targeting and internalization of radionu-
metastatic renal cell cancer. J Clin Oncol 2004;22:3003–15. clide. J Clin Onc 1991;9:478–490.
613. Rubenstein JL, Firdlyand J, Abrey L, et al. Phase I study of intra- 634. Schlaeppi JM, Wood JM. Targeting vascular endothelial growth
ventricular administration of rituximab in patients with recurrent factor (VEGF) for anti-tumor therapy, by anti-VEGF neutralizing
CNS and intraocular lymphoma. J Clin Oncol 2007;25:1350–6. monoclonal antibodies or by VEGF receptor tyrosine-kinase
614. Rudders RA, Levin A, Jespersen D, et al. Crossreacting human inhibitors. Cancer Metastasis Rev 1999;18:473–81.
lymphoma idiotypes. Blood 1992;80:1039–1044. 635. Schlom J, Wunderlich D, Teramoto YA. Generation of human
615. Ryan KP, Dillman RO, DeNardo SJ, et al. Breast cancer imaging monoclonal antibodies reactive with human mammary carcinoma
with In-111 human IgM monoclonal antibodies. Radiol cells. Proc Natl Acad Sci 1980;77:6841–45.
1988;167:71–5. 636. Schneck D, Butler F, Dugan W, et al: Phase I studies with a murine
616. Sabbatini P, Dupont J, Aghajanian C, et al. Phase I study of aba- monoclonal antibody vinca conjugate (KS1/4-DAVLB) in patients
govomab in patients with epithelial ovarian, fallopian tube, or with adenocarcinoma antibody. Immunoconj Radiopharm 1989;
primary peritoneal cancer. Clin Cancer Res 2006;12:5503–10. 2:93–100.
617. Sacchi S, Federico M, Vitolo U, et al. Clinical activity and safety 637. Schnipper LE. Clinical implications of tumor cell heterogeneity.
of combination immunotherapy with IFN-alpha 2a and Rituximab N Engl J Med 1986; 314:1423–31.
in patients with relapsed low grade non-Hodgkin’s lymphoma. 638. Schoof DD, Selleck CM, Massaro AF, et al. Activation of human
Hematologica 2001;86:951–8. tumor-infiltrating lymphocytes by monoclonal antibodies directed
618. Sacchi S, Pozzi S, Marcheselli R, et al. Rituximab in combination to the CD3 complex. Cancer Res 1990; 50:1138–43.
with fludarabine and cyclophosphamide in the treatment of patients 639. Schroff RW, Farrell MM, Klein RA, et al. T65 antigen modula-
with recurrent follicular lymphoma. Cancer 2007a;110:121–8. tion in a phase I monoclonal antibody trial with chronic lympho-
619. Sacchi S, Pozzi S, Marcheselli L, et al. Introduction of rituximab cytic leukemia patients. J Immunol 1984; 133:1641–48.
in front-line and salvage therapies has improved outcome of 640. Schroff RW, Foon KA, Wilburn SB, et al. Human antimurine
advanced-stage follicular lymphoma patients. Cancer 2007b;109: immunoglobulin responses in patients receiving monoclonal
2077–82. antibody therapy. Cancer Res 1985; 45:879–85.
402 Monoclonal antibody therapy

641. Schulz H, Klein SK, Rehwald U, et al. Phase 2 study of a com- 661. Shan D, Ledbetter JA, Press OW. Signaling events involved in
bined immunochemotherapy using rituximab and fludarabine anti-CD20-induced apoptosis of malignant human B cells.
in patients with chronic lymphocytic leukemia. Blood Cancer Immuno Immunother 2000;68:673–83.
2002;100:3115–20. 662. Shaw DR, Khazaeli MB, LoBuglio AF. Mouse/human chimeric
642. Schwaab T, Lewis LD, Cole BF, et al. Phase I pilot trial of the antibodies to a tumor-associated antigen: biologic activity of the
bispecific antibody MDXH210 (anti-Fc gamma RI X anti-HER-2/ four human IgG subclasses. J Natl Cancer Inst 1988; 80:1553–9.
neu) in patients whose prostate cancer overexpresses HER-2/neu. 663. Shawler DL, Miceli MC, Wormsley SB, et al. Induction of in
J Immunother 2001;24:79–87. vitro and in vivo antigenic modulation by the anti-human T cell
643. Schwartz RS. Shattuck lecture – diversity of the immune reper- monoclonal antibody T101. Cancer Res 1984;44:5921–7.
toire and immunoregulation. N Engl J Med 2003;348:1017–26. 664. Shawler DL, Bartholomew RM, Smith LM, et al. Human immune
644. Schwartz RS. Paul Ehrlich’s magic bullets. N Engl J Med response to multiple injections of murine monoclonal IgG. J
2004;350:1079–80. Immunol 1985;135:1530–35.
645. Scott AM, Lee FT, Hopklins W, et al. Specific targeting, biodis- 665. Shawler DL, McCallister TJ, Sobol RE, et al. Serologic and cel-
tribution, and lack of immunogenicity of chimeric anti-GD3 lular assays to monitor therapy with murine monoclonal antibod-
monoclonal antibody KM871 in patients with metastatic mela- ies. J Clin Lab Anal 1987; 2:184–90.
noma: results of a phase I trial. J Clin Oncol 2001;19:3976–87. 666. Shen JW, Atkinson B, Koprowsky H, et al. Binding of murine
646. Scott AM, Wiseman G, Welt S et al. A Phase I dose-escalation immunoglobulin to human tissues after immunotherapy with
study of sibrotuzumab in patients with advanced or metastatic anti-colorectal carcinoma monoclonal antibody. Int J Cancer
fibroblast activation protein-positive cancer. Clin Cancer Res 1984;33:465–8.
2003;9:1639–47. 667. Siberil S, Dutertre CA, Fridman WH et al. FcgammaR: The key
647. Scott AM, Lee FT, Jones R, et al. A phase I trial of humanized to optimize therapeutic antibodies? Crit Rev Oncol Hematol
monoclonal antibody A33 in patients with colorectal carcinoma: 2007;62:26–33.
biodistribution, pharmacokinetics, and quantitative tumor uptake. 668. Siena S, Peeters M, Van Cutsem E, et al. Association of progres-
Clin Cancer Res 2005;11:4810–7. sion-free survival with patient-reported outcomes and survival:
648. Scott AM, Lee FT, Tebbutt N, et al. A phase I clinical trial with results from a randomised phase 3 trial of panitumumab. Br J
monoclonal antibody ch806 targeting transitional state and Cancer 2007;97:1469–74.
mutant epidermal growth factor receptors. Proc Natl Acad Sci U 669. Sikora K, Wright R. Human monoclonal antibodies to lung can-
S A. 2007a;104:4071–6. cer antigens. Br J Cancer 1981;43:696–700.
649. Scott AM, Tebbutt N, Lee FT, et al. A phase I biodistribution and 670. Sikora K, Alderson T, Phillips J, et al. Human hybridomas from
pharmacokinetic trial of humanized monoclonal antibody malignant gliomas. Lancet 1982;1:11–4.
Hu3s193 in patients with advanced epithelial cancers that express 671. Sikora K, Alderson T, Ellis J, et al. Human hybridomas from
the Lewis-Y antigen. Clin Cancer Res 2007b;13:3286–92. patients with malignant disease. Br J Cancer 1983; 47:135–45.
650. Seagaert S, van Cutsem E. Clinical signs, pathophysiology, and 672. Singh R, Bandyopadhyay D. MUC1: a target molecule for cancer
management of skin toxicity during therapy with epidermal therapy. Cancer Biol Ther 2007;6:481–6.
growth factor receptor inhibitors. Ann Oncol 2005;16:1425–33. 673. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer:
651. Sears HF, Atkinson B, Mattis J, et al. Phase I clinical trial of correlation of relapse and survival with amplicfication of the
monoclonal antibody in treatment of gastrointestinal tumors. HER2–2/neu oncogene. Science 1987;235:177–82.
Lancet 1982;1:762–5. 674. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy
652. Sears HF, Herlyn D, Steplewski Z, et al. Effects of monoclonal plus a monoclonal antibody against HER2 for metastatic breast can-
antibody immunotherapy on patients with gastrointestinal adeno- cer that overexpresses HER2. N Engl J Med 2001;344:783–92.
carcinoma. J Biol Response Modif 1984;3:136–50. 675. Sliwkowski MX, Lofgren JA, Lewis GC, et al. Nonclinical
653. Sears HF, Herlyn D, Steplewski Z, et al. Phase II clinical trial of studies addressing the mechanism of action of trastuzumab
a murine monoclonal antibody cytotoxic for gastrointestinal (Herceptin). Semin Oncol 1999;26(4 suppl 12):60–70.
adenocarcinoma. Cancer Res 1985; 45:5910–13. 676. Small EJ, Tchekmedyian NS, Rini BI, et al. A pilot trial of CTLA-4
654. Sebastian M, Passlick B, Friccius-Quecke H, et al. Treatment of blockade with human anti-CTLA-4 in patients with hormone-
non-small cell lung cancer patients with the trifunctional mono- refractory prostate cancer. Clin Cancer Res 2007;13:1810–5.
clonal antibody catumaxomab (anti-EpCAM x anti-CD3): a 677. Smith I, Procter M, Gelber RD, et al. 2-year follow-up of trastu-
phase I study. Cancer Immunol Immunother 2007;56:1637–44. zumab after adjuvant chemotherapy in HER2-positive breast
655. Sehn LH, Donaldson J, Filewich A, et al. Rapid infusion ritux- cancer: a randomised controlled trial. Lancet 2007;369:29–36.
imab in combination with corticosteroid-containing chemother- 678. Smith KL, Dang C, Seidman AD. Cardiac dysfunction associated
apy or as maintenance therapy is well tolerated and can safely be with trastuzumab. Expert Opin Drug Saf 2006;5:619–29.
delivered in the community setting. Blood 2007;109:4171–3. 679. Sobol RE, Dillman RO, Smith JD, et al. Phase I evaluation of
656. Seiden MV, Burris HA, Matulonis U, et al. Phase II trial of murine monoclonal anti-melanoma antibody in man: preliminary
EMD72000 (matuzumab), a humanized anti-EGFR monoclonal observations. In: Hybridomas in Cancer Diagnosis and Treatment.
antibody, in patients with platinum-resistant ovarian and primary Mitchell MS, Oettgen HF, eds. New York: Raven Press,
peritoneal malignancies. Gynecol Oncol 2007;104:727–31. 1981;21:199–206.
657. Seidman AD, Fornier MN, Esteva FJ, et al. Weekly trastuzumab 680. Sodee DB, Malguria N, Faulhaber P, et al. Multicenter ProstaScint
and paclitaxel therapy for metastatic breast cancer with analysis imaging findings in 2154 patients with prostate cancer. The
of efficacy by HER2 immunophenotype and gene amplification. ProstaScint Imaging Centers. Urology 2000;56:988–93.
J Clin Oncol 2001;19:2587–95. 681. Soh EY, Eigelberger MS, Kim KJ, et al. Neutralizing vascular
658. Seidman A, Hudis C, Pierri MK et al. Cardiac dysfunction in the endothelial growth factor activity inhibits thyroid cancer growth
trastuzumab clinical trials experience. J Clin Oncol 2002;20: in vivo. Surgery 2000;128:1059–65.
1215–21. 682. Soiffer RJ, Chapman PB, Murray C, et al. Administration of R24
659. Shah MA, Ramanathan RK, Ilson DH, et al. Multicenter phase II monoclonal antibody and low-dose interleukin-2 for malignant
study of irinotecan, cisplatin, and bevacizumab in patients with melanoma. Clin Cancer Res 1997;3:17–24.
metastatic gastric or gastroesophageal junction adenocarcinoma. 683. Solal-Celigny P, Salles GA, Brousse N, et al. Single 4-dose ritux-
J Clin Oncol 2006;24:5201–6. imab treatment for low-tumor burden follicular lymphoma: sur-
660. Shakib F, ed. Basic and clinical aspects of IgG subclasses. New vival analyses with a follow-up of at least 5 years. Blood
York: Carger, 1986. 2004;104:169a [abstract 585].
Robert O. Dillman 403

684. Sosman JA, Weiss GR, Margolin KA, et al. Phase IB clinical 705. Taetle R, Honeysett JM. Effects of monoclonal anti-transferrin
trial of anti-CD3 followed by high dose bolus interleukin-2 in receptor antibodies on in vitro growth of human solid tumor cells.
patients with metastatic melanoma and advanced renal cell car- Cancer Res 1987;47:2040–4.
cinoma; clinical and immunologic effects. J Clin Oncol 706. Tan AR, Moore DF, Hidalgo M, et al. Pharmacokinetics of cetux-
1993;11:1496–505. imab after administration of escalating single dosing and weekly
685. Starlets D, Gore Y, Binsky I, et al. Cell-surface CD74 initiates a fixed dosing in patients with solid tumors.Clin Cancer Res
signaling cascade leading to cell proliferation and survival. Blood 2006;12:6517–22.
2006;107:4807–16. 707. Tan-Chiu E, Yothers G, Romond E, et al. Assessment of cardiac
686. Starling J, Cote RJ, Marder P, et al. Tissue distribution and cel- dysfunction in a randomized trial comparing doxorubicin and
lular location of the antigens recognized by human monoclonal cyclophosphamide followed by paclitaxel, with or without trastu-
antibodies 16.88 and 28A32. Cancer Res 1988;48:7273–78. zumab as adjuvant therapy in node-positive, human epidermal
687. Stein H, Hummel H. Cellular origin and clonality of classic growth factor receptor 2-overexpressing breast cancer: NSABP
Hodgkin’s lymphoma: immunophenotypic and molecular stud- B-31. J Clin Oncol 2005;23:7811–9.
ies. Semin Hematol 1999;36:233–41. 708. Tang SC, Hewitt K, Reis MD, Berinstein NL. Immunosuppressive
688. Stein R, Qu Z, Cardillo TM et al. Antiproliferative activity of a toxicity of CAMPATH-1H monoclonal antibody in the treatment
humanized anti-CD74 monoclonal antibody, hLL1, on B-cell of patients with recurrent low grade lymphoma. Leuk Lymphoma
malignancies. Blood 2004;104:3705–11. 1996;24:93–101.
689. Stein R, Mattes MJ, Cardillo TM et al. CD74: a new candidate 709. Tarhini AA, Kirkwood JM. Tremelimumab, a fully human mono-
target for the immunotherapy of B-cell neoplasms. Clin Cancer clonal IgG2 antibody against CTLA4 for the potential treatment
Res 2007;13(18 Pt 2):5556s–63s. of cancer. Curr Opin Mol Ther 2007;9:505–14.
690. Stemmler HJ, Kahlert S, Brudler O, et al. High efficacy of gem- 710. Tedesco KL, Thor AD, Johnson DH, et al. Docetaxel combined
citabine and cisplatin plus trastuzumab in patients with HER2- with trastuzumab is an active regimen in HER-2 3 + overexpress-
overexpressing metastatic breast cancer: a phase II study. Clin ing and fluorescent in situ hybridization-positive metastatic
Oncol 2005;17:630–5. breast cancer: a multi-institutional phase II trial. J Clin Oncol
691. Steplewski Z, Sun LK, Shearman CW, et al. Biological activity of 2004;22:1071–7.
human-mouse IgG1, IgG2, IgG3, and IgG4 chimeric monoclonal 711. Teillaud LJ. Engineering of monoclonal antibodies and antibody-
antibodies with antitumor specificity. Proc Natl Acad Sci USA based fusion proteins: successes and challenges.Expert Opin Biol
1988;85:4852–6. Ther 2005;5 Suppl 1:S15–27.
692. Stevenson FK, Wrightham M, Glennie MJ, et al. Antibodies to 712. Telli ML, Hunt SA, Carlson RW, et al. Trastuzumab-related
shared idiotypes as agents for analysis and therapy for human B cardiotoxicity: calling into question the concept of reversibil-
cell tumors. Blood 1986;68:430–6. ity. J Clin Oncol 2007;25:3525–33.
693. Stevenson FK, Bell AJ, Cusack R, et al. Preliminary studies for an 713. Tempero MA, Sivinski C, Steplewski Z, et al. Phase II trial of
immunotherapeutic approach to the treatment of human myeloma interferon gamma and monoclonal antibody 17–1A in pancre-
using chimeric anti-CD38 antibody. Blood 1991;77:1071–79. atic cancer: biologic and clinical effects. J Clin Oncol
694. Stevenson GT. CD38 as a therapeutic target. Mol Med 1990;8:2019–26.
2006;12:345–6. 714. Thieblemont C, Bouafia F, Hornez E, et al. Maintenance therapy
695. Stevenson GT, Anderson VA, Leong WS. Engineered antibody for with a monthly injection of alemtuzumab prolongs response
treating lymphoma. Recent Results Cancer Res 2002;159:104–12. duration in patients with refractory B-cell chronic lymphocytic
696. Straatsma BR, Nusinowitz S, Young TA, et al. Surveillance of the leukemia/small lymphocytic lymphoma (B-CLL/SLL). Leuk
eye and vision in clinical trials of CP-675,206 for metastatic Lymphoma. 2004;45:711–4.
melanoma. Am J Ophthalmol 2007;143:958–69. 715. Thienelt CD, Bunn PA Jr, Hanna N, et al. Multicenter phase I/II
697. Strauss SJ, Morschhauser F, Rech J, et al. Multicenter phase II study of cetuximab with paclitaxel and carboplatin in untreated
trial of immunotherapy with the humanized anti-CD22 antibody, patients with stage IV non-small-cell lung cancer. J Clin Oncol
epratuzumab, in combination with rituximab, in refractory or 2005;23:8786–93.
recurrent non-Hodgkin’s lymphoma. J Clin Oncol 716. Thomas DA, O’Brien S, Giles FJ, et al. Single agent Rituxan in
2006;24:3880–6. early stage chronic lymphocytic leukemia. Blood 2001;98:364a
698. Sugimoto H, Hamano Y, Charytan D, et al. Neutralization of cir- [abstract 1533].
culating vascular endothelial growth factor (VEGF) by anti- 717. Thomas DA, O’Brien S, Buesco-Ramos C, et al. Rituximab in
VEGF antibodies and soluble VEGF receptor 1 (sFlt-1) induces relapsed or refractory hairy cell leukemia. Blood 2003;102:3906–11.
proteinuria. J Biol Chem 2003;278:12605–8. 718. Thomas DA, Faderl S, O’Brien S, et al. Chemoimmunotherapy
699. Sumner WC, Foraker AG. Spontaneous regression of human mel- with hyper-CVAD plus rituximab for the treatment of adult
anoma: clinical and experimental studies. Cancer 1960;13:79–81. Burkitt and Burkitt-type lymphoma or acute lymphoblastic leu-
700. Surfus JE, Hank JA, Oosterwijk E, et al. Anti-renal cell carci- kemia. Cancer 2006;1;106:1569–80.
noma chimeric antibody G250 facilitates antibody-dependent 719. Thompson CH, Stacker SA, Salehi N, et al. Immunoscintigraphy
cellular cytotoxicity with in vitro and in vivo interleukin-2- for detection of lymph node metastases from breast cancer.
activated effectors. J Immunother Emphas Tumor Immunol 1984;2:1245–7.
1996;184–91. 720. Thurin J, Thurin M, Kimoto Y, et al. Monoclonal antibody-
701. Swanson JA, Hoppe AD. The coordination of signaling during Fc defined correlations in melanoma between levels of GD2 and
receptor-mediated phagocytosis. J Leukoc Biol. 2004;76:1093–103. GD3 antigens and antibody-mediated cytotoxicity. Cancer Res
702. Swisher EM, Shawler DL, Collins HA, et al. Expression of 1987;47:1229–33.
shared idiotypes in chronic lymphocytic leukemia and small lym- 721. Tibben JG, Boerman OC, Massuger LF, et al. Pharmacokinetics,
phocytic lymphoma. Blood 1991;77:1977–82. biodistribution and biological effects of intravenously adminis-
703. Tabernero J, Van Cutsem E, Diaz-Rubio E, et al. Phase II trial of tered bispecific monoclonal. antibody OC/TR F(ab’)2 in ovarian
cetuximab in combination with fluorouracil, leucovorin, and carcinoma patients. Int J Cancer 1996;66:477–83.
oxaliplatin in the first-line treatment of metastatic colorectal can- 722. Tibes R, Keating MJ, Ferrajoli A, et al. Activity of alemtuzumab
cer. J Clin Oncol 2007;25:5225–32. in patients with CD52-positive acute leukemia.Cancer.
704. Taetle R, Castagnola J, Mendelsohn J. Mechanisms of growth 2006;106:2645–51.
inhibition by anti-transferrin receptor Mab. Cancer Res 723. Tobinai K, Kobayashi Y, Itoh K, et al. Japanese multicenter phase
1986;46:1759–63. II and pharmacokinetic study of rituximab in relapsed or refractory
404 Monoclonal antibody therapy

patients with aggressive B-cell lymphoma. Ann Oncol 2004; 743. van der Kolk LE, Grillo-Lopez AJ, Baars JW, et al. Treatment of
15:821–30. relapsed B-cell non-Hodgkin’s lymphoma with a combination of
724. Tolcher AW, Mita M, Meropol NJ, et al. Phase I pharmacokinetic chimeric anti-CD20 monoclonal antibodies (rituximab) and G-CSF:
and biologic correlative study of mapatumumab, a fully human final report on safety and efficacy. Leukemia. 2003;17:1658–64.
monoclonal antibody with agonist activity to tumor necrosis 744. Vanhoefer U, Tewes M, Rojo F, et al. Phase I study of the
factor-related apoptosis-inducing ligand receptor-1. J Clin Oncol humanized antiepidermal growth factor receptor monoclonal
2007;25:1390–5. antibody EMD72000 in patients with advanced solid tumors
725. Tomas JF, Montalban C, Martinez-Lopez J, et al. Induction with that express the epidermal growth factor receptor. J Clin Oncol
fludarabine, cyclophosphamide, and rituximab followed by 2004;22:175–84.
maintenance with rituximab: results of a prospective study in 75 745. Van Oers MH, Klasa R, Marcus RE, et al. Rituximab mainte-
patients (LNHF-03). Blood 2006;108:782a [abstract 2762]. nance improves clinical outcome of relapsed/resistant follicular
726. Treon SP, Agus DB, Link B, et al. CD-20 directed antibody-me- non-Hodgkin lymphoma in patients both with and without ritux-
diated immunotherapy induces responses and facilitates hemato- imab during induction: results of a prospective randomized phase
logic recovery in patients with Waldenstrom’s macroglobulinemia. 3 intergroup trial. Blood 2006;108:3295–301.
J Immunother 2001;24:272–9. 746. Van Sorge NM, Van der Pol WL, van de Winkel JG. FcgammaR
727. Treon SP, Pilarski LM, Belch AR, et al. CD20-directed serotherapy polymorphisms: Implications for function, disease susceptibility
in patients with multiple myeloma: biologic considerations and and immunotherapy. Tissue Antigens 2003;61:189–202.
therapeutic applications. J Immunother. 2002;25:72–81. 747. Van Zaanen HC, Lokhorst HM, Aarden LA, et al. Chimaeric anti-
728. Treon SP, Emmanouilides C, Kimby E, et al. Extended ritux- interleukin 6 monoclonal antibodies in the treatment of advanced
imab therapy in Waldenstrom’s macroglobulinemia. Ann Oncol multiple myeloma: a phase I dose-escalating study. Br J Haematol
2005a;16:132–8. 1998;102:783–90.
729. Treon SP, Hunter Z, Barnagan AR, et al. CHOP plus rituximab 748. van Zanten-Przybysz I, Molthoff C, Gebbinck JK et al. Cellular
therapy in Waldenstrom’s macroglobulinemia. Clin Lymphoma and humoral responses after multiple injections of unconjugated
2005b;5:273–7. chimeric monoclonal antibody MOv18 in ovarian cancer patients:
730. Tripathy D, Seidman A, Keefe D, et al. Effect of cardiac dysfunc- a pilot study. J Cancer Res Clin Oncol 2002;128:484–92.
tion on treatment outcomes in women receiving trastuzumab for 749. Varga Z, de Mulder P, Kruit W, et al. A prospective open-label
HER2-overexpressing metastatic breast cancer. Clin Breast single-arm phase II study of chimeric monoclonal antibody
Cancer 2004;5:293–8. cG250 in advanced renal cell carcinoma patients. Folia Biol
731. Trowbridge IS, Domingo DL. Anti-transferrin receptor monoclo- 2003;49:74–7.
nal antibody and toxin-antibody conjugates affect growth of 750. Venturini M, Bighin C, Monfardini S, et al. Multicenter phase II
human tumour cells. Nature 1981;294:171–3. study of trastuzumab in combination with epirubicin and doc-
732. Trowbridge IS, Lopez F. Monoclonal antibody to transferrin etaxel as first-line treatment for HER2-overexpressing metastatic
receptor blocks transferrin binding and inhibits human tumor cell breast cancer. Breast Cancer Res Treat 2006;95:45–53.
growth in vitro. Proc Natl Acad Sci USA 1982;79:1175–9. 751. Vermorken JB, Trigo J, Hitt R, et al. Open-label, uncontrolled,
733. Tubbs RR, Pettay JD, Roche PC, et al. Discrepancies in clinical multicenter phase II study to evaluate the efficacy and toxicity of
laboratory testing of eligibility for trastuzumab therapy: apparent cetuximab as a single agent in patients with recurrent and/or meta-
immunohistochemical false-positives do not get the message. static squamous cell carcinoma of the head and neck who failed to
J Clin Oncol 2001;19:2714–21. respond to platinum-based therapy. J Clin Oncol 2007;25:2171–7.
734. Uemura H, Nakagawa Y, Yoshida K, et al. MN/CA IX/G250 as a 752. Vincenzi B, Santini D, Rabitti C, et al. Cetuximab and irinotecan
potential target for immunotherapy of renal cell carcinomas. Br J as third-line therapy in advanced colorectal cancer patients: a
Cancer 1999;81:741–6. single centre phase II trial. Br J Cancer 2006;94:792–7.
735. Ullenhag GJ, Spendlove I, Watson NF, et al. A neoadjuvant/adju- 753. Vitetta ES, Uhr JW. Monoclonal antibodies as agonists: an
vant randomized trial of colorectal cancer patients vaccinated expanded role for their use in cancer therapy. Cancer Res
with an anti-idiotypic antibody, 105AD7, mimicking CD55. Clin 1994;54:5301–9.
Cancer Res 2006;12:7389–96. 754. Vitolo U, Boccomini C, Ladetto M, et al. High clinical and
736. Uppenkamp M, Engert A, Diehl V, et al. Monoclonal antibody molecular response rate in elderly patients with advanced stage
therapy with CAMPATH-1H in patients with relapsed high- and follicular lymphoma treated at diagnosis with a brief chem-im-
low-grade non-Hodgkin’s lymphomas: a multicenter phase I/II munotherapy FND + rituximab. Blood 2002;100:359a [1392].
study. Ann Hematol 2002;81:26–32. 755. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of
737. Urba WJ, Ewel C, Kopp W, et al. Anti-CD3 monoclonal antibody trastuzumab as a single agent in first-line treatment of HER2-
treatment of patients with CD3-negative tumors: a phase IA/B overexpressing metastatic breast cancer. J Clin Oncol
study. Cancer Res 1992; 52:2394–2401. 2002;20:719–26.
738. Vadhan-Raj S, Cordon-Cardo C, Carswell E, et al. Phase I trial of 756. Vonderheide RH, Flaherty KT, Khalil M, et al. Clinical activity
mouse monoclonal antibody against GD3 ganglioside in patients and immune modulation in cancer patients treated with
with melanoma: induction of inflammatory responses at tumor CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin
sites. J Clin Oncol 1988; 6:1636–48. Oncol 2007;25:876–83.
739. Valesini G, Iannuccelli C, Marocchi E et al. Biological and 757. Vose JM, Link BK, Grossbard ML, et al. Phase II study of ritux-
clinical effects of anti-TNF alpha treatment. Autoimmun Rev imab in combination with CHOP chemotherapy in patients with
2007;7:35–41. previously untreated, aggressive non-Hodgkin’s lymphoma. J
740. Valone FH, Kaufman PA, Guyre PM, et al. Phase I1/Ib trial of Clin Oncol 2001;19:389–97
bispecific antibody MDX-210 in patients with advanced breast or 758. Vose JM, Link BK, Grossbard ML, et al. Long-term update of a
ovarian cancer that overexpresses the proto-oncogene HER-2/ phase II study of rituximab in combination with CHOP chemo-
neu. J Clin Oncol 1995;13:2281–92. therapy in patients with previously untreated, aggressive non-
741. Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial Hodgkin’s lymphoma. Leuk Lymphoma 2005;46:1569–73.
of panitumumab plus best supportive care compared with best 759. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Phase II
supportive care alone in patients with chemotherapy-refractory trial of bevacizumab and irinotecan in recurrent malignant
metastatic colorectal cancer. J Clin Oncol 2007;25:1658–64. glioma. Clin Cancer Res 2007;13:1253–9.
742. Van Cutsem E, Siena S, Humblet Y, et al. An open-label, single- 760. Vulfovich M, Saba N. Technology evaluation: mapatumumab.
arm study assessing safety and efficacy of panitumumab in Curr Opin Mol Ther 2005;7:502–10.
patients with metastatic colorectal cancer refractory to standard 761. Waldmann T. Anti-Tac (daclizumab, Zenapax) in the treatment
chemotherapy. Ann Oncol 2008;19:92–8. of leukemia, autoimmune diseases, and in the prevention of
Robert O. Dillman 405

allograft rejection: a 25-year personal odyssey. J Clin Immunol 783. Wierda W, O’Brien S, Wen S, et al. Chemoimmunotherapy
2007a;27:1–18. with fludarabine, cyclophosphamide, and rituximab for
762. Waldmann TA. The structure, function, and expression of inter- relapsed and refractory chronic lymphocytic leukemia. J Clin
leukin-2 receptors on normal and malignant lymphocytes. Oncol 2005;23:4070–8.
Science 1986;232:727–32. 784. Wilkinson I, Jackson C-JC, Lang GM, et al. Tolerance induction
763. Waldmann TA. The IL-2/IL-15 receptor systems:targets for in mice by conjugates of monoclonal immunoglobulins and
immunotherapy. Clin Immunol 2002;22:51–6 monomethoxypolyethylene glycol: transfer of tolerance by T
764. Waldmann TA. Daclizumab (anti-Tac, Zenapax) in the treatment cells and by T cell extracts. J Immunol 1987;139:326–31.
of leukemia/lymphoma. Oncogene 2007b;26:3699–703. 785. Willett CG, Boucher Y, di Tomaso E, et al. Direct evidence that
765. Waldmann TA, Goldman CK, Bongiovanni KF, et al. Therapy of the VEGF-specific antibody bevacizumab has antivascular effects
patients with human T-cell lymphotropic virus I-induced adult in human rectal cancer. Nat Med 2004;10:145–7.
T-cell leukemia with anti-tac, a monoclonal antibody to the 786. Wilson WH, Gutierrez M, O’Connor P, et al. The role of ritux-
receptor for interleukin-2. Blood 1988;72:1805–16. imab and chemotherapy in aggressive B-cell lymphoma: a pre-
766. Waldmann TA, White JD, Goldman CK, et al. The interleukin-2 liminary report of dose-adjusted EPOCH-R. Semin Oncol
receptor: a target for monoclonal antibody treatment of human 2002;29(1 Suppl 2):41–7.
T-cell lymphotrophic virus I-induced adult T-cell leukemia. 787. Winkler U, Jensen M, Manzke O, et al. Cytokine-release syndrome
Blood 1993;82:1701–12. in patients with B-cell chronic lymphocytic leukemia and high
767. Wallach D, Fellous M, Revel M. Preferential effect of gamma lymphocyte counts after treatment with an anti-CD20 monoclonal
interferon on the synthesis of HLA antigens and their mRNAs in antibody (rituximab, IDEC-C2B8). Blood 1999;94:2217–24.
human cells. Nature 1982;299:833–835. 788. Winter G, Milstein C. Man-made antibodies. Nature 1991;349:293–9.
768. Walport MJ. Complement. N Engl J Med 1991;344:1058–66, 789. Wiseman C, Hammock V, Barton L, et al. Clinical responses of
1140–4. primary human CNS tumors to OKT3/cyclophosphamide. J
769. Ward RL, Packham D, Smythe AM, et al. Phase I clinical trial of Immunother 1994;16:245 [abstract].
the chimeric monoclonal antibody (c30.6) in patients with meta- 790. Witzig TE, Gordon LI, Cabanillas F, et al. Randomized controlled
static colorectal cancer. Clin Cancer Res 2000;6:4674–83. trial of yttrium-90-labeled ibritumomab tiuxetan radioimmuno-
770. Watanabe M, Wallace PK, Keler T, et al. Antibody dependent cel- therapy versus rituximab immunotherapy for patients with
lular phagocytosis (ADCP) and antibody dependent cellular relapsed or refractory low-grade, follicular, or transformede B-cell
cytotoxicity (ADCC) of breast cancer cells mediated by bispe- non-Hodgkin’s lymphoma. J Clin Oncol 2002;20:2453–63.
cific antibody, MDX-210. Breast Cancer Res 1999;53:199–207. 791. Witzig TE, Vukov AM, Habermannn TM, et al. Rituximab ther-
771. Wedam SB, Low JA, Yang SX, et al. Antiangiogenic and antitu- apy for patients with newly diagnosed, advanced-stage, follicular
mor effects of bevacizumab in patients with inflammatory and grade I non-Hodgkin’s lymphoma: a phase II trial in the North
locally advanced breast cancer. J Clin Oncol 2006;24:769–77. Central Cancer Treatment Group. J Clin Oncol 2005;23:1103–8.
772. Weiner LM. Bispecific antibodies in cancer therapy. Cancer J 792. Wohrer S, Puspok A, Drach J, et al. Rituximab, cyclophosph-
2000;6(suppl 3):S265–69. amide, doxorubicin, vincristine and prednisone (R-CHOP) for
773. Weiner LM, Steplewski Z, Koprowski H et al. Biologic effects of treatment of early-stage gastric diffuse large B-cell lymphoma.
gamma interferon pre-treatment followed by monoclonal anti- Ann Oncol 2004;15:1086–90.
body 17–1A administration in patients with gastrointestinal car- 793. Wong JF, Colvin RB. Bi-specific monoclonal antibodies.
cinoma. Hybridoma 1986;5 Suppl 1:S65–77. Selective binding and complement fixation to cells that express
774. Weiner LM, Moldofsky PJ, Gatenby RA et al. Antibody delivery two different surface antigens. J Immunol 1987;139:1369–74.
and effector cell activation in a phase II trial of recombinant gam- 794. Woodhouse CS, Morgan AC Jr. Murine monoclonal IgG3 anti-
ma-interferon and the murine monoclonal antibody CO17–1A in bodies to human colorectal tumor-associated antigens: produc-
advanced colorectal carcinoma. Cancer Res 1988a;48:2568–73. tion and characterization of antibodies active in both
775. Weiner LM, Steplewski Z, Koprowski H, et al. Divergent dose- antibody-dependent cellular cytotoxicity and complement-medi-
related effects of gamma-interferon therapy on in vitro antibody- ated cytolysis. Cancer Res 1989;49:2766–72.
dependent cellular and nonspecific cytotoxicity by human 795. Wright PW, Hellstrom KE, Hellstrom IE, Bernstein ID. Serotherapy
peripheral blood monocytes. Cancer Res 1988b;48:1042–46. of malignant disease. Med Clin North Am 1976:60:607–22.
776. Weiner LM, Harvey E, Padavic-Shaller K, et al. Phase II multi- 796. Wurflein D, Dechant M, Stockmeyer B, et al. Evaluating antibod-
center evaluation of prolonged murine monoclonal antibody ies for their capacity to induce cell-mediated lysis of malignant B
17–1A therapy in pancreatic carcinoma. J Immunother 1993; cells. Cancer Res 1998;58:3051–58.
13:110–6. 797. Xiong HQ, Rosenberg A, LoBuglio A, et al. Cetuximab, a mono-
777. Welt S, Divgi CR, Scott AM, et al. Antibody targeting in meta- clonal antibody targeting the epidermal growth factor receptor, in
static colon cancer: a phase I study of monoclonal antibody F19 combination with gemcitabine for advanced pancreatic cancer: a
against a cell-surface protein of reactive tumor stromal fibro- multicenter phase II Trial. J Clin Oncol 2004;22:2610–6.
blasts. J Clin Oncol 1994;12:1193–203. 798. Yang H, Rosove HM, Figlin RA. Tumor lysis syndrome occur-
778. Welt S, Ritter G, Williams C Jr, et al. Phase I study of anticolon ring after the administration of rituximab in lymphoproliferative
cancer humanized antibody A33. Clin Cancer Res disorders: high-grade non-Hodgkin’s lymphoma and chronic
2003;9:1338–46. lymphocytic leukemia. Am J Hematol 1999;62:247–50.
779. Welte K, Miller G, Chapman PB, et al. Stimulation of T lympho- 799. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of
cyte proliferation by monoclonal antibodies against GD3 ganglio- bevacizumab, an anti-vascular endothelial growth factor
side. J Immunol 1987;139:1763–71. antibody, for metastatic renal cancer. N Engl J Med
780. Wendtner CM, Ritgen M, Schweighofer CD, et al. Consolidation 2003;349:427–34.
with alemtuzumab in patients with chronic lymphocytic leuke- 800. Yang JC, Hughes M, Kammula U, et al. Ipilimumab (anti-CTLA4
mia (CLL) in first remission – experience on safety and efficacy antibody) causes regression of metastatic renal cell cancer associated
within a randomized multicenter phase III trial of the German with enteritis and hypophysitis. J Immunother 2007;30:825–30.
CLL Study Group (GCLLSG). Leukemia 2004;18:1093–101. 801. Yasutomi J, Koda K, Saito N, et al. Identification of the immuno-
781. Wettendorff M, Hiopoulos D, Tempero M, et al. Idiotypic cas- reactive peptide sequence for AgSK1, an adenocarcinoma-
cades in cancer patients treated with monoclonal antibody restricted antigen. Tissue Antigens 2000;55:157–61.
CO17–1A. Proc Natl Acad Sci USA 1989; 86:3787–91. 802. Yoshikawa K, Ueda R, Obata Y, et al. Human monoclonal anti-
782. White CA. Rituximab immunotherapy for non-Hodgkin’s lym- body reactive to stomach cancer produced by mouse-human
phoma. Cancer Biother Radipharm. 1999;14:241–50. hybridoma technique. Jpn J Cancer Res 1986;77:1122–33.
406 Monoclonal antibody therapy

803. Younes A, Romaguera J, Hagemeister F. A pilot study of ritux- tion of patients with Her2-overexpressing disease. Lung Cancer
imab in patients with recurrent, classic Hodgkin disease. Cancer. 2004;44:99–110.
2003;98:310–4. 813. Zinzani PL, Pulsoni A, Perrotti A, et al. Fludarabine plus mitox-
804. Yu AL, Martina M, Uttenreuther-Fischer MM, et al. Phase I trial antrone with and without rituximab versus CHOP with and with-
of a human-mouse chimeric anti-disialoganglioside monoclonal out rituximab as front-line treatment for patients with follicular
antibody ch14.18 in patients with refractory neuroblastoma and lymphoma. J Clin Oncol 2004;22:2654–61.
osteosarcoma. J Clin Oncol 1998;16:2169–80. 814. Zinzani PL, Alinari L, Tani M, et al. Preliminary observations of a
805. Zhang D, Pier T, McNeel DG, et al. Effects of a monoclonal anti- phase II study of reduced-dose alemtuzumab treatment in patients
alphavbeta3 integrin antibody on blood vessels – a pharmacody- with pretreated T-cell lymphoma. Haematologica 2005;90:702–3.
namic study. Invest New Drugs 2007;25:49–55. 815. Wong KK, Sweet DL, Variakoujis D. The treatment of lympho-
806. Zhang W, Gordon M, Press OA, et al. Cyclin D1 and epidermal blastic lymphoma with antithymocyte globulin. Cancer 1981;50:
growth factor polymorphisms associated with survival in patients 57–61.
with advanced colorectal cancer treated with Cetuximab. 816. Foon KA, Bernhard MI, Oldham RK. Monoclonal antibody ther-
Pharmacogenet Genomics 2006;16:475–83. apy: assessment by animal tumor models. J Biol Response Modif
807. Zhou J, Chen J, Mokotoff M, et al. Bombesin/gastrin-releasing pep- 1982;1:277–304.
tide receptor: a potential target for antibody-mediated therapy 817. Davidson MB, Thakkar S, Hix JK, et al. Pathophysiology, clini-
of small cell lung cancer. Clin Cancer Res 2003a;9:4953–60. cal consequences, and treatment of tumor lysis syndrome. Am J
808. Zhou J, Johnson Je, Ghetie V, et al. Generation of mutated vari- Med 2004;116:546–54.
ants of the human form of the MHC class I-related receptor, 818. Tiu RV, Mountantonakis SE, Dunbar AJ, et al. Tumor lysis syn-
FcRn, with incrased affinity for mouse immunoglobuolin G. J drome. Semin Thromb Hemost 2007; 33:397–407.
Mol Biol 2003b;332:901–13. 819. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the
809. Zhu AX, Blaszkowsky LS, Ryan DP, et al. Phase II study of gem- treatment of cladribine-failed patients with hairy cell leukemia.
citabine and oxaliplatin in combination with bevacizumab in Blood 2003;102:810–813.
patients with advanced hepatocellular carcinoma. J Clin Oncol 820. Armstrong A, Eck SL. EpCAM: A new therapeutic target for an
2006;24:1898–903. old cancer antigen. Cancer Biol Ther 2003;2:320–326.
810. Ziada A, Barqawi A, Glode LM, et al. The use of trastuzumab in 821. Mellstedt H, Frodin J-E, Masucci G, et al. The therapeutic use of
the treatment of hormone refractory prostate cancer; phase II monoclonal antibodies in colorectal carcinoma. Semin Oncol
trial. Prostate 2004;60:332–7. 1991; 18:462–477.
811. Ziegler LD, Palazzolo, Cunningham J, et al. Phase I trial of 822. Riethmuller G, Schneider-Gadicke E, Schlimok G, et al.
murine monoclonal antibody L6 in combination with subcutane- Randomized trial of monoclonal antibody for adjuvant therapy
ous interleukin-2 in patients with advanced carcinoma of the of resected Dukes C colorectal carcinoma. Lancet 1994;
breast, colorectum, and lung. J Clin Oncol 1992;10:1470–8. 343:1177–1183.
812. Zinner RG, Glisson BS, Fossella FV, et al. Trastuzumab in com- 823. Spina M, Jaeger U, Sparano JA et al. Rituximab plus infusional
bination with cisplatin and gemcitabine in patients with Her2- cyclophosphamide, doxorubicin, and etoposide in HIV-associated
overexpressing, untreated, advanced non-small cell lung cancer: non-Hodgkin lymphoma: pooled results from 3 phase 2 trials.
report of a phase II trial and findings regarding optimal identifica- Blood 2005;105:1891–1897.
11 Immunotoxins
ARTHUR E. FRANKEL, JUNG-HEE WOO, AND DAVID M. NEVILLE

Introduction The information on toxin structure has been extremely


useful in immunotoxin design.
Immunotoxins are protein molecules composed of a cell
surface-directed ligand covalently linked to a peptide
cytotoxin. This definition excludes a number of impor-
Type I plant A-B Toxins
tant therapeutic compounds with distinct pharmacologic The type I A-B toxins are produced by plants and bacte-
properties which react with intracellular targets. Such ria. The plant toxins – ricin, abrin, and viscu-min (mistle-
molecules would be unlikely to find and react with their toe lectin I) – are encoded by single exons. The sequence
intracellular targets due to the permeability barrier of the from the N-terminus for each includes a signal peptide, a
plasma membrane. Thus, the ligands must bind cell sur- catalytic domain, and an alkaline protease-sensitive
face receptors or antigens. Ligands which have been disulfide loop, followed by a cell binding domain [40–42].
used include monoclonal antibodies and antibody frag- The linker is cleaved by plant vesicle endoproteases,
ments, adhesion molecules, growth factors, and cytok- N-linked glycosyl groups are added, and the signal
ines. The toxophore must be peptide in nature. This peptide is removed by a signal peptidase prior to toxin
excludes radiolabels such as 90Y, 213Bi, or 131I and small secretion [43]. Thus, the fully processed, native proteins
molecular weight drugs such as calicheamicin and doxo- are hetero-dimers of approximately 60,000 Mr, with a
rubicin [1, 2]. These immunoconjugates are the subjects 30,000 Mr catalytic subunit (A chain) and a 30,000 Mr
of other chapters in this book. We have further restricted cell binding subunit (B chain). The B chains have Ω-loop
the toxin moieties to cytotoxins. Thus, peptides which structures (Fig. 1a) [44–46]. There are three lectin binding
modify coagulation [3], complement [4], or immune sites/subunit which bind (3-galactosyl residues on mam-
responses [5] are not considered here. Conjugates with malian cell surface glycoproteins (Fig. 2a) [47, 48]).
these compounds are important potential therapeutics There are also extensive salt and hydrophobic bonds
and are considered separately in chapters discussing between the A and B chains which stabilize the heterodi-
anti-angiogenesis and cell-directed therapies. There are meric structures [44]. After cell binding, the heterodimers
three major classes of peptide cytotoxins [6, 7]. Class I internalize into endosomes using both clathrin-dependent
toxins are proteins which are intracellular enzymes. They and clathrin independent pathways [49], and, via a Rab9-
catalytically modify critical intracellular functions. Class independent process, reach the transreticular Golgi [50].
II toxins bind to cell surfaces and trigger intracellular In the Golgi, the toxins bind endoplasmic reticulum
signal pathways. Class III toxins are pore-forming pep- shuttle proteins which undergo retrograde transport via
tides which cause leaks in the plasma membrane. COP-I-independent Rab6 regulated vesicles [51]. The
Immunotoxins have been prepared with toxins of each toxins are then transported to the endoplasmic reticulum
class, as listed in Table 1 and discussed below. We will (ER) [52]. In the ER, the toxin is reduced, the A chain
first review the structure and molecular mechanisms of unfolds with the help of chaperones, and the A chain
cell intoxication for the peptide cytotoxins used in prepa- translocates to the cytosol using the Sec61p transposon
ration of immunoconjugates. [53]. In the cytosol the A chain refolds. The A chain has
an α-helical/β-sheet open sandwich tertiary structure
with a large open catalytic cleft. The A chains function as
Peptide Cytotoxins RNA N-glycosidases removing a critical adenine base
from the 28S ribosomal RNA (Fig. 3) [54]. This alters the
The DNA sequence, amino acid sequence, and three- interaction of elongation factor 2 (EF2) with the ribosome
dimensional structures for most of the toxins used in and prevents the structural modifications leading the GTP
conjugate synthesis have been defined. In most cases hydrolysis (Fig. 4). Protein elongation is blocked irre-
separate domains contribute to different toxin functions. versibly (Fig. 5). Cell death follows.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 407
© Springer Science + Business Media B.V. 2009
408 Immunotoxins

Table 1. Peptide cytotoxins used in immunotoxin synthesis


Protein Type Structure Order Source (species) Reference
Ricin I A-B Plant Ricinus communis [8]
Abrin I A-B Plant Abrus precatorias [9]
Viscumin I A-B Plant Viscumalbum [10]
Nigrin b I A-B Plant Sambucus nigra [607]
Gelonin I A Plant Geloniummulfiflorum [11]
Saporin I A Plant Saponaria officinalis [12]
Pokeweed antiviral protein I A Plant Phytolacca americana [13]
Luffin I A Plant Luffaa egyptiaca [14]
Bouganin I A Plant Bougainvillea spectabilis [15]
Trichokirin I A Plant Tiichosantheskifilowfi [16]
Trichosanthin I A Plant Tiichosantheskifilowfi [349]
BRIP I A Plant Hordeum vulgare [17]
Ebulin I A Plant Sambucus ebulus [558]
Bryodin I A Plant Bryonia dimca [18, 21]
Momordin I A Plant Momordica charanfia [19]
Momorcochin I A Plant Momorfica coohinchinensis [20]
Moschatin I A Plant Cucurbita moschata [571]
Dianthin 30 I A Plant Dianthus caryophyllus [22]
Ocymoidine I A Plant Saponaria ocymoides [23]
Pyramidatine I A Plant Vaccaria pyramidata [23]
Colocin 1 I A Plant Citrullus colocynthis [440]
Botulinum neurotype type C I A-B Bacteria Clostridia botulinum [560]
Diphtheria toxin I A-B Bacteria Corynbacterium diphthefiae [24]
Pseudomonas exotoxin I A-B Bacteria Pseudomonas aeruginosa [25]
Anthrax toxins I Binary Bacteria Bacillus anthracis [26]
Seminal ribonuclease I A Cow Bos taurus [27]
Bosinophil neurotoxin I A Human Homo sapiens [28]
Angiogenin I A Human Homo sapiens [29]
Pancreatic ribonuclease I A Human Homo sapiens [30]
RibonucleaseA I A Cow BosTaurus [31]
Ranpirnase I A Frog Rana pipiens [32]
Barnase I A Bacteria Bacillus amyloliquefaciens [579]
a-Sarcin I A Fungi Aspergillus giganteus [33]
Restrictocin I A Fungi Aspergillus restfictus [34]
Clavin I A Fungi Aspergillus clavatus [35]
Mitogillin I A Fungi Aspergillus fumigatus [93]
Bax I A Human Homo sapiens [167]
D-(KLAKLAK)2 I A Synthetic [563]
Phospholipase C II Bacteria Clostfidium perfringens [36]
TNF II Human Homo sapiens [574]
sTRAIL II Human Homos sapiens [590]
Hemolytic toxin III Sea anemone Stoichactis helianthus [37]
Cecropin/Shiva III Insect Hyalophora ceaopia [38]
Pyrularia thionin III Plant Pyrulafia pubera [39]
Proaerolysin III Bacteria Aeromonas hydrophila [548]
Cyt1Aa Toxin III Bacteria Bacillus thuringiensis [559]

targeting sequence of 25–29 amino acid residues. The


Type I plant A toxins signal peptide and C-terminal extension are processed
The type I plant A toxins are 25–31 kDa M, proteins. on secretion, thus protecting the cytosolic plant ribo-
They have similar primary amino acid sequences [55– somes from intoxication. The three-dimensional struc-
57], and similar three-dimensional structures [55, 56, ture contains eight α-helices and a β-sheet composed
58–60], and are all ribosomal RNA N-glycosidases of six strands. The sequence, structure, and enzyme
which remove the same adenine base from the 28S activity are homologous to the catalytic A subunit of
ribosomal RNA of the mammalian ribosomes [61]. type I plant A-B toxins. There is an open cleft contain-
However, they do not crossreact immunologically. The ing a conserved glutamic acid residue, an arginine resi-
preproproteins have a signal peptide, the catalytic due, and a tyrosine and a tryptophan residue. These
domain, and a C-terminal extension with a vacuolar residues function to bind and stabilize the transition
Arthur E. Frankel et al. 409

state for the C-N bond cleavage [62]. There are three DT has a 25-residue leader sequence followed by 535
conserved lysyl residues in the C-terminus of the amino acid residues [63, 64]. The mature protein has
mature proteins which likely are involved in ribosomal three domains (Fig. 1b) [65, 66]. There is an N-terminal
substrate recognition [56]. catalytic domain (amino acid residues 186), also called
the A fragment. This domain is followed by a 14 amino
acid loop bordered by Cys-186 and Cys-201. The loop is
arginine-rich and is a substrate for endosomal furin endo-
Type I Bacterial A-B Toxins protease. The second domain is the translocation domain
The type I bacterial A-B toxins which have been modi- (amino acid residues 187–381) and contains multiple
fied for targeted therapy include diphtheria toxin (DT) amphipathic helices with two negatively charged amino
and Pseudomonas exotoxin (PE). These toxins will be acid residues (Glu-349 and Asp-352) in a helical hairpin
discussed separately. between two of the helices (TH8 and TH9). The translo-

Figure 1. Drawings of peptide toxin a-carbon backbone structures. (a) ricin; (b) DT; (c) PA; (d) RNase; (e) restrictocin; (f)
Clostridia perfringens PLC; (g) cecropin (similar to Shiva-1); (h) crambin (similar to PT). Based on coordinates determined
from references described in the text. Note that the type I A-B and binary toxins have multiple domains which may subserve
distinct functions (see Fig. 2). Cylinders are α-helices and ribbons are β-strands. In the ricin structure (a), red tubes are RTB
subunit S2-loops, and yellowblue structures are RTA enzymatic subunit. In the DT structure (b), red is β-sheet binding domain,
yellow is translocation domain with amphipathic helices, and green in enzymatic domain. In PA (c), blue is receptor binding
β-sheet domain, green is the heptamerforming and membrane-inserting domain, and red is 20 kDa protease-sensitive frag-
ment. EDN (d) has an α + β structure with two α-helices, a β-sheet, a third α-helix and four additional β-sheets separated by
loops. There are binding sites for bases and phosphates of RNA. The catalytic histidine residues are at the two ends
410 Immunotoxins

Figure 1. (continued) Restrictocin (e) shows structural homology to Rnases. It has catalytic histidines similar to RNase. There
is a three-turn α-helix packed against a five-stranded antiparallel β-sheet. PLC (f) has an N-terminal domain (residues 1–246)
composed of six stacked α-helices. It has the phospholipase C active site. There is a flexible linker (residues 247–255) followed
by a C-terminal domain (residues 256–370) with an eight β-strand jelly-roll topology with Ca++-dependent phospholipid-binding
function. The cecropins (g) are small 36 amino acid residue peptides with a strongly basic N-terminal amphipathic helix linked
via a flexible Gly-Pro linker to a neutral C-terminal amphipathic helix. These stack in membranes as shown creating pores. PT
toxin has two amphipathic Ochelices connected to two antiparallel β-sheets. The related crambin (h) lacks β-sheets, but the
loops serve the same function (Fig. 1 h)
a Ricin
c PE

Galactosyl-terminated Alpa2-macroglobulin
oligosaccharides
Endosomes

KDEL-receptor Endosome

N-glycosidase reaction on
Trans-reticular ribosomes
Golgi
Trans-recticular
Golgi
37κD PE fragment
ER
ER
Translocation
EF2
of RTA
Ribosomes

b DT
d
Heparin-binding 20κDa LF
EGF fragment EF
63κDa
PA fragment
Protease

Endosome
PA receptor

DTA
EF2

Endosome

Ribosomes MAPKK

cAMP

Figure 2. Steps in peptide toxin intoxication of cells. (a) ricin; (b) DT; (c) PE; (d) anthrax toxins. Note for all toxins there is
an initial cell binding event. For the shown type I toxins (a–d), cell binding is followed by internalization. After intracellular
processing and transport the type I toxins escape to the cytosol where they catalytically inactivate key cellular processes. For
type II and III toxins the membrane perturbations lead to altered cell physiology or cell death

Figure 3. Atomic stick model of the S/R stem loop (red) portion of 28S rRNA (black). In addition to elongation factor binding
to the ribosome A site which displaces the peptidyl-tRNA (see Figs. 4 and 5), there is also an rRNA conformation change which
occurs spontaneously to drive elongation. This is catalyzed by other EF2 (EFG) domains (II-III) in which the GTPase activity
modifies ribosome structure. Ricin and type I plant A toxins remove an adenine base (rRNA N-glycosidase activity) from the
large rRNA at a specific location on the S/R conserved stem-loop (S = α-sarcin site and R = ricin site). This stem-loop normally
switches between two states which change overall rRNA and ribosome structure in the large subunit. Once the site is chemically
modified, EF2 or EFG can no longer bind properly and elongation stops. Note that EF2 or EFG interacts with the ribosome at
least two sites in domain IV and II-III in addition to its interaction with the mRNA at the anticodon loop and each of these two
sites are affected separately by type I peptide toxins
412 Immunotoxins

c
TOXIN

O
His O
H C OPO AMP
N
H O
N O

H
OH OH

Glu
N
R N Diphthamide
H
EF2

Figure 4. (a) Structure of EF-G and EF-Tu-Phe-tRNA-GTP ternary complex. Note that domain IV (the lower tip of EF-G and EF2)
resembles the tRNA portion of the EF-Tu-Phe-tRNA-GTP complex. Additional data show that this region is adjacent to the anti-
codon loop and binds the A site of the ribosome. (b) Model of EF2 α-carbon backbone showing in yellow the location of diph-
thamide residue at the tip of domain IV. The diphthamide residue is the target for DT and PE ADP-ribosylation. (c) The enzymatic
reaction catalyzed by DT and PE. The His and Arg residues of the catalytic site of DT and PE interact with the nicotinamide ring
and stabilize the position of the NAD molecule in such a way that the electrophilic carbon atom of the ribose group can interact
with the nucleophilic residues of diphthamide on EF2

aa-tRNA-EF-Tu-GTP cation domain ends in a flexible spacer (amino acid resi-


dues 382–390) which is then connected to a β-sheet rich
50S cell-binding domain (amino acid residues 391–535). DT
EF-Tu-GDP binds to cells via amino acid residues in the cell-binding
A-site
P-site domain to cell-surface-expressed heparin-binding epider-
30S mal growth factor-like growth factor (HB-EGF) precursor
[67, 68]. The amino acid residues of the DT cell-binding
domain, in particular Lys-516, binds a crevice in the
extracellular EGF-like domain of the HB-EGF precursor
(amino acid residues 122–148). There is a critical salt
bridge interaction between Lys-516 of DT and Glu-141
EF-G-GDP of HB-EGF precursor. The HB-EGF precursor associ-
ates on the plasma membrane with CD9 and heparan
EF-G-GDP
sulfate proteoglycan [69, 70]. This complex has a higher
Figure 5. Protein synthesis steps affinity for DT. Animals and cell lines with absent or
Arthur E. Frankel et al. 413

low-affinity receptors for DT are insensitive to the toxin Domain II is the translocation domain. Domain III is the
[67]. After cell binding the DT-HB-EGF precursor com- carboxylterminal third of the molecule and includes
plex undergoes clathrin-dependent endocytosis [71]. residues 405–613. It has an extended catalytic cleft.
Interestingly, the cytoplasmic domain of the HB-EGF Domain III is the enzymatic domain. PE in the blood
precursor lacks an internalization motif (although it has reacts with plasma carboxypeptidase, and the terminal
a membrane proximal Tyr-192) [72]. The complex lysine residue is cleaved [97]. PE612 then binds the
internalizes at 1%/min, and dynamin is required to com- α2-macroglobulin receptor/low density lipoprotein
plete vesicle formation [73]. Once internalized into early receptor-related protein (α2MR/LRP) on the surface of
endosomes [74], DT u Glu zndergoes furin cleavage at mammalian cells including fibroblasts and hepatocytes
the arginine-rich loop [75], low pH-induced protonation [98]. PE612 domain Ia, residue Lys-57 is important in
of the helical hairpin aspartate and glutamate [76], inser- this binding reaction [99]. The PE612, α2 MR/LRP
tion of the TH8 and TH9 amphipathic helices of the trans- complex undergoes receptor-mediated endocytosis
location domain into the vesicle membrane [77], unfolding [100]. Once reaching the endosomes the PE612 under-
of the catalytic domain [78], reduction of the disulfide goes furin cleavage between Arg-279 and Gly-280 [101,
bridge linking the A fragment with the remainder of DT 102], a pH-dependent conformation change [103] fol-
[79, 80], and transfer of the A fragment through the mem- lowed by reduction of the disulfide bond between Cys-
brane to the cytosol (Fig. 2b) [81]. The DT A fragment 265 and Cys-287 [104]. This creates a 28 kDa N-terminal
then catalytically ADP-ribosylates elongation factor 2 fragment and a 37 kDa C-terminal fragment. The
(EF2) on the diphthamide residue in EF2 domain IV [82, C-terminal REDL of the 37 kDa fragment then binds to
83, 92]. A detailed molecular mechanism for the enzy- the KDEL receptor and the complex is routed to the
matic activity has been proposed. First, residues 39–46 of endoplasmic reticulum (ER) [105, 106]. In the ER the
the A fragment ‘active site’ loop bind NAD [84]. The dis- 37 kDa fragment utilizes residues in domain II to trans-
lodged loop then binds EF2 in combination with the locate the cytosol [107]. Thirty-four amino acid residues
exposed NAD. A fragment Glu-148 then attacks the at the N-terminus of the 37 kDa fragment appear critical
strained N-glycosidic bond of the NAD. The Glu-148 for translocation, including Trp-281, Leu-284, and Tyr-
carboxylate group activates the diphthamide imidazole of 289 [108, 109]. There is an unidentified saturable pro-
EF2 for a nucleophilic attack on Cl’N in an SN2-type tein which binds the fragment and facilitates transfer.
displacement reaction. This leads to addition of ADP to Most of the translocation domain helices, with the
the domain IV diphthamide residue. This irreversible exception of the first and last helix, are essential for the
modification prevents the elongation factor from displac- translocation process [110]. The translocon may be part
ing the tRNA petidyl complex from the A site to the P site of the machinery for PE transfer to the cytosol, since the
[85, 86]. Protein synthesis is inactivated, and cells die by translocation occurs in the ER (Fig. 2c). The cytosolic
lysis or programmed cell death [87, 88]. The prolonged C-terminal fragment then catalyzes the ADP-ribosylation
action of a single A fragment molecule in the cytosol is of EF2 similar to DT [111]. Glu-553 is important for
sufficient to inhibit protein synthesis and prevent cell NAD binding. The NAD is cleaved and the ADP moiety
growth and division [81, 89]. Cellular resistance to DT transferred to the diphthamide residue of bound EF2
can occur due to altered DT receptors [67], lack of inter- [112]. Cell death follows by a process indistinguishable
nalization [73], failure of arginine-rich loop proteolysis from DT [81, 89]. Cell resistance to PE has been seen
[75], lack of endosomal acidification [90], too great a dis- secondary to alterations of individual steps in the intoxi-
tance between the DT translocation domain and the vesi- cation process. Cells may lack receptors [113]; cell
cle membrane [71], incomplete inter-domain disulfide intracellular trafficking may be disrupted [114–115];
reduction [79], incomplete A fragment unfolding [78], too endosomal acidification may not occur [116]; furin-
rapid cytosolic proteolysis of the A fragment [81], and mediated cleavage may not occur due to low or abnor-
resistant EF2 [91]. mal furin [117]; EF2 may be mutated so that the
PE is a 66 kDa Mr, protein produced by Pseudomonas diphthamide residue is absent [91]; finally, cells may be
aeruginosa. It consists of 638 amino acid residues resistant to PE-induced apoptosis [87].
including a 25 amino acid hydrophobic leader peptide
[95]. The molecule has three distinct domains [96].
Domain I is an antiparallel β-structure with 17 β-strands.
Type I Bacterial Binary Toxin
It includes amino acid residues 1–252 (Ia) and 365–404 Anthrax toxin produced by Bacillus anthracis is a binary
(Ib). The first 13 strands form an elongated β-barrel. toxin. The catalytic domain and binding domains are on
Domain Ia, is the cell-binding domain. Domain II consists completely separate proteins. Anthrax toxins have two
of residues 253–364 and has six consecutive α-helices. catalytic proteins – edema factor (EF) and lethal factor
414 Immunotoxins

(LF). Either of these proteins can interact with the cell- (14–16 kDa Mr) proteins with an α + β structure (Fig.
binding protein, protective antigen (PA). 1d) [140]. From the N-terminus there are two α-helices,
PA has 764 amino acid residues with a 29 residue sig- a β-sheet, a third α-helix, and four additional β-sheets
nal peptide. Mature PA is a 735 amino acid residue, separated by loops. There are binding sites for bases and
83 kDa secreted protein [118]. It has four domains [119]. phosphates of RNA. The catalytic residues include His-
PA domain 4 (residues 596–735) has an initial hairpin 12, Lys-41, Thr45, and His-119. His-12 and His-119 act
and helix which serves as a flexible linker followed by an as the base and acid, respectively, in the transphospho-
immunoglobulin-like fold which functions in receptor rylation step that generates the cyclic phosphate inter-
binding (Fig. 1c) [120, 121]. The nature of the cell sur- mediate. The roles of His-12 and His-119 are reversed
face receptor is unknown, but there are approximately in the subsequent hydrolysis step (Fig. 6) [141]. Lys-41
30,000 receptors/macrophage [122]. After cell binding, and Thr-45 bind the substrate phosphate and pyrimidine
cell surface furin or PACE4 nicks PA in the middle of base, respectively, to stabilize the transition state
domain 1 (residues 1–258) at residues 164–167 (Fig. 2d) intermediate.
[123]. This releases the N-terminal 20 kDa Mr fragment, The conjugated vertebrate RNases include bovine
PA20. The remainder of PA, PA63, remains on the cell seminal RNase (BS-RNase), human eosinophil derived
surface. A large hydrophobic surface on the residual neurotoxin (EDN), human angiogenin (ang), frog ranpir-
domain 1 of PA63 serves as a site for EF or LF binding. nase, bovine pancreatic RNase A, and human pancreatic
EF and LF are 90 kDa Mr, 770 amino acid residue pro- ribonuclease I (hRNase 1). They each have distinct prop-
teins with a similar N-terminal 250 amino acid domain erties. BS-RNase is a dimeric protein (32 kDa Mr) with
which mediates binding to PA63 [124, 125]. These pro- strong and selective cytotoxicity to tumor cells, germ
teins have distinct C-terminal domains which have ade- cells, and T cells [142]. EDN is a 15 kDa, 134 amino acid
nylate cyclase and Zn2+-dependent metalloprotease residue, highly basic protein which has selective toxicity
activity, respectively [124–132]. The EF/LF-PA63 and to Purkinje cells, parasites, and tumor cells [28, 143]. Ang
PA63 oligomerize on the cell surface to heptamers facili- is a 123 amino acid residue, 14 kDa Mr protein that spe-
tated by the new N-terminal domain 1 and domain 2 cifically degrades tRNA. It binds 170 kDa and 42 kDa Mr
(residues 259–487) of PA63 [133]. There can be up to receptors on endothelial and vascular smooth muscle cell
seven LF or EF molecules bound per heptamer. Both surfaces and induces cell proliferation, cell adhesion, pro-
monomers and heptamers then undergo endocytosis
[134]. The low pH of the endosomes triggers protonation
of critical histidines and unfolding of a large amphip- His119
R
athic hairpin loop in PA domain 2 which is hypothesized His12
HN NH
to insert into the membrane and, together with those of O
O
other PA molecules in the heptamer, form a 14-strand P
O
β-barrel [135]. EF or LF then unfolds and translocates to N NH
the cytosol through the barrel pore [136, 137]. Once in O O H
the cytosol, EF and LF refold. EF is a calcium-depen-
dent-calmodulin-dependent adenylate cyclase that raises
the cytosolic cyclic AMP (cAMP). Elevated cAMP leads B
O
to paralysis of several macrophage physiological path- His12
ways [138]. LF proteolytically cleaves members of the His119
H
mitogen-activated protein kinase kinase (MAPKK or NH
MEK) family. In the case of MEKs I and 2, this cleavage HN N H O O O HN
P
results in their inactivation. Consequently, they cannot O O
activate their substrate, MAPK. Depending on the cell
type, this may have a variety of consequences including O B
apoptosis or altered physiology [139].
Figure 6. Steps in RNase hydrolysis. This is a two-step pro-
cess in which initially a cyclic phosphate intermediate is formed.
Type I Vertebrate A Toxins His-12 acts as a general-base catalyst and His-119 acts as a
general acid to protonate the leaving group (the RNA fragment
Several members of the vertebrate ribonuclease (RNase) designated R). The second step is a hydrolysis step in which
superfamily have been linked to tumor selective ligands. His-119 activates the attack of water by general base catalysis
These ribonucleic acid-degrading proteins are small and His-12 is the acid catalyst, protonating the leaving group
Arthur E. Frankel et al. 415

duction of cell-associated proteases, and cell migration Type II Toxins


and invasion. The protein’s ribonucleolytic activity is
essential in these activities. The protein is internalized by Type II toxins interact with cell surfaces and produce
receptor-mediated endocytosis, translocated to the cytosol changes leading to secondary intracellular signals and
and then the nucleus [29, 144]. Ranpirnase is a basic often cell death. Clostridia perfringens α-toxin is a 370
14 kDa Mr protein which binds tumor cells and vascular residue, zinc metalloenzyme with phospholipase C
endothelium and degrades tRNA selectively [32, 145]. activity [149]. It is the key virulence determinant in gas
Unlike ang, ranpirnase triggers cell apoptosis. Neither gangrene. It binds to membranes in the presence of cal-
BS-RNase nor ranpirnase is bound by cytosolic ribonu- cium. It has two domains (Fig. 1f). The N-terminal
clease inhibitor protein (RI), and, since they are not inac- domain (residues 1–246) is composed of six stacked
tivated in the cell, have potent cell toxicity. RI constitutes a-helices. It has the phospholipase C active site. There is
>0.01% of cytosolic proteins and inactivates ribonu- a flexible linker (residues 247–255) followed by a
cleases by forming tight complexes that prevent RNA C-terminal domain (residues 256–370) with an eight
substrates from entering the active site. Human pancre- (three-strand jelly roll topology). The C-terminal domain
atic RNase 1 and bovine pancreatic RNase A are not toxic resembles eukaryotic C2 domains and has Ca Z′-dependent
to cells, lack cell-binding functions, and are good sub- phospholipid binding function. Membrane phospholipid
strates for RI. Interestingly, a variant of RNasel – des. hydrolysis results in the perturbation of cell metabolism
1-7hpRNase 1 – is not a substrate for RI and is 100-fold and activation of the arachidonic acid cascade and
more cytotoxic when delivered to cells by conjugation to protein kinase C. Cell death and altered physiology such
epidermal growth factor (EGF) [30]. The nature of the as endothelial cell integrin and cytokine expression and
cell-binding and internalization functions of BS-RNase, platelet aggregation ensues.
EDN, ang, and ranpirnase are unknown.
Type III Toxins
Type I Fungal A Toxins Type III toxins produce pores in cell membranes leading
The fungal cytotoxins include α-sarcin, mitogillin, clavin, to cell death. Sticholysin II from the tentacle nemato-
and restrictocin. These are 17 kDa Mr, basic proteins with 20 cysts of the sea anemone Stichodactpla helianthus is a
lysines, four arginines, and eight histidines. α-Sarcin, restric- member of the actinoporin family. It is a 175 amino acid,
tocin, clavin, and mitogillin share 86–99% amino acid 19 kDa Mr basic protein containing five tryptophans, 12
homology [94]. They have four cysteines forming two disul- tyrosines, and no cysteines [150]. This water-soluble
fide bridges between residues 6 and 148 and residues 76 and protein binds membrane lipid (principally sphingomy-
132. The first disulfide bond is not essential for the catalytic elin) and partially unfolds [151]. The protein’s β-sheets
activity. The proteins show structural homology to RNase facilitate tetramer formation [152]. This is followed by
T1 and have catalytic histidines similar to RNase T1 and membrane insertion of the N-terminal amphipathic helices.
other RNases (Fig. 1f). There is a three-turn α-helix packed One nanometer pores are produced, which causes colloid
against a five-stranded antiparallel β-sheet [146, 147]. Large osmotic shock and cell death.
positively charged peripheral loops near the active site con- Cecropins are peptides found in the hemolymph of the
struct a platform with a concave surface for RNA binding. A giant silk moth Hpalophora cecropia [153]. The synthetic
large 39-residue loop L3 may contribute to membrane bind- cecropin analog, Shiva-1, is a 36 residue basic peptide
ing. They have acquired a mechanism to enter cells – inter- devoid of cysteine with a strongly basic N-terminal
acting with negatively charged phospholipids in membranes amphipathic helix linked via a flexible Gly-Pro linker to
and translocating across membranes. Further, in the cytosol, a neutral C-terminal amphipathic helix (Fig. 1g) [154].
they cleave a single phosphodiester bond (3′ to guanosine The peptide is toxic to bacteria, parasites, and tumor
residue 4325) in a highly conserved stem-loop structure of cells. The peptide binds cell surface acidic lipids and
28S ribosomal RNA. This target is two bases removed from forms channels in the membranes followed by cell lysis.
the target for depurination by the plant rRNA N-glycosidases Pyrularia thionin (PT), from the parasitic plant
[148]. This stemloop rRNA structure is a binding site for Pyrularia pubera, is a 47 amino acid basic peptide with
elongation factors, and its modification irreversibly blocks five lysines, four arginines, and eight cysteines (which
protein synthesis (Fig. 3). These toxins share antigenicity form four disulfide bonds) [155]. The molecule consists
with Aspergillus fumigatus allergen I, and produce hyper- of two amphipathic α-helices connected to two anti-
sensitivity reactions in patients. These fungal toxins are parallel β-sheets (Fig. 1h). PT has a hydrophobic and
cytotoxic to tumor cells [33–35, 93]. hydrophilic surface. It binds to cell membrane phos-
416 Immunotoxins

phatidylserine with its hydrophilic side and interacts with approached in a similar way to the plant toxins. Trypsin
phospholipid hydrocarbon tails through the hydrophobic cleavage followed by reduction and purification have been
domain. This destabilizes the membrane bilayer with used to isolate DT A fragment [161]. DT and PE mutants
formation of inverted micelles [156]. The altered with altered amino acid residues important in receptor
membrane organization can lead to cell death. The mem- binding have been used [24, 162]. The most common
brane depolarization also produces an influx of calcium approach for the bacterial A-B-toxins has been to geneti-
and induction of phospholipaseA2 [157]. cally delete the receptor binding domain. This has been
Proaerolysin from Aeromonas hydrophila is a 53 kDa accomplished successfully for DT with the generation of
Mr protein. It exists as a dimer and binds glycophos- DT385, DT388, DAB389, DT390 and DT486 and for PE
phatidylinositol-anchored proteins found on the surface with synthesis of PE35, PE38, and PE40 [163, 164].
of most mammalian cells. It contains a C-terminal inhib- The type I binary toxin, anthrax toxin, has been modi-
itory domain that is cleaved by membrane proteases such fied both by removing critical receptor-binding residues
as furin. The N-terminal fragment, aerolysin, oligomerizes of PA domain 4 and by changing the protease cleavage
and enters plasma membranes to form highly stable pores sequence in PA domain 1 so that the toxin is processed
causing cell death [549]. only on tumor cells [165, 166]. Further, the anthrax
toxin lethal factor has been genetically modified to
attach the PE catalytic domain to the lethal factor PA
Toxin Modification and Ligand binding domain to produce FP59 [550].
Conjugation Addition of sphingomyelin to type III actinoporin
To prepare immunotoxins, three tasks must be com- conjugates, including sticholysin and equinatoxin II,
pleted. The toxin normal tissue binding and toxicity blocks normal tissue binding [151, 234]. The type III
functions must be removed or modified; target cell toxin, proaerolysin, has been modified to replace the
selective peptide ligands must be identified; and the furin site with the prostate cancer specific cleavage pro-
modified toxin and ligand must be covalently linked in tease, PSA, cleavage sequence [548, 549].
such a manner that there are no alterations to the toxin The type I A toxins lack normal tissue-binding func-
translocation and enzymatic functions or the ligand- tion and hence do not require structural modification for
binding or internalization functions. An alternative reducing toxicities.
approach can be used to deliver peptide toxins to cells.
Bispecific monoclonal antibodies that have one binding Ligand Selection
domain for target cell surfaces and the other binding
domain to react with peptide toxins have been synthe- The peptide ligand must bind all the target cells and, in
sized. These molecules have shown potent antitumor the case of type I toxins, undergo internalization. In addi-
activity in vitro and in animal models [480]. Because of tion, the ligand should have minimal or no binding to
space limitations we have focused our review only on vital normal tissues. The identification of selective
covalently linked ligandtoxin conjugates. ligands for immunotoxin synthesis is one of the most
important steps. Table 2 shows the variety of ligand-
receptor systems which have been used for immunotoxin
Toxin Modification targeting. Antibodies, antibody fragments, cytokines,
The type I A-B, type I binary toxin, type II, and type III growth factors, antigens, and receptor fragments have
toxins may react with normal vital tissues and, conse- been linked to toxins. Only a subset of the synthesized
quently, produce serious side-effects. The structure/ immunotoxins have shown selective cytotoxicity to target
function studies described above suggest solutions for cells. Problems with constructs have included steric
the type I and one of the type III toxins. hindrance or misfolding of the ligand, insufficient
The lectin subunit of the plant type I A-B toxins may internalization for type I toxin conjugates, and unex-
be removed either by reduction and purification of the A pected normal tissue reactivities. Ligands which make
subunit [8] or production of the A subunit by heterolo- inactive conjugates with one toxin may be highly effica-
gous expression [158]. A third approach is to use affinity cious when either (a) combined with another toxin, (b)
crosslinkers to block the B subunit lectin sites [159]. conjugated or fused with a different linker, or (c)
Genetic engineering has been used to produce B subunit expressed using a different vector or host. These will be
variants with modifications of amino acid residues at the discussed in greater detail in the next section. Recently,
lectin sites [160]. bispecific immunotoxins have been made by Vallera
The normal tissue receptor-binding domains of the bac- employing two ligands with one toxin (e.g., EGF and
terial type I A-B toxins and the binary toxins have been IL3, sFv anti-ErbB2 with sFv anti-EpCAM, IL13 with
Arthur E. Frankel et al. 417

Table 2. Ligands used to prepare immuntoxins


Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
UCHT1F(ab)’2 Fab’2 fragment CD3ε C UCHT1F(ab’)2-RTA T-cell ALL [401]
BisFv Bivalent sFv CD3ε G A-dmDT390-bisFv T-cell ALL [168]
SFv-Cys SFv-Cys mCD3ε G (DT390-sFv-Cys)2 T-cell ALL [169]
sFv sFv mCD3ε G DT390anti-CD3sFv T-cell ALL [170]
SPV-T3a MAb mCD3 C SPV-T3a-RTA T-cell ALL [411]
UCHT1 MAb CD3ε C UCHT1-CRM9, UCHT1-DTA, T-cell ALL [171,
UCHT1-MSPSA, UCHT1-DT, 172, 312]
UCHT1-ricin
WT32 MAb CD3 C WT32-RTA T-cell ALL [329]
64.1 MAb CD3 C 64.1-RTA T-cell ALL [420]
F(ab’)2 sheep Polyclonal CD3-anti-CD3 C F(ab )2-dianthin, F(ab’)2-saporin, T-cell ALL [20]
anti-mouse Ig F(ab )2-PAP, F(ab’)2-bryodin,
F(ab )2-momordin, F(ab’)2-
momorcochin, F(ab’)2-trichokirin
FN18 MAb rCD3 C FN18-CRM9, DT390-C207 T-cell ALL [173,
diabody 613]
898112-6-15 MAb pCD3 C pCD3-CRM9 T-cell ALL [24]
HB-EGF Growth factor EGFR G HBEGF-saporin Lung cancer [437]
EGF Growth factor EGFR G, C DAB339EGF, DAB486EGF, Lung cancer [30, 175,
EGF-DTA, EGF-RTA, EGF- 176, 477,
hpRNasel, EGF-ang, SAP-EGF 600]
EGF Growth factor EGFR, IL13R G DTEGF13 Prostate cancer, [551,
glioma 556]
TGFα Growth factor EGFR G TGFα-PE40, PEα53L/TGFα/KDEL Pancreas cancer [264,
295]
Mints MAb EGFR C Mint5-ocymoidine, Mint5- Breast cancer [23]
pyramidatine
425.3 MAb EGFR C 425.3-PE Breast cancer [283]
425 sFv EGFR G 425(scFv)-ETA’ Pancreas cancer [584]
528 MAb EGFR C 528-RTA Lung cancer [425]
r3 Mab EGFR C egf/r3Mab-HT Lung [557]
adenocarcinoma
B4G7 MAb EGFR C B4G7-gelonin Lung cancer [472]
14171(dsFv) dsFv EGFRvIII G sFv(14E1)-ETA Gliomas [291]
14171(Fv) sFv EGFRvIII G dsFv(14E1)-ETA Gliomas [291]
Anti- sFv EGFRvIII G Anti-EGFRvIII(Fv)-PE40 Gliomas [262]
EGFRvIII(Fv)
MR1-1 dsFv mEGFRvIII G MR1-1(dsFv)PE38KDEL Gliomas [608]
L2 Growth factor IL2R G DAB389IL2, DAB486IL2, Lymphomas [160, 162,
IL2-PE66GIu4, IL2-Bax, 167, 177,
IL2-ricin, IL2-PE40, IL2-PAP, 178, 413,
IL2-pancRNase1 476]
Mik-betal(Fv) sFv UR G Mik-beta1(Fv)-PE40 Lymphomas [253]
Bell 0 MAb IL2R C BB10-saporin Lymphomas [447]
RFT5 MAb IL2R C RFT5-RTA Lymphomas [382]
Anti-Tac MAb IL2R C Anti-Tac-RTA, Anti-CD25-MLA Lymphomas [314,
413, 10]
Anti-Tac(Fab) Fab IL2R G AntiTac(Fab)-PE40, Anti- Lymphomas [36, 260]
Tac(Fab)-phospholipase C
Anti-Tac(Fv) sFv IL2R G Anti-Tac(Fv)-PE40, Anti- Lymphomas [260,
Tac(Fv)-PE38 279]
RTF5(Fv) sFv IL2R G RFT5(Fv)-ETA’ Lymphomas [285]
GMCSF Growth factor GMCSFR, uPAR G DTU2GMCSF AML [555]
GMCSF Cytokine GMCSFR G DT388GMCSF, DT385-L-GMCSF, AML [179, 180,
GMCSF-PE40, GMCSF-ricin 181]
mGMCSF Cytokine mGMCSFR G DT390mGMCSF, DT388mGMCSF AML [182, 183]
IL3 Cytokine IL3R G DT388IL3, DT388K116W AML [184, 558]
(continued)
418 Immunotoxins

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
26292 sFv CD123 G 26292(Fv)-PE38-KDEL AML [605]
mIL3 CytoKne mIL3R G DT390mIL3, DT389 L-mIL3 AML [185, 186]
Transferrin Growth factor Transferrin C Tf-CRM9, Tf-CRM107, Gliomas, [187, 188,
receptor/CD71 Tf-equinatoxin II, Tf-PE, T-mellALL 234, 259,
Tf-Saporin, Tf-gelonin, 402, 412,
Tf-RNase, Tf-KFT25-RTA 421, 426]
anti-TfR(Fv) sFv Transferrin G DT388-anti-TfR(Fv),anti- AML [28, 29,
receptor/CD71 TfR(Fv)-PE40, anti-TfR(Fv)-EDN, 34, 179,
Anti-TfR(Fv)-pancRNase, 407]
Anti-TfR(Fv)-angL2, Antii-TfR(Fv)
L1, Anti-TfR(Fv) restrictocin
454A12 MAb Transferrin C 454A12-CRM107, 454A12-RTA, Gliomas, breast [188–191]
receptor/CD71 454A12-rRTA, 454A12-RNase cancer
HB21 MAb Transferrin C HB21-PE, HB21-RTA, HB21- Breast cancer [14, 33,
receptor/CD71 gelonin, HB21-bryodin, 191,
HB21-luffin, HB21-α-sarcin 360]
mIL18 Growth factor mIL18R G mIL18-PE38 T cell leukemia [585]
B7 Modulator CD28 G B7-2-L-PE40KDEL GVHD [602]
IP-10 Chemokine CXCR3 G DT390-IP-10-Sralpha Multiple sclerosis [604]
CCL17 Chemokine CCR4 G CCL17-EDN, CCL17-PE38 T lymphoma [606]
HB21 (Fv) sFv Transferrin G HB21(Fv)-PE40 Breast cancer [287]
receptor/CD71
51E9 MAb Transferrin C 5E9-gelonin Breast cancer [414]
receptor/CD71
B2/25 MAb Transferrin C B2/25-saporin Breast cancer [442]
receptor/CD71
OKT9 MAb Transferrin C OKT9-gelonin Breast cancer [459]
receptor/CD71
R17217 MAb Murine transferrin C R17217-rRTA Breast cancer [237]
receptor
Anti-Ly2.1 MAb mTcell antigen C Anti-Ly2.1-ricin T-cell ALL [330]
IL7 Cytokine IL7R G DAB389IL7 Pre-B cell ALL [200]
ASF Lectin Asialogly C ASF-DTA Hepatocarcinoma [201]
coprotein
receptor
IL4 Cytokine IL4R G DAB389IL4, IL-4(38-37)- Gliomas, KS [202, 203,
PE38KDEL, cpIL-4(13D)- 587]
PE38KDEL
mIL4 Cytokine mIL4R G DAB389mIL4, DT390mIL4, AML, [204–206]
mIL4-PE40 mastocytosis
IL13 Cytokine IL 13R G IL 13-PE38QQR, DT388IL 13, Renal cell cancer, [207, 247,
DTEGF13, DTAT13 gliomas 551, 553,
556]
αMSH Growth factor MSHR G DAB389αMSH, DAB486αMSH Melanoma [208, 209]
GRP Hormone GRPR/bb2 G DAB389GRP Adenocarcinoma [210]
NT-4 Growth factor TrkB G DAB389NT4 Neuroblastoma [211]
CD4 T cell HIVgp120 G DAB389CD4, CD4-PE40, HIV + tumors [212, 213,
co-receptor CD4-RTA 337]
3B3(Fv) sFv HIVgp120 G 3B3(Fv)-PE38 HIV + tumors [235]
Anti-gp120 MAb HIVgp120 C Anti-gp120-RTA HIV + tumors [236]
0.5beta MAb HIVgp120 C 0.5beta-RTA HIV + tumors [336]
Anti-gp120 Polyclonal HIVgp120 C Anti-gp120-RTA HIV +tumors [355]
CyanovirinN Antiviral protein HIVgp120 G FLAG-CV-N-PE38 HIV +tumors [274]
7B2 MAb HIVgp41 C Anti-gp41-RTA HIV + tumors [236, 318,
564]
HRG13 Growth factor HER4 G HRG13-PE38KDEL Breast cancer [299]
(continued)
Arthur E. Frankel et al. 419

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
HRGbetal Growth factor HER4 G HRGbeta2-PE38KDEL Breast cancer [299]
HRGbeta2 Growth factor HER4 G HRGbetal-PE38KDEL Breast cancer [299]
Betacellulin Growth factor HER4 G BTC-TX50, BTC-TX48 Breast cancer [300]
48–50
Hrg Growth factor HER4 G DT389hrg, hrg-PE38KDEL, Breast cancer [214, 248,
hrg-PE40 277]
HERB sFv HER2 G hERB-hRNase Breast cancer [582]
FRP5 Mab HER2 G chFRP5-ZZ-PE38 Breast cancer [614]
FRP5(SFv) SFv HER2 G SFv(FRP5)-ETA Prostate cancer [281]
e23(dsFv)2 (dsFv)2 HER2 G e23(dsFv)2-PE38 Lung cancer [290]
e23(dsFv) dsFv HER2 G e23(dsFv)-PE38 Lung cancer [290]
e23(sFv) sFv HER2 G e23(Fv)-PE38KDEL Prostate cancer [292]
520C9 MAb HER2 C 520C9-RTA Breast cancer [316]
741 F8 MAb HER2 C 741 F8-RTA Breast cancer [422]
454C11 MAb HER2 C 454C11-RTA Breast cancer [422]
Mgr6 MAb HER2 C Mgr6-clavin Breast cancer [35]
BACH250 Humanized MAlk HER2 C BACH250-gelonin Breast cancer [11]
Anti-HER2 MAb HER2 C Anti-HER2-saporin Breast cancer [444]
ConA Lectin Mannose C ConA-DTA, ConA-RTA Carcinoma [215, 216]
TRH Hormone TRHR C TRH-CRM45, TRH-CRM26 Pituitary tumors [217]
Ricin Lectin Galactose C Ricin-DTA Carcinoma [218]
sIL 15 Cytokine IL 15R G DAB389IL 15 Lymphoma [219]
D3 MAb p148 C D3-DTA Hepatocarcinoma [220]
GCSF Cytokine GMCSFR G DAB389GCSF, GCSF-PE40 AML [223, 224]
H65 MAb CD5 C H65-RTA, H65-mitogillin, T-cell ALL [93, 394,
H65-gelonin 466]
STI MAb CD5 C STI-RTA T-mellALL [395]
T101 MAb CD5 C Anti-CD5-CRM9, T101-RTA, T-cell ALL [187, 190,
T101-RNase, T101-ricin, 228, 312,
T101-ricin-125l, anti-CD5-MLA, 327, 328,
T101 Fab-RTA, T101 F(ab’)2- 10, 19,
RTA, anti-CD5-momordin 25]
anti-CD5-Pyrularia thionin
OKT1 MAb CD5 C OKT1-sap or in T-cell ALL [451]
SM5-1 sFv gp230 G SMFv-PE38KDELmut1 Hepatocellular [612]
cancer
MEL sFv gp240 G MELsFv-rGel, scFvMEL/TNF Melanoma [569, 574]
ZME01 8 MAb Proteoglycan, C ZME018-RTA, ZME018-gelonin Melanoma [353]
p250
9.2.27 MAb Proteoglycan, C 9.2.27-DTA, 9.2.27-abrin, Melanoma [225, 254,
p250 9.2.27-PE, 9.2 27-gelonin, 307, 311]
9.2.27-RTA, 9.2.27-PAP
NR-ML-05 MAb Proteoglycan, C NR-ML-05-abrin, NR-ML-05-PE Melanoma [254]
p250
SV10016 MAb Proteoglycan, C SV10016-CRM103 Melanoma [187]
p250
Ep2 MAb Proteoglycan, C Ep2-saporin Melanoma [448]
p250
BrE3 MAb Mucin, MUC1 C BrE3-RTA SCLC [375]
Anti-MUC1 sFv MUC1 G sFv(MUC1)-ETA Breast cancer [603]
Mc5 MAb Mucin C Mc5-DTA Breast cancer [226]
BM7 MAb Mucin C BM7-PE Breast cancer [283]
C242 MAb Mucin C C242-PE, C242-NlysPE40, Breast cancer [255, 367]
C242-RTA
C242rF(ab’) Antibody Mucin C C242F(ab’)-PE38QQR Colon cancer [268]
fragment
MBR1 MAb Mumn C MBR1-restrictocin Breast cancer [322]
(continued)
420 Immunotoxins

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
260F9 MAb p55 C 260F9-RTA, 260F9-rRTA Breast cancer [158, 191]
171A MAb Ep-CAM C 171A-DTA, 171A-RTA Breast cancer [227]
Anti-EpCAM sFv EpCAM G DTEpCAM23 Colon cancer [552]
4D5MoCB sFv EpCAM G 4D5MoCB-ETA Colon cancer [568]
A9(Fab)’ Fab fragment p100 C A9(Fab)’-DTA A.castellani [229]
Anti-SV40 Polyclonal SV40 antigens C Anti-SV40-DT SV40 + tumors [230]
Cholera toxin B Toxin fragment GM1 ganglioside C Cholera B-DTA Gliomas [231]
Anti-ConA Polyclonal ConA C Anti-ConA-DTA ConA-treated [232]
tumors
RTB Toxin fragment Galactose C RTB-DTA, RTB-MLA, RTB- Carcinomas [233, 387,
momordin 417]
TEClgM MAb IgM Fc C TEClgM-saporin Myeloma [455]
Protein A Toxin fragment ImmunoglobulinFc C Protein A-RTA Ig + tumors [238]
74124 MAb pCD4 C 74124-PAP Transplants [431]
MT151 MAb CD4 C Anti-CD4-PAP, MT151-RTA, HIV + tumors [239–241,
MT151-blocked ricin, 453]
anti-CD4-saporin
OVB3 MAb Ovarian antigen C OVB3-PE Ovarian cancer [242]
NR-LU-10 MAb Breast cancer C NR-LU-10-PE Breast cancer [243]
antigen
ATF Protease Urokinase PA G, C ATF-PE38, ATF-PE38KDEL, AML, gliomas [244–246,
fragment receptor uPA-SAP, DT388N-termURO, 434]
ATF-saporin
11A8 MAh bFGFR C 11 A8-saporin Breast cancer [435]
bFGF Growth factor bFGFR G bFGF-PE40, hFGF-PE66Glu4, Breast cancer [249, 435,
hFGF-saporin, DT389bFGF 599]
aFGF Growth factor aFGFR G aFGF-PE40, aFGF-PE66Glu4KDEL Breast cancer [249, 252]
PR1(Fv) sFv PR1 antigen G PR1(Fv)-PE38KDEL Prostate cancer [250]
MRK16 MAb P-glycoprotein C MRK16-PE, MRK-RTA, Renal cancer [251, 365,
MRK-saporin 450]
PA Toxin peptide PA receptor G FP59 + PA, FP59 + PA(MMP) Gliomas, [256, 286]
carcinomas
IGF-I Growth factor IGF-IR G IGF-I-PE40 Breast cancer [257]
Anti-CD8 MAb CDS C Anti-CD8-ricin, Anti-CDs-saporin Tcell lymphoma [331, 453]
Lym-1 MAb HLA-DR C Lym-l-gelonin Lymphoma [463]
2G5 MAb HLA-DR C 2G5-RTA Lymphoma [361]
HB55 MAb HLA-DR C HB55-ricin Lymphoma [343]
KM231 MAb Sialyl-Lea-antigen C KM231-RTA Colon cancer [356]
84.1C MAb mlgE C 84.1c-RTA Allergies [396]
Phlp56 Allergan Ant-P5 G P5-ETA’ Allergies [593]
B3 MAb Lewisy antigen G B3-LysPE38 Breast cancer [266]
B3(Fab) Fab fragment Lewisy antigen G B3(Fab)-PE38 Breast cancer [297]
B3(dsFv) dsFv Lewisy antigen G B3(dsFv)-PE38, dsFvB3-gran- Breast cancer [297, 580]
zyme B
B3(Fv) sFv Lewisy antigen G B3(Fv)-PE38 Breast cancer [258]
B1(Fv) sFv Lewisy antigen G B1(Fv)-PE38 Breast cancer [267]
B1(dsFv) dsFv) Lewisy antigen G B1(dsFv)-PE38 Breast cancer [267]
BR96(sFv) sFv Lewisy antigen G BR96sFv-PE40 (SGN10), Breast cancer [21, 278]
BR96sFv-bryodin
Folate Vitamin Folate receptor C Folate-momordin, Folate- KS [261]
LysPE38, Folate-Cys-PE35
Anti-FRbeta Mab Folate receptor C anti-FRbeta-PEA RA [592]
IL9 Cytokine IL9R G rhIL9-ETA’ Hodgkin’s disease [263]
55.1(Fv) sFv Colon antigen G 55.1(Fv)PE38, 55.1(Fv)PE38KDEL Colon cancer [265]
55.1 (dsFv) dsFv Colon antigen G 55.1(dsFv)-PE38, 55.1(dsFv)- Colon cancer [265]
PE38KDEL
ME20 MAh p105 C ME20-lysPE40 Melanoma [269]
(continued)
Arthur E. Frankel et al. 421

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
G28- sFv CD40 G G28-5sFv(VL-VH)-PE40, Lymphoma [18, 270]
5sFv(VL-VH) G28-5sFv(VL-VH)-hryodin
GnRH Hormone GnRHR C GnRH-PE66, GnRH-PE40, Breast cancer [271, 293,
GnRH-PAP 13]
MOC31 MAb EGP-2 C MOC31-ETA252-61 3 SCLC [272]
WF lectin Lectin N-acetyl- C WF-DTA Carcinoma [221]
Galactosamine
hPL Hormone hPLR C hPL-DTA Breast cancer [222]
AntiCD30sFv sFv CD30 G Anti-CD30(sFv)-PE38, Anti- Hodgkin’s [280]
CD30(Fv)-PE38KDEL disease
Ki4 MAb CD30 C Ki4-RTA Hodgkin’s [398]
disease
Ki4(sFv) sFv CD30 G Ki(sFv)-ETA’, scFvKit4- Hodgkin’s [294, 565]
angiogenin disease
BerH2 sFv CD30 G BerH2-scFv-hpRNase Hodgkin’s [609]
disease
BerH2 MAb CD30 C BerH2-saporin, BerH2-RTA, Hodgkin’s [22, 373,
BerH2-momordin, BerH2- disease 416, 428]
dianthin, BerH2-PAP
HRS3 MAb CD30 C HRS3-RTA Hodgkin’s [338]
disease
HRS3Fab’ Fab’ fragment CD30 C HRS3Fah’-RTA Hodgkin’s [338]
disease
TP3 Mab p80 C TP3-PAP Osteosarcoma [174]
TP3(sFv) sFv Osteosarcoma G TP-3(sFv)-PE38 Osteosarcoma [26]
antigen
TP3(dsFv) dsFv Osteosarcoma G TP-3(dsFv)-PE38 Osteosarcoma [26]
antigen
RANTES Chemokine CCR5 G RANTES-PE40, DT390-RANTES Rheumatoid [282, 561]
arthritis
HD6 MAb CD22 C HD6-saporin Lymphomas [452]
HD39 MAb CD22 C HD39-saporin Lymphomas [452]
OM124 MAb CD22 C OM124-PAP, OM124-saporin Lymphomas [348]
RFB4 MAb CD22 C RFB4-RTA Lymphomas [332]
RFB4(dsFv) dsFv CD22 G RFB4(dsFv)PE38, RFB4(dsFv) Lymphomas [288, 562]
PE38KDEL, HA22
LL2 MAb CD22 C LL2-onconase Lymphomas [32]
RFB4 sFv CD22 G DT2219ARL B-cell ALL, CLL [554]
AchRα Antigen Anti-AchR G AchRα-gelonin Myasthenia [594]
gravis
Dsg3ΔNI Antigenic Anti-desmoglen3 G DsgΔN1-PE40KDEL, PE37- Pemphigus [289]
peptide Dsg3ΔN1-KDEL vulgaris
hMN14sFv sFv CEA G hMN14(Fv)-PE40 Colon cancer [296]
C110 MAb CEA C C110-RTA-s°Y Colon cancer [357]
C19 MAb CEA C C19-RTA Colon cancer [304]
CB-CEA-1 MAb CEA C CB-CEA-1-hemolytic toxin Colon cancer [37]
I-1 MAb CEA C I-1-blocked ricin Colon cancer [391]
C27 MAb CEA C C27-ahrin A Colon cancer [9]
E4 MAb Prostate antigen C E4-PE35KDEL Prostate cancer [298]
E4(Fv) sFv Prostate antigen G E4Fv-PE38KDEL Prostate cancer [298]
M6 MAb Idiotype C M6-ricin, M6-RTA Lymphoma [302]
38.13 MAb Pk antigen C 38.13-RTA, 38.13-gelonin, Burkitt’s [306]
38.13-PAP lymphoma
Fab’anti-L3T4 Fab’ fragment Murine C Fab’anti-L3T4-RTA Lymphoma [308]
Tcellantigen
486P3121 MAb Bladder cancer C 486P-RTA + 486P-RTB Bladder cancer [310]
antigen
RFT11 MAb CD2 C RFT11-RTA T-cell ALL [326]
(continued)
422 Immunotoxins

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
35.1 MAb CD2 C 35.1-ricin, 35.1-RTA T-cell ALL [312, 326]
OKT11 MAb CD2 C Anti-CD2-RTA, Anti-CD2-saporin, T-cell ALL [393, 473]
OKT11-gelonin
452D9 MAh p74 C 452D9-RTA Ha-ras + tumors [313]
Thyroglobulin Antigen Ig anti- C Thyroglobulin-RTA Thyroiditis [305]
thyroglobulin
Anti-CD64 Mab CD64 C Anti-CD64-RTA Rheumatoid [598]
arthritis
H2-D(d)tet Tetramer MHC C H2-D(d)tetramer-biotin-avidin- GVHD [601]
saporin
2F8 Mab SR-A/CD163 C 2F8-Saporin Ovarian cancer [610]
E9 sFv FCRL1 G E9(Fv)-PE38 CLL [615]
96.5 MAb p97 C 96.5-RTA Melanoma [424]
HB2 MAb CD7 C HB2-saporin T-cell ALL [434]
TXU MAb CD7 C TXU-PAP T-cell ALL [390]
3A1e(Fv) Fv-Cys CD7 C 3A1e(Fv)Cys-RTA T-cell ALL [408]
3A1f sFv CD7 G Rnase-TRHRQPRGWEQL-anti- T-cell ALL [588]
CD7-sFv
Anti-CD7 sFv CD7 G scFvCD7:sTRAIL T-cell lymphoma [590]
3A1 e MAb CD7 C 3A1e-RTA T-cell ALL [427]
WTI MAb CD7 C WT1-RTA T-cell ALL [317]
791T/36 MAb p72 C 791T/36-RTA Colon cancer [319]
8A MAb Myeloma antigen C 8A-momordin, 8A-saporin Myeloma [320, 321]
IORT6 MAb T-cell antigen C IORT6-hemolytic toxin T-cell ALL [323]
SN5d MAb CD10 C SN5d-RTA Pre-Bcell ALL [324]
Anti-CALLA MAb CD10 C Anti-CALLA-RTA Pre-B cell ALL [380]
AntiGE2 MAb GE2 C Anti-GE2-ricin, AntiGE2-RTA Gliomas [325]
AR3 MAb CAR-3 C AR3-RTA, AR3-ricin, AR3-gelonin Gastric cancer [333, 461]
8C MAb Ovarian cancer C 8C-RTA Ovarian cancer [334]
antigen
My9 MAb CD33 C My9-blocked ricin AML [346]
p67.7 MAb CD33 C p67.7-RTA AML [352]
HuM195 Humanized CD33 C HuM195-gelonin AML [460]
MAlk
Anti-CD33 sFv CD33 G Anti-CD33(sFv)-ETA AML [597]
Anti- MAb Vasopressin C Anti-vasopressin-RTA Pituitary tumor [347]
vasopressin
Hepama-1 MAb Hepatoma antigen C Hepama-l-trichosanthin Hepatoma [349]
Cluster 2 MAb MAb Cluster 2 C Cluster 2 MAb-RTA SOLO [350]
antigen-SCLC
SOKTI MAb T-cell antigen C SOKT1-RTA T-cell antigen [351]
Fib75 MAb Bladder cancer C Fih75-RTA, Fih75-a-sarmn, Bladder cancer [354, 471]
antigen Fih75-gelonin
MAb2 MAb Gastric antigen C MAh2-RTA Gastric cancer [358]
16 MAb Oncofetal antigen C 16-RTA, 16-MLA Lymphoma [359, 374]
Anti-laryngeal MAb Laryngeal cancer C Anti-laryngeal cancer-RTA Laryngeal cancer [362]
cancer antigen
W3 MAb ICAM C W3-RTA Myeloma [363]
Bispecific Bispecific MAlk CD4 and CD26 C Bispecific (CD4/CD26)-blocked Tissue allografts [364]
MAb(CD4/ ricin
CD26)
SWAII MAb CD24 C SWAII-RTA Lymphoma [366]
HAE9 MAb Erythroblast C HAE9-RTA Eyrtholeukemia [368]
antigen
HAE3 MAb Glycophorin C HAE3-RTA Erythroleukemia [368]
(continued)
Arthur E. Frankel et al. 423

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
Bispecific Bispecific MAlk CD4 and CD29 C Bispecific (CD4/CD29)-blocked Tissue allografts [369]
MAb(CD4/ ricin
CD29)
CLL2m MAb CLLantigen C CLL2m-RTA CLL [370]
SEN31 MAb Cluster 5a SOLO C SEN31-blocked ricin SCLC [371]
antigen
317G5 MAb p42 C 317G5-RTA Breast cancer [372]
SEN36 MAb NCAWCD56 C SEN36-RTA SCLC [376]
SEN7 MAb NCAWCD56 C SEN7-PE, SEN7-blocked ricin SCLC [383]
N901 MAb NCAWCD56 C N901-blocked ricin SCLC [400]
BDI-1 MAb Bladder antigen C BDI-1-ricin Bladder cancer [377]
Anti-mu MAb IgM Fc C Anti-mu-RTA Myeloma [378]
Anti-CD6 MAb CD6 C Anti-CD6-blocked ricin GVHD [379]
Anti-CRF MAb Corticotropin- C Anti-CRF-RTA Pituitary tumor [381]
releasing factor
CRF Hormone CRFreceptor C CRF-saporin Pituitary tumor [436]
ScFvH17 sFv Placental alkaline C scFvH17-BSRNase Germ cell tumor [27, 384]
phosphatase
Anti-la MAb la C Anti-la-RTA Carcinoma [385]
Ng76 Mab Melanoma antigen C Ng76-luffin, Ng76-moschatin Melanoma [571, 572]
E6 Mab PSMA C E6-dgA Prostate cancer [583]
A5 sFv PSMA G A5-PE40 Prostate cancer [595]
7E4B11 Mab RPTPβ C 7E4B11-saporin Gliomas [589]
IB4 Mab CD38 C IB4-saporin-S6 Myeloma [596]
HB7 MAb CD38 C HB7-blocked ricin Myeìoma [386]
Anti- MAb Asialo-GM2 C AntiasialoGM2-RTA Carcinoma [388]
asialoGM2
Anti-vbeta6 MAb vbeta6 T cell C Antivbeta6-RTA Myasthenia [389]
receptor gravis
14G2a MAb Disialoganglioside C 14G2a-RTA Neumblastomm [392]
Peanut Lectin D-glucosyl moiety C Peanut agglutinin-RTA Lymphoma [397]
agglutinin
BU12 MAb CD19 C BU12-saporin Lymphoma [441]
B43 MAb CD19 C B43-PAP Lymphoma [432]
HD37 MAb CD19 C HD37-RTA, HD37-saporin Lymphoma [399, 452]
B4 MAb CD19 C B4-blocked ricin Lymphoma [415]
B-c3 Mab CD19 C Anti-CD19:K CLL [563]
Anti-CD19 sFv CD19 G CD19-ETA’ CLL [611]
Anti-CD19 sFv CD19 G DT2219ARL ALL [554]
Fv5191/cys sFv-Cys CD19 C Fvs191-Cys-RTA Lymphoma [406]
Rituximab Mab CD20 C Rituximab/Saporin-S6, Lymphoma [581, 586]
Rituximab-allinase
ONSM21(Fv) sFv Medulloblastoma C ONSM21 (Fv)-RTA Medulloblastoma [419]
antigen
ONSM21 MAb Medulloblastoma C ONSM21-RTA Medulloblastoma [403]
antigen
35 MAb Nicotinic C 35-ricin Strabismus [404]
acetylcholine
receptor
K42C10 MAb Endoglin/CD105 C K42C10-RTA Breast cancer [405]
44G4 Mab CD105 C 44G4-ebulin, 44G4-nigrin b Breast cancer [558, 607]
SN6j MAb Endoglin/CD105 C SN6j-RTA Breast cancer [418]
8OG MAb Alphafetoprotein C 8OG-gelonin Hepatoma [471]
15A8 MAb Breast cancer C 15A8-gelonin Breast cancer [467]
antigen
HB5 MAb C3d receptor C HB5-gelonin EBV infection [474]
Anti-Lyt2.2 MAb Lyt2.2 antigen C Anti-Lyt2.2-gel on in T-cell lymphoma [475]
(continued)
424 Immunotoxins

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
SN7 MAb B cell antigen C SN7-RTA B-cell ALL [409]
Anti-VIP MAb Vasoactive C Anti-VIP-RTA Pituitary tumor [410]
intestinal peptide
Anti-CMV MAb mCMV antigen C Anti-mCMV-RTA, Anti-mCMV- CMV infection [411, 462]
gelonin
Anti-CMV Polyclonal Human CMV C Anti-huCMV-gelonin CMV infection [464]
antigens
Anti-d(beta)h MAb Dopamine beta- C Antid(beta)h-saporin Pitirflary tumors [12]
hydroxlase
M24 MAb CD80 C M24-bouganin, M24-gelonin, Hodkgin’s [15]
M24-saporin disease
1G10 MAb CD86 C IG10-bouganin, IG10-gelonin, Hodkgin’s [15, 570]
IG10-saporin, IG10-gelonin disease
LHRH Hormone LHRHR C LHRH-bovineRNase Breast cancer [31]
Anti-Pbs21(Fv) sFv Pbs21 G Anti-Pbs21(Fv)-Shiva-1 Malaria [38]
Anti- MAb Melanoma antigen C Anti-melanoma-BRIP Melanoma [17]
melanoma
48127 MAb gp54 C 48127-PAP, 48127-saporin Bladder cancer [429]
J3109 MAb CD72 C J3109-PAP B-mellALL [430]
0×7 MAb mCD90 C 0 × 7-DT, 0 × 7-saporin T-cell ALL [199, 315]
O × 7F(ab’)2 F(ab )2 mCD90 C O × 7F(ab’)-saporin T-mellALL [315]
fragment
Anti-Thy1.2 MAb mCD90 C Anti-Thy1-trichokirin, Anti-Thy1- T-mellALL [16, 458]
gelonin
Anti-Thy1.2 Polyclonal mCD90 C Anti-Thy1-RTA T-mellALL [342]
mFclgE Fcfragment mFcεRI receptor G Fc(2′-3)-PE40 Allergies [273]
IR162 MAb rIgEFc receptor C IR162-RTA, IR162-ricin Allergies [303, 309]
mSCF Cytokine mc-kit G mSCF-PE40 SCLC, AML [275]
K1 MAb Mesothelin p40 G K1-LysPE38QQR Mesothelioma [284]
GPI-anchored
K1(Fv) sFv Menothelin p40 G Anti-mesothelin(Fv)-PE38 Menothelioma [276]
GPI-anchored
SS1(dsFv) dsFv Mesothelin p40 G SS1(dsFv)-PE38, D-SS1P Mesothelioma [543, 616]
GPI-anchored
Anti-0X40 MAb T cell antigen C Anti-OX40-RTA Multiple sclerosis [423]
MBP(66-88) Antigenic Anti-MBP Ig G MBP(66-88)-PE40 Multiple sclerosis [301]
peptide
MBPp8799 Peptide anti-MBP G, C MBPp87-99-cyt1Aa Multiple sclerosis [559]
CD64 Mab CD64 C CD64-riA Rheumatoid [566]
arthritis
m22 sFv CD64 G m22(scFv)-ETA’ AML [573]
anti-JL1 Mab JL1 C Anti-JL1-gelonin Leukemias [567]
Campath-1 MAb CD52 C Campath-l-saporin Lymphoma [335]
L6 MAb Lung cancer C L6-ricin Lung cancer [339]
antigen
Anti-CTLA4 Mab CTLA-4 C Anti-CTLA4-saporin Tolerance [575]
Anti-CTLA4 sFv CTLA-4 G scFvCTLA4-perforin Tolerance [576]
8H9 dsFv glycoprotein G 8H9(dsFv)-PE38 Osteosarcoma [577]
LL1 Fusion L2-H2 CD74 G 2L-Rap-hLL1-gamma4P Myeloma [591]
Anti-mFcD MAb msIgD C Anti-mFcD-RTA Lymphoma [340]
SWA11 MAb SCLC antigen C SWA11-RTA SCLC [341]
Anti-T.cruzi MAb T. muzi antigen C Anti-T. cruziRTA, anti-T. Chagas disease [344]
cruzi-abrin A
Anti-CD45RO Mab CD45RO C Anti-CD45RO-RTA HIV [578]
BMAC1 MAb CD45 C BMAC1-RTA, MBAC1-MLA Renal transplant [345]
OX1 MAb rCD45 C OX1-RTA, OX1-MLA Renal transplant [345]
(continued)
Arthur E. Frankel et al. 425

Table 2. (continued)
Type
Ligand Type Receptor/antigen linkage Compounds Disease Reference
M20.4 MAb primate NGFR p75 C M20.4-saporin Neuromas [437]
1921g MAb rat NGFR p75 C 192Ig-saporin, Neuromas [433]
192Ig-trichosanthin
OKT10 MAb CD38 C OKT10-saporin Myeloma [434]
Insulin Hormone Insulin receptors C Insulin-saporin Breast cancer [438]
BB2 MAb Myeloma antigen C BB2-saporin Myeloma [439]
BB4 MAb Myeloma antigen C BB4-saporin Myeloma [439]
Anti-epithelial MAb Epithelial antigen C Anti-epithelial Lung cancer [440]
antigen antigen-colocin 1
Anti-Ab1 MAb Idiotype anti-DNA C Anti-Ab1-saporin SLE [443]
Anti-Id MAb Idiotype C Anti-Id-saporin Lymphoma [456]
lymphoma
Anti-CD37 MAb CD37 C Anti-CD37-saporin Lymphoma [445]
ML30 MAb Heat shock protein C ML30-saporin AML [446]
Anti-AML MAb AML-M5 antigen C Anti-AML-saporin AML [449]
8A MAb Myeloma antigen C 8A-saporin Myeloma [454]
62B1 MAb Myeloma antigen C 62B1-saporin Myeloma [454]
TTC Toxin fragment p15 G DAB389TTC Neuromas [192]
SP Hormone Neurokinin-1 G DAB389SP CML, neuromas [193]
IL6 Cytokine IL6R G DAB389IL6, IL6-PE40, Myelomas, KS [194, 195]
IL6-PE66GIu4
VEGF165 Growth factor flk-1, flk-1/KDR G, C DT390VEGF165, KS [196, 197]
VEGF165-DT385
VEGF121 Growth factor flk-1/KDR G, C DT390VEGF121, KS, breast cancer [196–198]
VEGF121-DT385,
VEGF121/rGel

oLH Hormone Ovine LHR C oLH-gelonin Prostate cancer [457]


gp330 Antigen Anti-gp330 Ig C gp330-gelonin Heyman’s [465]
nephritis
Anti-pichinde MAb, Pichinde virus C Anti-Pichinde virus-gelonin Pichinde virus [468]
virus polyclonal antigens
NDA4 MAb NDA4 Tcell antigen C NDA4-gelonin T-cell ALL [469]
14G2a MAb GD2 ganglioside C 14G2a-gelonin Melanoma [470]
MSN-1 MAb Endometrial Ca C MSN-1-gelonin Endometrial [547]
antigen cancer

RTA, ricin toxin A chain; sFv, single chain antibody; Cys, cysteine; DT, diphtheria toxin; DTA, diphtheria toxin A fragment; CRM9, binding
site DT mutant; HBEGF, heparin-binding epidermal growth factor; ETA, Pseudomonas exotoxin; PE, Pseudomonas exotoxin; PE40,
40 kDa PE fragment; GMCSF, granulocyte-macrophage colony-stimulating factor; IL3, interleukin-3; CRM107, binding site DT mutant;
rRTA, recombinant RTA; mIL3, mouse IL3; mGMCSF, mouse GMCSF; EGF, epidermal growth factor; TGFα, tumor growth factor-alpha;
dsFv, disuffide-stabilized sFv; MAb, monoclonal antibody; R, receptor; G, genetic fusion; C, chemical linkage; RNase, ribonuclease;
AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; CA, cancer; IL7, interleukin-7; IL4, interleukin-4; mIL4, murine IL4;
IL13, interleukin-13; aMSH, alpha-melanocyte-stimulating hormone; NT-4, neurotrophin-4; GRP, gastrin-releasing peptide; HRG,
heregulin; TRH, thyroid releasing hormone; hPL, human prolactin; Wf, Wistaria floribunda; ConA, concanavalin A; HIV, human immu-
nodeficiency virus; SCLC, small cell lung cancer; IL15, interleukin-15; Ep-CAM, epithelial cell adhesion molecule; RTB, ricin toxin B
chain; MLA, mistletoe lectin A chain; KS, Kaposi’s sarcoma; LH, luteinizing hormone; VEGF, vascular endothelial growth factor; CML,
chronic myeloid leukemia; IL6, interleukin-6; SLE, systemic lupus erythematosis; Id, idiotype; SP, substance P; TTC, tetanus toxin
C-terminal domain fragment; T. cruzi, trypanosoma cruzi; SCF, stem cell factor; Gel, gelonin; PAP, pokeweed antiviral protein; Ig, immu-
noglobulin; NGF, nerve growth factor.MBP, myelin basic protein; VIP, vasoactive intestinal peptide; CMV, cytomegalovirus; LHRH,
luteinizing hormone releasing hormone; CRF, corticotropin releasing factor; GVHD, graft-versus host disease; CLL, chronic ymphocytic
leukemia; GnRH, gonadotropin-releasing hormone; IL9, interleukin-9; Tf, transferrin; GCSF, granulocyte colony-stimulating facor; ASF,
asialofetuin; IGF, insulin-like growth factor; FGF, fibroblast growth factor; a, acidic; b, basic; o, ovine
426 Immunotoxins

the N-terminal fragment of urokinase, sFv anti-CD19 to internalize is also a critical parameter, at least for
with sFv anti-CD22) [551–554]. Dual specific immuno- conjugates with type I toxins [329]. While chemical
toxins have also been tested by Frankel (urokinase conjugates represent the vast majority of reported immu-
activated DT with GMCSF ligand) [555]. notoxins, recombinant toxin conjugates offer advantages
for production [164]. The molecules are homogeneous
Conjugation of Toxin and Ligand with a single site of linkage between the haptophore and
toxophore. Furthermore, the synthesis does not require
The linkage of the modified toxin with the ligand must be manipulations of different chemical components. The
covalent and stable, so that metabolism in the bloodstream specificity of the immunotoxin is important, since
or interstitial tissues will not occur. Both amide linkage crossreaction with vital normal tissues can produce
by genetic engineering and chemical crosslinking using severe toxicities (see the next section). Both normal
bifunctional reagents such as thiolating compounds – human tissue immunostaining and testing in nonhuman
3-(2-pyridyldithio)proprionicacid N-hydroxysuccinimide primate models have been used to identify normal
ester (SPDP) [8] or 3-maleimidobenzoic acid N-hydroxy- tissues likely to be damaged by systemic immunotoxin
succinimide ester (MBS) [24] – have been used. The administration [478, 479]. The size of the immunotoxin
coupling must not severely alter the ligand affinity for appears to influence both efficacy and safety. Larger
its receptor or impair the ability of the toxin domains to conjugates have greater difficulty in reaching extravas-
translocate to the cytosol and enzymatically inactivate cular sites of disease in most cases [228]. In one report,
cell functions (type I toxins) or interact and alter cell a larger immunotoxin targeting HER2 was more effective
membrane functions (type II and III cytotoxins). Genetic in vivo [614]. The larger molecules have prolonged
fusions are well defined and controlled, and generally circulating half-life, leading to increased vascular
have less effect on toxin functions than chemical conju- endothelial injury and vascular leak syndrome (VLS)
gation. Further, the fusion can be designed so that the [8]. However, with conjugates much smaller than 58 kDa
linkage is remote from ligand and toxin domains critical Mr, the immunotoxin will be rapidly cleared by the
for their normal functions. Chemical conjugation meth- kidneys, leading to very short half-life and insufficient
ods yield a heterogeneous product due to the reaction of antitumor effect. Further, renal tubular injury is more
the derivatizing reagent with multiple amino acid resi- likely [169]. Immunotoxins made with human ligands
dues in the ligand and/or toxin. Cysteines and lysines and toxins are likely to be far less immunogenic than
are the most frequent substrates for bifunctional those made with non-human components [28, 29, 32,
reagents. Both chemical conjugates and genetic fusions 190, 407, 426]. Some toxins perform better for particular
have been developed and used on patients with benefi- classes of tumors or target cells. DT conjugates are more
cial results. Successful clinical development has been toxic to myeloid malignancies than PE, ricin, or other
done with both chemical conjugates and genetic type I conjugates. This is likely due to the requirement
fusions. for routing to the endoplasmic reticulum for the type I
toxins other than DT and anthrax toxins. Since most
myeloid cells rapidly traffic internalized materials to the
Preclinical Studies with lysosome for degradation, conjugates requiring trans-
port to the endoplasmic reticulum do not intoxicate
Immunotoxins these cells well [179, 181]. Consequently, improved
Over a 1,000 immunotoxins have been synthesized in efficacy has been observed with DT conjugates for
the past 3 decades, but most have been evaluated with myeloid disorders. Stability in the bloodstream is also
markedly different tissue culture and animal model important. Disulfide-stabilized single chain immuno-
assays. Thus, comparisons are fraught with potential toxins or tandem or dimeric single chain immunotoxins
errors. Nevertheless, in the spirit of a review, we will appear to be more stable in vivo than single-chain Fv
attempt to draw some general observations. fragment conjugates [163, 168]. These molecules also
The more potent immunotoxins show higher affinity have a better in vivo therapeutic index.
binding to their cell surface receptors and have more None of the immunotoxins which have been tested in
receptors/cell [189]. Immunotoxin efficacy is also influ- clinical trials to date is perfect. The ligands are generally not
enced by the location of interaction of the ligand and the tumor-specific. Many of the conjugates are not recombinant.
receptor. Antibodies which bind different epitopes on The protein components are frequently immunogenic.
receptors can vary greatly in conjugate cytotoxicity The size is often too large. In some cases the target cell
[326, 422]. The ability of the ligand-receptor complex may not always endocytose the immunotoxin-receptor
Arthur E. Frankel et al. 427

complex. The patients have often not been optimally transaminasemia, fatigue, edema. Thus, BL22 immuno-
selected for the individual agent. However, many of the toxin appears to be the current best salvage treatment
newly described immunotoxins which have entered clin- for relapsed HCL patients. An improved higher affinity
ical trials in the past few years, or will enter clinical trials variant, HA22, has begun clinical testing in patients
in the near future, have improved characteristics, and with refractory B-cell malignancies [562].
better patient selection is being used. The recent results LMB2 is composed of an anti-CD25 sFv fused to
in the clinic are very exciting and will be described in PE38. LMB2 has been administered to 35 patients with
detail in the next section. lymphomas at doses of 2–63 μg/kg i.v. over 30 min q.o.d.
× 3 [279]. A complete remission was observed in HCL
which was ongoing at 20 months. There were seven partial
Clinical Experience remissions in patients with HCL (three patients),
CTCL (one patient), CLL (one patient), HD (one patient),
with Immunotoxins and ATL (one patient). Responders received at least
Clinical trials have been conducted with only several 60 μg/kg total dose of LMB-2 per cycle. The durations of
dozen immunotoxins over the past 3 decades. The com- remissions have not been determined, but exceed 20 and
plexities in synthesis and purification of these multi- 6 months for two patients and 1 month for the remaining
domain polypeptides, and the necessary but extensive patients. Responding patients had clearance of circulat-
safety and regulatory hurdles, have limited the rapid ing malignant cells, improvement in skin lesions, and
application of this technology. Direct comparisons are regression of lymph nodes and splenomegaly. DLT was
difficult due to the different conjugate constructions; the reversible transaminase elevations, diarrhea and cardio-
different patient populations; and the different routes, myopathy. MTD was 40 μg/kg/dose. Other side effect
doses, and schedules of drug used in the various trials. was fever. 17% of patients developed neutralizing anti-
Nevertheless, we will address the efficacy, pharmacoki- bodies after the first cycle. Drug half-life was 4 h.
netics, immune responses, and toxicities for each agent Responders received at least 60 μg/kg total dose of
and try to present common principles. Excitingly, sev- LMB-2 per cycle. LMB-2 was also administered with
eral of these drugs are showing significant anticancer MART-1/gp-100 vaccine to patients with metastatic mel-
activity in phase I and II clinical studies in patients with anoma in an effort to augment anti-melanoma immunity
chemotherapy-refractory cancers. via Treg depletion [618].
Denileukin diftitox is a fusion protein composed of
the catalytic and translocation domains of DT fused to
Efficacy human IL-2. Among CTCL patients with stage IB to
Immunotoxins with the highest response rate in clinical IVA disease refractory to other therapies and treated
studies have been active in tissue culture in the picomo- with 9 or 18 μg/kg/day for 5 days every 3 weeks, there
lar range, have been targeted to hematologic malignan- were 10% CRs and 20% PRs lasting a median of 6
cies or diseases, and have been fully recombinant months [486, 488, 489]. Denileukin diftitox produced a
molecules. The only exceptions in which good response durable complete remission in a patient with transplant-
rates were seen in non-hematologic diseases were in refractory large cell lymphoma that was ongoing after 5
cases in which immunotoxins were administered locally years [485]. Two patients with low-grade lymphoma
by intracavitary or interstitial infusions. obtained partial remissions. Based on the above results,
BL22 consists of a disulfide-stabilized anti-CD22 sFv in 1999 the FDA made denileukin diftitox the first
fused to PE38. When given at doses of 3–50 μg/kg i.v. approved immunotoxin [539]. Drug half-life was 30 min,
over 30 min every other day for three doses every month and side effects included an acute cytokine reaction
for up to 14 cycles to patients with chemotherapy refrac- (fever, chills, nausea, vomiting, myalgias, chest pain,
tory B cell malignancies, there were 61% CR and 19% arthralgias, back pain), transient liver enzyme transami-
PR in purine-analog refractory hairy cell leukemia nasemia, and a vascular leak syndrome (hypotension,
(HCL) [481, 482, 617]. The remissions were durable hypoalbuminemia, dyspnea, edema). Most patients
with only 27% responders relapsing after 10–23 months. developed an immune response to the agent, but this did
Retreatment again produced CRs. Responses were dose not correlate with toxicities or response. In patients with
dependent and the MTD was 40 μg/kg/dose. Neutralizing non-CTCL relapsed/refractory T-cell non-Hodgkin’s
antibodies developed in 24% of patients. A reversible lymphoma, the agent produced a 48% remission rate
hemolyticuremic syndrome requiring plasmapheresis with 22% CRs and 26% PRs with median response
was observed. Other side effects were hypoalbuminemia, duration of 6 months [619]. Denileukin diftitox also
428 Immunotoxins

produced remissions in CLL for a total of 27% overall in central nervous system toxicity in 22% of patients at
response (4% CRs and 23% PRs) and median response the MTD of 6 μg/mL × 40 mL [635]. MRI showed tumor
duration of 6 months [620]. Denileukin diftitox given necrosis following treatment. Median survival was 7
for patients with relapsed or refractory B cell non- months. NBI-3001 was administered IV at 0.008–
Hodgkin’s lymphoma yielded 7% CRs and 18% PRs 0.27 mg/m2 days × 5 every 28 days to patients with renal
with median response duration of 7 months [621]. There cancer and non-small cell lung cancer [636]. DLT was
are case reports of denileukin diftitox-induced remis- transient transaminasemia. Neutralizing antibodies were
sions in patients with systemic mastocytosis [622], detected in 71% of patients after two cycles. No responses
extranodal natural killer/T-cell lymphoma [623], periph- were seen.
eral T-cell lymphoma [490], subcutaneous panniculitis- The binding site deleted Pseudomonas exotoxin PE38
like T cell lymphoma [624], and adult T-cell leukemia fragment used to construct BL22 and LMB-2 was fused
[625]. Among non-malignant conditions, denileukin to an anti-mesothelin disulfide stabilized Fv to create
diftitox depletes autoimmune activated T cells yielding the recombinant immunotoxin, SS1P. SS1P was given
a 71% response rate after two to six doses at 9 μg/kg in as 30 min IV infusions every other day for three to six
patients with steroid refractory acute GVHD [626] and doses to patients with advanced mesothelioma, ovarian
yielding remissions in patients with psoriasis [491–493]. cancer and pancreatic cancer at 18–45 μg/kg/dose [637].
Additional studies evaluated the role of expression of DLT was pleuritis, and the MTD was 45 μg/kg × 3 doses.
the individual IL2R subunits in CTCL response. The There were a few minor responses and over half of
alpha subunit expression was measured by immunohis- patients had stable disease. Phase II clinical studies are
tochemistry [627]. Denileukin diftitox responses underway. Combinations with gemcitabine and pacli-
occurred in both CD25 positive and negative tumors, taxel are planned [638, 639]. Novel constructs with
although patients with strongly positive tumors were releasable PEGylation have been tested in animals and
more likely to respond. Responses were also more likely they maintain efficacy with reduced immunogenicity
at 18 μg/kg versus 9 μg/kg [486]. Rare toxicities included and toxicity [616].
thyrotoxicosis and optic neuritis [628, 629]. PRX302 is a furin cleavage site modified proaerolysin.
Combinations of denileukin diftitox with other agents The new site is selectively cleaved by PSA. It was
including bexarotene, rituximab and hyper-CVAD administered intratumorally to 24 patients with locally
enhanced response rates in CTCL, B-cell non-Hodk- recurrent prostate cancer after primary radiotherapy
gin’s lymphoma, and adult T-cell leukemia, respectively failure [640]. The drug was well tolerated without DLT
[487, 630, 631]. The effects of denileukin diftitox on at doses of 0.03–3 μg/g prostate. Delivery was by a single
IL2R expressing T-regulatory cells has been used to multi-deposit, transrectal ultrasound-guided transperineal
augment immune responses to poxvirus vaccine and intraprostatic injection using a modified brachytherapy
melanomas [632, 633]. technique. PSA levels decreased in 63% of patients. The
TransMid is a chemical conjugate of transferrin and the percentage of positive prostate biopsy cores post-therapy
DT-binding site mutant, CRM107. Forty-four patients revealed a decrease in 75% of patients with three patients
with recurrent high-grade gliomas had placement of one showing no positive biopsy cores at 30 days post-therapy.
to two catheters into the tumor beds [483, 484]. Forty A phase IB study has been initiated at multiple institutions
milliliters of TransMid (0.66 μg/ml) was delivered by to optimize the dose and delivery method.
convection over 5 days. There were 12 responders IL13-PEQQR or cintredekin besudotox is composed
including four complete responsers by magnetic reso- of interleukin-13 fused to PE38 with a C-terminal QQR.
nance imaging (MRI) with median survival of >71 Cintredekin besudotox was administered by convection-
weeks. Toxicity was brain injury. TransMid is being enhanced delivery to recurrent glioblastoma multiforme
tested in low grade gliomas and metastatic cancer to tumors for up to 6 days [641]. Optimal dose was
brain [634]. 0.4 mL/h at 0.5 μg/mL over 50 h through two catheters
NBI-3001, IL4(38-37)PE38KDEL, was administered with systemic corticosteroids. Toxicity was tumor
by one to three stereotactic catheters at 0.2–6 μg/mL in necrosis and cerebral edema. 33% of patients had remis-
30–185 mL over 4–8 days to nine patients with recurrent sions lasting 7–117+ weeks. Response and survival
high-grade gliomas [517]. Six of nine patients showed depended upon optimal catheter placement. Unique
tumor necrosis by MRI and histopathology. There was characteristics of patient brain and tumor growth caused
one unmaintained CR lasting >18 months. NB-3001 complex fluid distribution patterns after catheter place-
given at 6 μg/mL × 40 mL to 9 μg/mL × 100 mL intratu- ment and convection delivery. Both 123I-labeled human
morally via one to three catheters over 4–8 days resulted serum albumin PET imaging and MR diffusion tensor
Arthur E. Frankel et al. 429

imaging predicted patient-specific drug distribution by development [158, 159, 228, 242, 266, 319, 415, 428,
convection-enhanced delivery and may improve pro- 494–538].
spective selection of catheter trajectories and targeted
toxin efficacy [642].
The catalytic and translocation domains of diphtheria
Pharmacokinetics and Tissue Distribution
toxin (DT388) were fused via a Met-His linker to human Larger molecules have longer half-lives in the circulation
interleukin-3 to make DT388IL3. DT388IL3 was admin- and poorer tissue penetration. The half-life of monoclonal
istered IV over 15 min every other day for up to six doses antibody conjugates with RTA, ricin, PAP, and PE ranged
to 45 patients with poor-risk, relapsed or refractory AML from 9 to 24 h [266, 427, 494, 497, 499]. The molecular
or MDS [643]. Half-life was 30 min, and peak levels weights were all around 200,000 Mr. There was likely
were dose dependent. An inter-patient dose escalation little clearance of these molecules via renal glomeruli,
schema was used with doses from 4 to 12.5 μg/kg/dose. and penetration into extravascular sites such as nodes,
DLT was not observed, but side effects included tran- marrow, or skin nodules was poor [158, 228, 536]. In con-
sient transaminasemia, hypoalbuminemia, fever, chills, trast, the smaller recombinant immunotoxins (60,000–
nausea and vomiting. Antibodies to drug developed 70,000 Mr) have had shorter half-lives of 1–5 h [279, 481,
between day 15 and day 30 in most patients. Responses 485, 501, 531] and should have better penetration into
included one CR lasting 8 months. And two PRs of 3 and extravascular sites of disease. However, there are no reports
4 months. Duration. A phase IB inter-patient dose escala- to date regarding the saturation of extravascular tumor
tion study in AML and MDS patients with 10–40% mar- deposits in clinical trials with recombinant immuno-
row blast index (cellularity fraction times percent blasts) toxins. The shorter circulation times and increased vascular
with five daily doses is ongoing. A higher affinity vari- permeability of the smaller recombinant immunotoxins
ant, DT388K116W, is in final stages of testing and is may contribute to their greater antitumor efficacy and
expected to begin clinical trials in late 2008 [644]. reduced endothelial toxicity (see below).
ScFv(FRP5)-ETA was prepared by fusing the scFv of
the anti-erbB2 monoclonal antibody FRP5 to domains II
and III of Pseudomonas exotoxin. ScFv(FRP5)-ETA was
Immune Responses
injected intratumorally once daily for 7–10 days with Because most individuals in the US are immunized with
daily doses of 60–900 μg in patients with subcutaneous diphtheria toxoid as children, almost half of adults have
tumor nodules of metastatic breast cancer, colorectal can- anti-DT antibody titers pretreatment with DT conjugates
cer and malignant melanoma [645]. Adverse reactions [540]. Further, between 10% and 20% of patients have
were local injection site pain and inflammation. Partial or had previous Pseudomonas infections (often subclinical)
complete regression of injected nodules was seen in 20% yielding pretreatment anti-PE antibody titers [541].
and 40% of patients, respectively. Next, patients with Patients exposed to castor oil may show anti-ricin anti-
metastatic breast cancer, prostate cancer, head and neck bodies. These circulating antibodies reduce the half-life
cancer, non-small cell lung cancer and transitional cell and AUC for immunotoxins and, in some cases, neutralize
carcinoma were treated on an inter-patient dose escala- cell cytotoxicity [266, 486, 542]. Even if patients lack pre-
tion schedule systemically with 2–20 μg/kg bolus infu- treatment immunity, after administration of the foreign
sion daily for 5 days of each wk for 2 weeks [646]. protein most patients develop anti-immunotoxin antibod-
The DLT was transient transaminasemia. The MTD ies. The exceptions include CLL patients and some
was 12.5 μg/kg, and the peak drug level was 100 ng/mL. patients with severe immunosuppression. PEGylation or
Most patients developed anti-scFv(FRP5)-ETA antibodies use of human ligands and toxins may reduce the immune
after 8 days. There were no responses. Phase II studies response [582, 616].
are ongoing.
Responses have been observed with other immuno-
toxins including Fab’(antiCD22)-dgA, HerH2-saporin,
Toxicities
TP40, DAB486IL2, LMB-1, IgG-HD37-dgA, DA7, There are two general classes of side-effects due to
DAB389EGF, antiB4-blocked ricin, T101-RTA, H65- immunotoxins. In some instances the targeted toxin
RTA, 260F9-rA, XMMME001-RTA, N901-blocked receptor/antigen is present on normal tissues. This can
ricin, IgRFT5-dgA, SPV-T3a-dgA/WT1-dgA, DT388 lead to significant toxicities. LMB-1 and LMB-7 bind the
GMCSF, B43-PAP, IgRFB4-dgA, 791/T36-RTA, anti- Lewisy antigen which is overexpressed on many carci-
TAC-PE, OVB3-PE, 454A12-rA, erb38, and LMB-7 nomas. The Lewisy antigen is also expressed on normal
but these molecules have not undergone further stomach mucosa. Initial DLTs for these immunotoxins
430 Immunotoxins

were nausea, vomiting, and diarrhea with endoscopic injury was reversible. VLS is a syndrome observed with
biopsy evidence of gastritis [266, 530]. After prophy- interleukin-2, monoclonal antibodies, some chemotherapy
laxis with omeprozole, antiemetics, and loperamide, drugs such as taxotere, and many immunotoxins. The
this side effect was alleviated and dose escalation could syndrome usually has onset of 4–6 days after initiating
proceed. 260F9-rA binds a 55 kDa Mr antigen found on therapy and last 410 days. ‘Leaky’ capillaries throughout
breast carcinomas and normal Schwann cells [158]. The the body produce low serum albumin, edema, weight
clinical trial was stopped after the occurrence of revers- gain, dyspnea, aphasia, rhabdomyolysis, and proteinuria.
ible, but prolonged, peripheral neuropathies with The abnormal vascular endothelium may be intoxicated
biopsy-confirmed axonal loss. Similarly, clinical trials by the immunotoxin directly or may be ‘activated’ by
with OVB3-PE and 454A12-rA were discontinued after elevated circulating cytokines. Evidence from tissue
CNS toxicity and ligand crossreactivity with normal culture and animal models has been reported for a direct
CNS tissue antigens (an antigen in the pons for OVB3 binding site of toxins on vascular endothelial cells and
and transferrin receptors on normal brain capillaries for for an indirect toxic effect of immunotoxin-triggered
454A12) were found [242, 524]. DAB389EGF and erb- inflammatory cytokines on endothelial cells [545, 546].
38 reacted with EGFR and HER-2 receptors present on There do not appear to be effective methods of prophy-
normal hepatocytes and produced dose-limiting liver laxis, although paradoxically active hydration appears
injury [527, 532]. DT388GMCSF reacts with normal to reduce the incidence and severity of VLS. Further,
liver macrophages – Kupffer cells. Subsequent cytokine smaller recombinant immunotoxins appear to have a
release triggers dose-limiting hepatocyte damage [531]. lower incidence, possibly due to their shorter circulating
Tf-CRM107 produced focal brain injury in the peritu- half-life. Excluding patients with prior radiotherapy
moral normal cortex [483]. Histopathology showed may also lower the incidence or severity of the VLS
crossreactivity and damage to normal brain capillaries. syndrome [8]. The rare cases of HUS and rhabdomyolysis
Toxicities that are independent of ligand have been may be patient-specific types of vascular injury or due to
observed in almost every immunotoxin clinical study. ligand (particularly anti-CD22 antibodies). Allergic or
The similarity of the toxicities observed with different anaphylactoid reactions occur very rarely, are IgE-
immunotoxin trials suggests that the lesions are due to mediated, and are treated in a similar way to allergic
the toxin moieties. The five classes of side effects are: reactions to other foreign proteins such as L-asparaginase.
(a) transient constitutional symptoms with fever, chills, Fluids, cardiac monitoring, oxygen, corticosteroids,
nausea, vomiting, myalgias, arthralgias, asthenia and diphenhydramine, theophyllines, and rarely epinephrine
hypotension; (b) transient hepatotoxicity with elevated can be given. Patients with anaphylactoid reactions
transaminases and, rarely, other liver enzymes; and (c) should not be retreated with the same immunotoxin.
transient vascular leak syndrome (VLS) consisting of
edema, hypoalbuminemia, weight gain, and, rarely, dys-
pnea and aphasia; (d) assorted syndromes including
rhabdomyolysis, hemolytic uremic syndrome, and renal Ongoing and Future Clinical
insufficiency with proteinuria and renal tubular acido-
sis; and (e) allergic reactions. The toxicities may be due
Studies
to binding of the toxin moieties to normal tissues. The current and near-future applications of immunotoxins
Reaction of immunotoxins with macrophages, lympho- are focused on expanding the use of some of the active,
cytes, or endothelium may lead to cytokine release with established agents described above, as well as testing
consequent constitutional symptoms and elevated circu- novel compounds. Further protocols are being performed
lating cytokines. Elevations of interleukin-6 and tumor with denileukin diftitox, TransMID, NBI-3001, DT388IL3,
necrosis factor alpha have been reported in some but not DT388K116W,LMB-2,Cintredekinbesudotox, AdmDT390-
all cases [481, 495, 497, 531]. Where studied, corticos- bisFv(UCHT1), BL22, HA22, PRX302, scFv(FRP5)-ETA,
teroids or infiiximab + rofecoxib appeared to dampen or PrAgU2/FP59, and DT2219ARL.
eliminate this side-effect [481, 487]. These studies should help define the niche for immu-
The hepatotoxicity may be directly due to immunotoxin notoxins in the eventual multi-agent armamentarium for
binding affected by the pI of the ligand in the conjugate therapy of cancer. The past 2 decades are finally showing
or secondary release of cytokine from macrophages the original promise for these ‘magic bullets’. Discoveries
may damage the liver [543, 544]. In most cases, impair- in the next few years of methods to modulate cell surface
ments of hepatic function such as factor VII production receptor number and physiology, as well as methods to
or bilirubin clearance were not observed, and the liver ameliorate normal tissue toxicities of these agents,
Arthur E. Frankel et al. 431

should refine the applications of these chimeric proteins 16. Casellas P, Dussossoy D, Falasca AI et al. Trichokirin, a ribosome-
in the management of cancer and autoimmune disorders. inactivating protein from the seeds of Trichosanthes kirilowii
Maximowicz. Purification, partial characterization and use for
preparation of immunotoxins. Eur J Biochem 1988; 176: 581–8.
17. Ovadia, M, Hager, CC, Oeltmann, TN. An anti-melanoma-barley
References ribosome inactivating protein conjugate is cytotoxic to melanoma
cells in vitro. Anticancer Res 1990; 10: 671–5.
1. Senekowitsch-Schmidtke R, Schuhmacher C, Becker KF et al. 18. Francisco JA, Gawlak SL, Siegall CB. Construction, expression
Highly specific tumor binding of a 213Bis-labeled monoclonal and characterization of BD 1-G28–5 sFv, a single-chain anti-CD40
antibody against mutant E-cadherin suggests its usefulness for immunotoxin containing the ribosome-inactivating protein bryodin 1.
locoregional alpha-radioimmunotherapy of diffuse-type gastric J Biol Chem 1997; 272: 24165–9.
cancer. Cancer Res 2001; 61: 2804–8. 19. Porro G, Bolognesi A, Caretto P et al. In vitro and in vivo properties
2. Hellstrom I, Hellstrom KF, Senter PD. Development and activities of an anti-CD5-momordin immunotoxin on normal and neoplastic
of the BR96-doxorubicin immunoconjugate. Meth Mol Biol 2001; T lymphocytes. Cancer Immunol Immunother 1993; 36: 346–50.
166: 3 16. 20.Bolognesi A, Tazzari PL, Tassi C, Groom G, Gobbi M, Stirpe F. A
3. Gottstein C, Wels W, Ober B, Thorpe PE. Generation and charac- comparison of anti-lymphocyte immunotoxins containing different
terization of recombinant vascular targeting agents from hybri- ribosome-inactivating proteins and antibodies. Clin Exp Immunol
doma cell lines. Biotechniques 2001; 30: 190–4. 1992; 89: 341–6.
4. Juhl H, Petrella EC, Cheung NK, Bredehorst R, Vogel CW. Additive 21. Siegall CB, Gawlak SL, Chace D, Wolf, EA, Mixan B, Marquardt
cytotoxicity of different monoclonal antibody cobra venom factor H. Characterization of ribosome-inactivating proteins isolated
conjugates for human neuroblastoma cells. Immunobiology 1997; from Bryonia dioica and their utility as carcinoma-specific immu-
197: 444–59. noconjugates. Bioconj Chem 1994; 5: 423–9.
5. Schrama D, thor Straten P, Fischer WH et al. Targeting of lympho- 22. Bolognesi A, Tazzari PL, Legname G et al. Anti-CD30 immuno-
toxin-alpha to the tumor elicits an efficient immune response asso- toxins with native and recombinant dianthin 30. Cancer Immunother
ciated with induction of peripheral lymphoid-like tissue. Immunity Immunother 1995; 40: 109–14.
2001; 14: 111–21. 23. Gasanov SF, Rael FD, Gasanov NF, Vernon LP. In vitro evaluation
6. Bhakdi S, Bayley H, Valeva A et al. Staphylococcal alphatoxin, of Pyrularia thionin-anti-CD5 immunotoxin. Cancer Immunol
streptolysin-O, and Escherichia coli hemolysin: prototypes of Immunother 1995; 41: 122–8.
pore-forming bacterial cytolysins. Arch Microbiol 1996; 165: 24. Huang CA, Yamada K, Murphy MC et al. In vivo T cell depletion in
73–9. miniature swine using the swine CD3 immunotoxin, pCD3-CRM9.
7. Sears CL, Kaper JB. Enteric bacterial toxins: mechanisms of Transplantation 1999; 68: 855–60.
action and linkage to intestinal secretion. Microbial Rev 1996; 60: 25. Onda M, Olafsen T, Tsutsumi Y, Bruland OS, Pastan I. Cytotoxicity
167–215. of anti-osteosarcoma recombinant immunotoxins composed of
8. Schindler J, Sausville E, Messmann R, Uhr JW, Vitetta ES. The TP-3 Fv fragments and a truncated Pseudomonas exotoxin A.
toxicity of deglycosylated ricin A chain-containing immunotoxins J Immunother 2001; 24: 144–50.
in patients with non-Hodgkin’s lymphoma is exacerbated by prior 26. Liu S, Bugge TH, Leppla SH. Targeting of tumor cells by cell sur-
radiotherapy: a retrospective analysis of patients in five clinical face urokinase plasminogen activator-dependent anthrax toxin. J
trials. Clin Cancer Res 2001; 7: 255–8. Biol Chem 2001; 276: 17976–84.
9. Tsai LC, Chen YL, Lee C et al. Growth suppression of human 27. Deonarain MP, Epenetos AA. Design, characterization and anti-
colorectal carcinoma in nude mice by monoclonal antibody C27- tumour cytotoxicity of a panel of recombinant, mammalian ribo-
abrin A conjugate. Dis Colon Rectum 1995; 38: 1067–74. nuclease-based immunotoxins. Br J Cancer 1998; 77: 537–46.
10. Tonevitsky AG, Agapov II, Shamshiev AT, Temyakov DE, Pohl P, 28. Newton DL, Nicholls PJ, Rybak SM, Youle RJ. Expression and
Kirpichnikov MP. Immunotoxins containing A chain of mistletoe characterization of recombinant human eosinophil derived neuro-
lectin I are more active than immunotoxins with ricin A-chain. toxin and eosinophil-derived neurotoxin-antitransferrin receptor
FFBS Lett 1996; 392: 166–8. sFv. J Biol Chem 1994; 269: 26739–45.
11. Rosenblum MG, Shawver LK, Marks JW, Brink J, Cheung L, 29. Newton DL, Xue Y, Olson KA, Fett JW, Rybak SM. Angiogenin
Langton-Webser B. Recombinant immumotoxins directed against single-chain immunofusions: influence of peptide linkers and spacers
the c-erb-2/HER2/neu oncogene product: in vitro cytodoxicity, between fusion protein domains. Biochemistry 1996; 35: 545–53.
pharmacokinetics, and in vivo efficacy studies in xenograft models. 30. Psarras K, Ueda M, Yamamura T et al. Human pancreatic Rnase
Clin Cancer Res 1999; 5: 865–74. l-human epidermal growth factor fusion: an entirely human
12. Ritter S, Bugarith K, Dinh TT. Immunotoxic destruction of distinct ‘immunotoxin analog’ with cytotoxic properties against squamous
catecholamine subgroups produces selective impairment of gluco- cell carcinomas. Protein Eng 1998; 11: 1285–92.
regulatory responses and neuronal activation. J Comp Neurol 31. Gho YS, Chae CB. Luteinizing hormone releasing hormone-
2001; 432: 197–216. RNase A conjugates specifically inhibit the proliferalion of LHRH-
13. Schlick J, Dulieu P, Desvoyes B, Adami P, Radom J, Jouvenot M. receptor-positive human prostate and breast tumor cells. Mol Cells
Cytotoxic activity of a recombinant GnRH-PAP fusion toxin on 1999; 9: 31–6.
human tumor cell lines. FEBS Lett 2000; 472: 241–6. 32. Newton DL, Hansen HJ, Mikalski SM, Goldenberg DM, Rybak
14. Ramakrishnan S, Enghlid JJ, Bryant HL, Xu FT. Characterization SM. Potent and specific antitumor effects of an anti-CD22-targeted
of a translation inhibitory protein from Luffa aegyptiaca. Biochem cytotoxic ribonuclease: potential for the treatment of non-Hodg-
Biophys Res Common 1989; 160: 509–16. kin’s lymphoma. Blood 2001; 97: 528–35.
15. Bolognesi A, Polito L, Tazzari PL et al. In vitro anti-tumour activity 33. Rathore D, Nayak SK, Bator JK. Overproduction of fungal ribot-
of anti-CD80 and anti-CD86 immunotoxins containing type 1 ribo- oxin alpha-sarcin in Escherichia coli: generation of an active
some-inactivating proteins. Br J Haematol 2000; 110: 351–61. immumotoxin. Gene 1997; 190: 311–35.
432 Immunotoxins

34. Goyal A, Batra JK. Inclusion of a furin-sensitive spacer enhances 54. Endo Y, Mitsui K, Motizuki M, Tsurugi K. The mechanism of
the cytotoxicity of ribotoxin restrictocin containing recombinant action of ricin and related toxic lectins on eukaryotic ribosomes. J
single-chain immunotoxins. Biochem J 2000; 345: 247–54. Biol Chem 1987; 262: 5908–12.
35. Parente D, Raucci G, Celano B, Pacilla et al. Clavin, a type 1 ribo- 55. Gawlak SL, Neubauer M, Klei HE, Chang CYY, Einspahr HM,
some-inactivating protein from Aspergillus clavatus IFO 8605. Siegall CB. Molecular, biological, and preliminary structural anal-
cDNA isolation, heterologous expression, biochemical and bio- ysis of recombinant bryodin 1, a ribosome inactivating protein
logical characterization of the recombinant protein. Eur J Biochem from the plant Bryonia dioica. Biochemistry 1997; 36: 3095–103.
1996; 239: 272–80. 56. Savino C, Federici L, Ippoliti R, Lendaro E, Tsernoglou D. The
36. Chovnick A, Schneider WP Tso JY, Queen C, Chang CN. A recom- crystal structure of saporin S06 from Saponaria officinalis and its
binant, membrane-acting immunotoxin. Cancer Res 1991; 51: interaction with the ribosome. FEBS Lett 2000; 470: 239–43.
465–7. 57. Legname G, Bellosta P, Gromo G et al. Nucleotide sequence of
37. Avila AD, Mateo de Acosta C, Lage A. A carcinoembryonic anti- cDNA coding for dianthin 30, a ribosome inactivating protein
gen-directed immunotoxin built by linking a monoclonal antibody from Dianthus caryophyllus. Biochim Biophys Acta 1991; 1090:
to a hemolytic toxin. Int J Cancer 1989; 43: 926–9. 119–22.
38. Yoshida S, Ioka D, Matsuoka H, Endo H, Ishii A. Bacteria express- 58. Monzingo AF, Collins EJ, Ernst SR, Irvin JD, Robertus JD. The
ing single-chain immunotoxin inhibit malaria parasite develop- 2.5 Angstrom structure of pokeweed antiviral protein. J Mol Biol
ment in mosquitoes. Mol Biochem Parasitol 2001; 113: 89–96. 1993; 233: 705–15.
39. Gasanov SE, Rael ED, Gasanov NE, Vernon LP. In vitro evalua- 59. Husain J, Tickle IJ, Wood SP. Crystal structure of momordin, a
tion of Pyrularia thionin-anti-CD5 immumotoxin. Cancer Immunol type I ribosome inactivating protein from the seeds of Momordica
Immunother 1995; 41: 122–8. charantia. FEBS Lett 1994; 342: 154–8.
40. Halling KC, Halling AC, Murray EE, Ladin BF, Houston LL, 60. Hosur MV, Nair B, Satyamurthy P, Misquith S, Surolia A, Kannan
Weaver RE. Genomic cloning and characterization of a ricin gene KK. X-ray structure of gelonin at 1.8 Angstrom resolution. J Mol
from Ricinus communis. Nucl Acids Res 1985; 13: 8019–33. Biol 1995; 250: 368–80.
41. Wood KA, Lord JM, Wawrzynczak EI, Piatak M. Preproabrin: 61. Stirpe F, Bailey S, Miller SP, Bodley JW. Modification of ribo-
genomic cloning, characterization and the expression of the somal RNA by ribosome-inactivating proteins from plants. Nucl
A-chain in Escherichia coli. Eur J Biochem 1991; 198: 723–32. Acids Res 1988; 16: 1349–57.
42. Eschenburg S, Krauspenhaar R, Mikhailov A, Stoeva S, Betzel C, 62. Frankel AE, Welsh P, Richardson J, Robertus JD. Role of arginine
Voelter W. Primary structure and molecular modeling of mistletoe 180 and glutamic acid 177 of ricin toxin A chain in enzymatic inac-
lectin I from Viscum album. Biochem Biophys Res Commun 1998; tivation of ribosomes. Mol Cell Biol 1990; 10: 6257–63.
247: 367–72. 63. Greenfield L, Bjorn MJ, Horn G et al. Nucleotide sequence of the
43. Lord JM. Synthesis and intracellular transport of lectin and storage structural gene for diphtheria toxin carried by corynebacteriophage
protein precursors in endosperm from castor beans. Eur J Biochem beta. Proc Natl Acad Sci USA 1983; 80: 6853–7.
1985; 146: 403–9. 64. Ratti G, Rappuoli R, Giannini G. The complete nucleotide sequence
44. Montfort W, Villafranca JE, Monzingo AF et al. The three-dimensional of the gene coding for diphtheria toxin in the corynephage omega
structure of ricin at 2.8 Angstroms. J Biol Chem 1987; 262: (tox+) genome. Nucl Acids Res 1983; 11: 6589–95.
5398–403. 65. Cloe S, Bennett MJ, Fujii G et al. The crystal structure of diphthe-
45. Tahirov TH, Lu TH, Liaw YC, Chen YL, Lin JY. Crystal structure ria toxin. Nature 1992; 357: 216–22.
of abrin-a at 2.14 Angstroms. J Mol Biol 1995; 250: 354–67. 66. Bell CE, Eisenberg D. Crystal structure of nucleotide-free diphthe-
46. Krauspenhuar R, Eschenburg S, Perbandt M et al. Crystal structure ria toxin. Biochemistry 1997; 36: 481–8.
of mistletoe lectin I from Viscum album. Biochem Biophys Res 67. Brooke JS, Cha JH. Molecular characterization of key diphtheria
Commun 1999; 257: 418–24. toxin:receptor interactions. Biochem Biophys Res Commun 2000;
47. Frankel AE, Burbage C, Fu T, Tagge E, Chandler J, Willingham 275: 374–81.
MC. Ricin toxin contains at least three galactose binding sites 68. Louie GV, Yang W, Bowman ME, Choe S. Crystal structure of the
located in B chain subdomains 1alpha, 1beta, and 2gamma. complex of diphtheria toxin with an extracellular fragment of its
Biochemistry 1996; 35: 14749–56. receptor. Mol Cell 1997; 1: 67–78.
48. Sleeves RM, Denton ME, Barnard FC, Henry A, Lambert JM. 69. Cha J, Brooke JS, Ivey KN, Eidels L. Cell surface monkey CD9
Identification of three oligosaccharide binding sites in ricin. antigen is a coreceptor that increases diphtheria toxin sensitivity and
Biochemistry 1999; 38: 11677–85. diphtheria toxin receptor affinity. J Biol Chem 2000; 275: 6901–7.
49. Llorente A, Rapak A, Schmidt SL, van Deurs B, Sandvig K. 70. Shishido Y, Sharma KD, Higashiyama S, Klagsbrun M, Mekada E.
Expression of mutant dynamin inhibits toxicity and transport of endo- Heparin-like molecules on the cell surface potentiate binding of
cytosed ricin to the Golgi apparatus. J Cell Biol 1998; 140: 553–63. diphtheria toxin to the diphtheria toxin receptor/membrane-
50. Grimmer S, Iversen TG, van Deurs B, Sandvig K. Endosome to anchored heparin-binding epidermal growth factor-like growth
Golgi transport of ricin is regulated by cholesterol. Mol Biol Cell factor. J Biol Chem 1995; 270: 29578–85.
2000; 11: 4205–16. 71. Lanzrein M, Sand O, Olsnes S. GPI-anchored diphtheria toxin
51. Girod A, Storrie B, Simpson JC et al. Evidence for a COP- receptor allows membrane translocation of the toxin without
independent transport route from the Golgi complex to the endo- detectable ion channel activity. EMBO J 1996; 15: 725–34.
plasmic raticulum. Nature Cell Biol 1999; 1: 423–30. 72. Almond BD, Eidels L. The effect of receptor rapid-internaliza-
52. Sandvig K, van Deurs B. Endocytosis and intracellular transport of tion signals on diphtheria toxin endocytosis and cell sensitivity.
ricin: recent discoveries. FEBS Lett 1999; 452: 67–70. Mol Microbiol 1995; 18: 623–30.
53. Wesche J, Rapak A, Olsnes S. Dependence of ricin toxicity on 73. Simpson JC, Smith DC, Roberts LM, Lord JM. Expression of
translocation of the toxin A-chain from the endoplasmic reticulum mutant dynamin protects cells against diphtheria toxin but not
to the cytosol. J Biol Chem 1999; 274: 34443–9. against ricin. Exp Cell Res 1998; 239: 293–300.
Arthur E. Frankel et al. 433

74. Lemichez E, Bomsel M, Devilliers G et al. Membrane transloca- 93. Better M, Bernhard SL, Lei SP Fishwild DM, Carroll SF. Activity
tion of diphtheria toxin fragment A exploits early to late endo- of recombinant mitogillin and mitogillin immunoconjugates. J
some trafficking machinery. Mol Microbiol 1997; 23: 445–57. Biol Chem 1992; 267: 16712–8.
75. Gordon V, Klimpel K, Arora N, Henderson M, Leppla S. 94. Fernandez-Luna JL, Lopez-Otin C, Soriano F, Mendez E.
Proteolytic activation of bacterial toxins by eukaryotic cells is per- Complete amino acid sequence of the Aspergillus cytotoxin mito-
formed by furin and by additional cellular proteases. Infect gillin. Biochemistry 1985; 24: 861–7.
Immum 1995; 63: 82–7. 95. Gray GL, Smith DH, Baldridge JS et al. Cloning, nucleotide
76. Silverman JA, Mindell JA, Finkelstein A, Shen WH, Collier RJ. sequence, and expression in Escherichia coli of the exotoxin A
Mutational analysis of the helical hairpin region of diphtheria toxin structural gene of Pseudomonas aeruginosa. Proc Natl Acad Sci
transmembrane domain. J Biol Chem 1994; 269: 22524–32. USA 1984; 81: 2645–9.
77. Falnes PO, Sandvig K. Penetration of protein toxins into cells. 96. Allured VS, Collier RJ, Carroll SF, McKay DR Structure of exo-
Curr Opin Cell Biol 2000; 12: 407–13. toxin A of Pseudomonas aeruginosa at 3.0-Angstrom resolution.
78. Falnes PO, Choe S, Madshus IH, Wilson BA, Olsnes S. Inhibition Proc Natl Acad Sci USA 1986; 83: 1320–4.
of membrane translocation of diphtheria toxin A-fragment by 97. Hessler JL, Kreitman RJ. An early step in Pseudomonas exotoxin
internal disulfide bridges. J Biol Chem 1994; 269: 8402–7. action is removal of the terminal lysine residue, which allows bind-
79. Papini E, Rappuoli R, Murgia M, Montecucco C. Cell penetra- ing to the KDEL receptor. Biochemistry 1997; 36: 14577–82.
tion of diphtheria toxin. Reduction of the interchain disulfide 98. Kounnas MZ, Morris RE, Thompson MR, FitzGerald DJ,
bridge is the rate-limiting step of translocation in the cytosol. J Strickland DK, Saelinger CB. The α2-macroglobulin receptor/
Biol Chem 1993; 268: 1567–74. low density lipoprotein receptor-related protein binds and inter-
80. Falnes PO, Olsnes S. Cell-mediated reduction and incomplete nalizes Pseudomonas exotoxin A. J Biol Chem 1992; 267:
membrane translocation of diphtheria toxin mutants with inter- 12420–23.
nal disulfides in the A fragment. J Biol Chem 1995; 270: 99. Jinno Y, Chaudhary VK, Kondo T, Adhya S, Pastan I. Mutational
20787–93. analysis of domain I of Pseudomonas exotoxin. Mutations in
81. Falnes PO, Ariansen S, Sandvig K, Olsnes S. Requirement for domain I of Pseudomonas exotoxin which reduce cell binding
prolonged action in the cytosol for optimal protein synthesis inhi- and animal toxicity. J Biol Chem 1988; 263: 13203–7.
bition by diphtheria toxin. J Biol Chem 2000; 275: 4363–8. 100. Manhart MD, Morris RE, Bonventre IT, Leppla S, Saelinger CB.
82. Bodley JW, Dunlop PC, Van Ness BG. Diphthamide in elonga- Evidence for Pseudomonas exotoxin A receptors on plasma
tion factor 2: ADP-ribosylation, purification, and properties. membrane of toxin-sensitive LM fibroblasts. Infect Immun 1984;
Methods Enzymol 1984; 106: 378–87. 45: 596–603.
83. Domenighini M, Rappuoli R. Three conserved consensus 101. Ogata M, Fryling CM, Pastan I, FitzGerald DJ. Cellmediated
sequences identify the NAD-binding site of ADP-ribosylating cleavage of Pseudomonas exotoxin between Arg279 and G1y280
enzymes, expressed by eurkaryotes, bacteria and T-even bacte- generates the enzymatically active fragment which translocates
riophages. Mol Microbiol 1996; 21: 667–74. to the cytosol. J Biol Chem 1992; 267: 25396–401.
84. Bell CE, Eisenberg D. Crystal structure of diphtheria toxin bound to 102. Inocencio NM, Moehring JM, Moehring TJ. Furin activates
nicotinamide adenine dinucleotide. Biochemistry 1996; 35: 1137–49. Pseudomonas exotoxin A by specific cleavage in vivo and in
85. Wilson KS, Noller HF. Molecular movement inside the transla- vitro. J Biol Chem 1994; 269: 31831–5.
tional engine. Cell 1998; 92: 337–49. 103. McKee ML, FitzGerald DJ. Reduction of furin-nicked
86. Gomez-Lorenzo MG, Spahn CMT, Agrawal RK et al. Three- Pseudomonas exotoxin A: an unfolding story. Biochemistry
dimensional cryo-electron microscopy localization of EF2 in the 1999; 38: 16507–13.
Saccharomyces cerevisiae 80S ribosome at 17.5A resolution. 104. Ogata, M, Chaudhary VK, Pastan, I, FitzGerald, DJ. Processing
EMBO J 2000; 19: 2710–8. of Pseudomonas exotoxin by a cellular protease results in the
87. Brinkmanm U, Brinkmanm E, Gallo M, Scharf U, Pastan I. Role generation of a 37,000-Da toxin fragment that is translocated to
of CAS, a human homologue to the yeast chromosome segrega- the cytosol. J Biol Chem 1990; 265: 20678–85.
tion gene CSEI, in toxin and tumor necrosis factor mediated 105. Jackson ME, Simpson JC, Girod A, Pepperkok R, Roberts LM,
apoptosis. Biochemistry 1996; 35: 6891–9. Lord JM. The KDEL retrieval system is exploited by Pseudomonas
88. Keppler-Hafkemeyer A, Brinkmann U, Pastan I. Role of caspases exotoxin A, but not by Shiga-like toxin-1, during retrograde
in immunotoxin-induced apoptosis of cancer cells. Biochemistry transport from the Golgi complex to the endoplasmic reticulum.
1998; 37: 16934–42. J Cell Sci 1999; 112: 467–75.
89. Yamaizumi M, Mekada E, Uchida T, Okada Y. One molecule of 106. Kreitman RJ, Pastan I. Importance of the glutamate residue of
diphtheria toxin fragment A introduced into a cell can kill the KDEL in increasing the cytotoxicity of Pseudomonas exotoxin
cell. Cell 1978; 17: 245–50. derivatives and for increased binding to the KDEL receptor.
90. Merion M, Schlesinger P, Brooks RM, Moehring JM, Moehring Biochem J 1995; 307: 29–37.
TJ, Sly WS. Defective acidification of endosomes in Chinese 107. Hwang J, FitzGerald DJ, Adhya S, Pastan I. Functional domains
hamster ovary cell mutants cross-resistant: to toxins and viruses. of Pseudomonas exotoxin identified by deletion analysis of the
Proc Nad Acad Sci USA 1983; 80: 5315–9. gene expressed in E coli. Cell 1987; 48: 129–36.
91. Foley BT, Moehring JM, Moehring TJ. Mutations in the elonga- 108. Theuer CP, Buchner J, FirzGerald D, Pastan I. The N-terminal region
tion factor 2 gene which confer resistance to diphtheria toxin and of the 37-kDa translocated fragment of Pseudomonas exotoxin A
Pseudomonas exotoxin A. Genetic and biochemical analysis. J aborts translocation by promoting its own export after microsomal
Biol Chem 1995; 270: 23218–25. membrane insertion. Proc Natl Acad Sci USA 1993; 90: 7774–8.
92. Chung DW, Collier RJ. The mechanism of ADP-ribosylation of 109. Zdanovsky AG, Chiron M, Pastan I, FitzGerald DJ. Mechanism
elongation factor 2 catalyzed by fragment A from diphtheria of action of Pseudomonas exotoxin. Identification of a rate-limit-
toxin. Biochim Biophys Acta 1977; 483: 248–57. ing step. J Biol Chem 1993; 268: 21791–9.
434 Immunotoxins

110. Taupiac MP, Bahian M, Alarm M, Beaumelle B. A deletion within nylate cyclase and of a catalytically inactive variant in which
the translocation domain of Pseudomonas exotoxin A enhances glutamine substitutes for lysine-346. Biochemistry 1991; 30:
translocation efficiency and cytotoxicity concomitantly. Mol 2619–24.
Microbiol 1999; 31: 1385–93. 128. Escuyer V, Duflot E, Sezer O, Danchin A, Mock M. Structural
111. Carroll SF, Collier RJ. Active site of Pseudomonas aeruginosa homology between virulence-associated bacterial adenylate
exotoxin A. Glutamic acid 553 is photolabeled by NAD and cyclases. Gene 1988; 71: 293–8.
shows functional homology with glutamic acid 148 of diphtheria 129. Leppla SH. Bacillus anthracis calmodulin-dependent adenylate
toxin. J Biol Chem 1987; 262: 8707–11. cyclase: chemical and enzymatic properties and interactions with
112. Li M, Dyda F, Benhar I, Pastan I, Davies DR. Crystal structure of eucaryotic cells. Adv Cyclic Nucl Protein Phosphor Res 1984;
the catalytic domain of Pseudomonas exotoxin A complexed with 17: 189–98.
a nicotinamide adenine dinucleotide analog: implications for the 130. Gupta P, Singh A, Chauhan V, Bhathagar R. Involvement of resi-
activation process and for ADP ribosylation. Proc Natl Acad Sci dues 147VYYE-IGK153 in binding of lethal factor to protective
USA 1996; 93: 6902–6. antigen of Bacillus anthracis. Biochem Biophys Res Commun
113. Laithwaite JE, Benn SJ, Yamate J, FitzGerald DJ, LaMarre J. 2001; 280: 158–63.
Enhanced macrophage resistance to Pseudomonas exotoxin A is 131. Quinn CP, Singh Y, Klimpel KR, Leppla SH. Functional mapping
correlated with decreased expression of the low density lipopro- of anthrax toxin lethal factor by in-frame insertion mutagenesis.
tein-related protein. Infect Immun 1999; 67: 5827–33. J Biol Chem 1991; 266: 20124–30.
114. Laurie SM, Robbins AR. A toxin-resistant mouse L-cell mutant 132. Duesbery NS, Webb CP, Leppla SH et al. Proteolytic inactivation
defective in protein transport along the secretory pathway. J Cell of MAP-kinase-kinase by anthrax lethal factor. Science 1998;
Physiol 1991; 147: 215–23. 280: 734–7.
115. Banker DE, Pastan I, Gottesman MM, Herschman HR. An epi- 133. Singh Y, Klimpel KR, Goel S, Swain PK, Leppla SH.
dermal growth factor ricin A chain (EGF-RTA)-resistant mutant Oligomerization of anthrax toxin protective antigen and binding
and an epidermal growth factor Pseudomonas exotoxin (EGF- of lethal factor during endocytic uptake into mammalian cells.
PE)-resistant mutant have distinct phenotypes. J Cell Physiol Infect Immun 1999; 67: 1853–9.
1989; 139: 51–7. 134. Gordon VM, Leppla SH, Hewlett EL. Inhibitors of receptor-
116. Merion M, Schlesinger P, Brooks RM, Moehring JM, MoehringTJ, mediated endocytosis block the entry of Bacillus anthracis ade-
Sly WS. Defective acidification of endosomes in Chinese ham- nylate cyclase toxin but not that of Bordetella pertussis adenylate
ster ovary cell mutants ‘cross-resistant’ to toxins and viruses. cyclase toxin. Infect Immun 1988; 56: 1066–9.
Proc Natl Acad Sci USA 1983; 80: 5315–9. 135. Miller CJ, Elliott JL, Collier RJ. Anthrax protective antigen:
117. Spence MJ, Sicic JF, Foley BT, Moehring TJ. Analysis of mutants prepore-to-pore conversion. Biochemistry 1999; 38: 10432–8.
in alleles of the fur gene from an endoprotease-deficient Chinese 136. Sellman BR, Mourez M, Collier RJ. Dominant-negative mutants
hamster ovary cell strain. Somat Cell Mol Genet 1995; 21: 1–18. of a toxin subunit: an approach to therapy of anthrax. Science
118. Welkos SL, Lowe JR, Eden-McCutchan F, Vodkin M, Leppla SH, 2001; 292: 695–7.
Schmidt JJ. Sequence and analysis of the DIVA encoding protec- 137. Batra S, Gupta P, Chauhan V, Singh A, Bhatnagar R. Trp 346 and
tive antigen of Bacillus anthracis. Gene 1988; 69: 287–300. Leu 352 residues in protective antigen are required for the expres-
119. Petrosa C, Collier RJ, Klimpel KR, Leppla SH, Liddington RC. sion of anthrax lethal toxin activity. Biochem Biophys Res
Crystal structure of the anthrax toxin protective antigen. Nature Commun 2001; 281: 186–92.
1997; 385: 833–8. 138. Rossi AG, McCutcheon JC, Roy N, Chilvers ER, Haslett C,
120. Little SF, Novak JM, Lowe JR et al. Characterization of lethal Dransfield I. Regulation of macrophage phagocytosis of apop-
factor binding and cell receptor binding domains of protective totic cells by cAMP. J Immunol 1998; 160: 3562–8.
antigen of Bacillus anthracis using monoclonal antibodies. 139. Duesbery NS, Webb CP, Vande Woude GF. MEK wars, a new
Microbiology 1996; 142: 707–15. front in the battle against cancer. Nature Med 1999; 5: 736–7.
121. Varughese M, Teixerira AV, Liu S, Leppla SH. Identification of a 140. Boix E, Leonidas DD, Nikolovski Z, Nogues MV, Cuchillo CM,
receptor-binding region within domain 4 of the protective antigen Acharya KR. Crystal structure of eosinophil cationic protein at
component of anthrax toxin. Infect Immun 1999; 67: 1860–5. 2.4 Angstrom resolution. Biochemistry 1999; 38: 16794–801.
122. Friedlander AM, Bhatnagar R, Leppla SH, Johnson L, Singh Y. 141. Fersht A. Enzyme Structure and Mechanism. New York: Freeman
Characterization of macrophage sensitivity and resistance to 1985: 426–31.
anthrax lethal toxin. Infect Immun 1993; 61: 245–52. 142. Antignani A, Naddeo M, Cubellis MV, Russo A, D’Alessio G.
123. Gordon VM, Rehemtulla A, Leppla SH. A role for PACE4 in the Antitumor action of seminal ribonuclease, its dimeric cytosolic
proteolytic activation of anthrax toxin protective antigen. Infect ribonuclease inhibitor. Biochemistry 2001; 40: 3492–6.
Immun 1997; 65: 3370–5. 143. Leonidas DD, Boix E, Pril R et al. Mapping the ribonucleolytic
124. Robertson DL, Tippetts MT, Leppla SH. Nucleotide sequence of active site of eosinophil-derived neurodoxin (EDN). J Biol Chem
the Bacillus anthracis edema factor gene (cya): a calmodulin- 2001; 276: 15009–17.
dependent adenylate cyclase. Gene 1988; 73: 363–71. 144. Hu GF, Xu CJ, Riordan JF. Human angiogenin is rapidly translo-
125. Bragg TS, Robertson D. Nucleotide sequence and analysis of the cated to the nucleus of human umbilical vein endothelial cells
lethal factor gene (lef) from Bacillus anthracis. Gene 1989; 81: and binds to DNA. J Cell Biochem 2000; 76: 452–62.
45–54. 145. Bretscher LE, Abel RL, Raines RT. A ribonuclease A variant with
126. Drum CL, Yan SZ, Sarac R et al. An extended conformation of low catalytic activity but high cytodoxicity. J Biol Chem 2000;
calmodulin induces interactions between the structural domains 275: 9893–6.
of adenylyl cyclase from Bacillus anthracis to promote catalysis. 146. Yang X, Moffat K. Insights into specificity of cleavage and
J Biol Chem 2000; 275: 36334–40. mechanism of cell entry from the crystal structure of the
127. Labruyere E, Mock M, Surewicz WK et al. Structural and ligand- highly specific Aspergillus ribotoxin, restrictocin. Structure
binding properties of a truncated form of Bacillus anthracis ade- 1996; 4: 837–52.
Arthur E. Frankel et al. 435

147. Perez-Canadillas JM, Santoro 1, Campos-Olivas R et al. The 166. Liu S, Bugge TH, Leppla SH. Targeting of tumor cells by cell
highly refined solution structure of the cytotoxic ribonuclease surface urokinase plasminogen activator-dependent anthrax
α-sarcin reveals the structural requirements for substrate recog- toxin. J Biol Chem 2001; 276: 17976–84.
nition and ribonucleolytic activity. J Mol Biol 2000; 299: 167. Aqeilan R, Yarkoni S, Lorberboum-Galski H. Interleukin 2-Bax:
1061–73. a novel prototype of human chimeric proteins for targeted ther-
148. Szewczak AA, Moore PB. The sarcin/ricin loop, a modular RNA. apy. FEBS Lett 1999; 457: 271–6.
J Mol Biol 1995; 247: 81–98. 168. Thompson J, Stavrou S, Weetall M et al. Improved binding of a
149. Naylor CE, Eaton IT, Howells A et al. Structure of the key toxin bivalent single-chain immumotoxin results in increased efficacy
in gas gangrene. Nature Struct Biol 1998; 5: 738–46. for in vivo T-cell depletion. Protein Eng 2001; 14: 1035–41.
150. de los Rios V, Onaderra M, Martinez-Ruiz A et al. Overproduction 169. Vallera DA, Kuroki DW, Panoskaltsis-Mortari A, Buchsbaum DJ,
in Escherichia coli and purification of the hemolytic protein Rogers BE, Blazar BR. Molecular modification of a recombinant
sticholysin II from the sea anemone Stichodactyla helianthus. anti-CD3ε-directed immumotoxin by inducing terminal cysteine
Protein Express Purif 2000; 18: 71–6. bridging enhances anti-GVHD efficacy and reduces organ toxicity
151. Mancheno JM, De los Rios V, Del Pozo AM, Lanio ME, Onaderra in a lethal murine model. Blood 2000; 96: 1157–65.
M, Gavilanes JG. Partially folded states of the cytolytic protein 170. Vallera DA, Panoskaltsis-Mortari A, Jost C et al. Anti-graft-
sticholysin II. Biochim Biophys Acta 2001; 1545: 122–31. versus-host disease effect of DT390-anti-CD3sFv; a single-chain
152. Martin-Benito J, Gavilanes F, de los Rios V, Mancheno JM, Fv fusion immunotoxin specifically targeting the CD3 epsilon
Fernandez JJ, Gavilanes JG. Two-dimensional crystallization on moiety of the T-cell receptor. Blood 1996; 88: 2342–53.
lipid monolayers and three-dimensional structure of stricholysin 171. Colombatti M, Greenfield L, Youle RJ. Cloned fragment of DT
II, a cytolysin from the sea anemone Stichodactyla helianthus. linked to T-cell specific antibody identifies regions of B chain
Biophys 1 2000; 78: 3186–94. active in cell entry. J Biol Chem 1986; 261: 3030–5.
153. Moore Al, Beazley WD, Bibby MC, Devine DA. Antimicrobial 172. Neville DM, Scharf, J, Srinivasachar K. In vivo T-cell ablation by
activity of cecropins. J Antimicrob Chemother 1996; 37: 1077–89. a halo-immunotoxin directed at human CD3. Proc Natl Acad Sci
154. Jaynes JM, Burton CA, Barr SB et al. In vitro cytocidal effect of USA 1992; 89: 2585–9.
novel lytic peptides on Plasmodium falciparum and Trypanosoma 173. Neville DM, Scharff J, Hu HZ. A new reagent for the induction of T
cruzi. FASEB J 1988; 2: 2878–83. cell depletion, anti-CD3-CRM9. J Immunother 1996; 19: 85–92.
155. Vernon LP. Pyrularia thionin: physical properties, binding to 174. Ek O, Waurzyniak B, Myers DE, Uckun FM. Antitumor activity
phospholipid bilayers, and cellular responses. Adv Exp Med Biol of TP3 (anti-p80)-pokeweed antiviral protein immunotoxin in
1996; 391: 279–91. hamster cheek pouch and severe combined immumodeficient
156. Gasnov SE, Vernon LP, Aripov TF. Modification of phospholipid mouse xenograft models of human osteosarcoma. Clin Cancer
membrane structure by the plant toxic peptide Pyrularia thionin. Res 1998; 4: 1641–7.
Arch Biochem Biophys 1993; 301: 367–74. 175. Shaw JP Akiyoshi DE, Arrigo DA et al. Cytotoxic properties of
157. Wang F, Naisbitt GH, Vernon LP, Glaser M. Pyrularia thionin DAB486EGF and DAB389EGF, epidermal growth factor (EGF)
binding to and the role of tryptophan-8 in the enhancement of receptor-targeted fusion toxins. J Biol Chem 1991; 266: 13449–55.
phosphatidylserine domains in erythrocyte membranes. 176. Cawley DR Herschman HR. Epidermal growth factor toxin A
Biochemistry 1993; 32: 12283–9. chain conjugates: EGF-ricin A is a potent toxin while EGF-
158. Gould BJ, Borowitz MJ, Groves ES et al. Phase I study of an anti- diphtheria fragment A is nontoxic. Cell 1980; 22: 563–70.
breast cancer immunotoxin by continuous infusion: report of a 177. Williams DP Snider CE, Strom TR Murphy JR. Structure/function
targeted toxic effect not predicted by animal studies. J Nat Cancer analysis of interleukin-2 toxin (DAB486IL-2): fragment B
Inst 1989; 81: 775–81. sequences required for the delivery of fragment A to the cytosol of
159. Grossbard ML, Gribben JG, Freedman AS et al. Adjuvant immu- target cells. J Biol Chem 1990; 265: 11885–9.
motoxin therapy with anti-B4-blocked ricin after autologous 178. Lorberboum-Galski H, Kozak RW, WaldmannTA, Bailm P,
bone marrow transplantation for patients with B-cell non-Hodgkin’s FitzGerald DJ, Pastan I. Interleukin 2 (IL2) PE40 is cytotoxic to
lymphoma. Blood 1993; 81: 2263–71. cells displaying either the p55 or p70 subunit of the IL2 receptor.
160. Frankel AE, Fu T, Burbage C, Chandler J, Willingham MC, Tagge J Biol Chem 1988; 263: 18650–6.
EP. IL2 fused to lectin-deficient ricin is toxic to human leukemia 179. Kreitman RJ, Pastan I. Recombinant toxins containing human
cells expressing the IL2 receptor. Leukemia 1997; 11: 22–30. granulocyte macrophage colony-stimulating factor and either
161. Gilliland DG, Steplewski Z, Collier RJ, Mitchell KF, Chang TH, Pseudomonas exotoxin or diphtheria toxin kill gastrointestinal
Koprowski H. Antibody-directed cytotoxic agents: use of mono- cancer and leukemia cells. Blood 1997; 90: 252–9.
clonal antibody to direct the action of toxin A chains to colorectal 180. Perentesis JP, Bendel AE, Shan Y et al. Granulocyte mac-
carcinoma cells. Proc Natl Acad Sci USA 1980; 77: 4539–43. rophage colony-stimulating factor receptor-targeted therapy of
162. Loberboum-Galski H, Garsia RJ, Gately M et al. IL2-PE664G1u, chemotherapy-and radiation-resistant human myeloid leukemias.
a new chimeric protein cytodoxic to human activated T lympho- Leuk Lymph 1997; 25: 247–56.
cytes. J Biol Chem 1990; 265: 16311–7. 181. Burbage C, Tagge EP, Harris B et al. Ricin fusion toxin targeted
163. Frankel AE, Powell BL, Vallera DA, Neville DM. Chimeric to the human granulocyte macrophage colony stimulating factor
fusion proteins (diphtheria toxin-based). Curr Opin Invest Drug receptor is selectively toxic to acute myeloid leukemia cells.
2001; 2: 1294–301. Leuk Res 1997; 21: 681–90.
164. Kreitman RJ, Pastan I. Recombinant toxins. Adv Pharmacol 182. Chan C-H, Blazar BR, Eide CR, Kreitman RJ, Vallera DA. A
1994; 28: 193–219. murine cytokine fusion toxin specifically targeting the murine
165. Varughese M, Teixeira AV, Liu S, Leppla SH. Identification granulocyte macrophage colony-stimulating factor (GM-CSF)
of a receptor-binding region within domain 4 of the protective receptor on normal committed bone marrow progenitor cells
antigen component of anthrax toxin. Infect Immum 1999; 67: and GM-CSF-dependent tumor cells. Blood 1995; 86:
1860–5. 2732–40.
436 Immunotoxins

183. Rozemuller H, Rombouts EJC, Touw IP et al. In vivo targeting of agent for the elimination of IL-7 receptor positive cells. Bioconj
leukemia cells using diphtheria toxin fused to murine GM-CSF. Chem 1998; 9: 201–7.
Leukemia 1998; 12: 710–7. 201. Cawley DR Simpson DL, Herschman HR. Asialoglycopro tein
184. Frankel AE, Ramage J, Kiser M, Alexander R, Kucera G, Miller receptor mediates the toxic effects of an asialoetuin diphtheria
MS. Characterization of diphtheria fusion proteins targeted to the toxin fragment A conjugate on cultured rat hepatocytes. Proc Natl
human interleukin-3 receptor. Protein Eng 2000; 13: 575–81. Acad Sci USA 1981; 78: 3383–7.
185. Chan CH, Blazar BR, Greenfield L, Kreitman RJ, Vallera DA. 202. Cai J, Zheng T, Murphy J, Waters CA, Lin GY, Gill PS. IL4R
Reactivity of murine cytokine fusion toxin, diphtheria toxin390- expression in AIDS-KS cells and response to rhIL-4 and IL-4
murine interleukin-3 (DT390-mIL-3), with bone marrow pro- toxin (DAB389-IL-4). Invest New Drugs 1997; 15: 279–87.
genitor cells. Blood 1996; 88: 1445–56. 203. Rand RW, Kreitman RJ, Patronas N, Varricchio F, Pastan I, Puri
186. Liger D, vanderSpek JC, Gaillard C et al. Characterization and RK. Intratumoral administration of recombinant circularly per-
receptor specific toxicity of two diphtheria toxin-related interleu- muted interleukin-4 Pseudomonas exotoxin in patients with high-
kin-3 fusion proteins DAB389-mIL3 and DAB389-(G4S)2- grade glioma. Clin Cancer Res 2000; 6: 2157–65.
mIL3. FEBS Lett 1997; 406: 157–61. 204. Vallera DA, Jin N, Baldrica JMR, Panoskaltsis-Mortari A, Chen
187. Neville DM, Srinivasachar K, Stone R, Scharf, J. Enhancement SY, Blazar BR. Retroviral immunotoxin gene therapy of acute
of immunotoxin efficacy by acid-cleavable cross-linking agents myelogenous leukemia in mice using cytotoxic T cells trans-
utilizing diphtheria toxin and toxin mutants. J Biol Chem 1989; duced with an interleukin 4/diphtheria toxin gene. Cancer Res
264: 14653–61. 2000; 60: 976–84.
188. Johnson VG, Wrobel C, Wilson D et al. Improved tumor specific 205. Lakkis F, Steele A, Pacheco-Silva A, Rubin-Kelley V, Strom TB,
immunotoxins in the treatment of CNS and leptomeningeal neo- Murphy JR. Interleukin 4 receptor targeted cytotoxicity: genetic
plasia. J Neurosurg 1989; 70: 740–8. construction and in vivo immunosuppressive activity of a diph-
189. Bjorn M, Ring D, Frankel AE. Evaluation of monoclonal anti- theria toxin-related murine interleukin 4 fusion protein. Eur J
bodies for the development of breast cancer immunotoxins. Immumol 1991; 21: 2253–8.
Cancer Res 1985; 45: 1214–21. 206. Puri RK, Ogata M, Leland P, Feldman GM, FitzGerald D, Pastan I.
190. Newton DL, Ilercil O, Laske DW, Oldfield E, Rybak SM, Youle Expression of high-affinity interleukin 4 receptors on murine
RJ. Cytotoxic ribonuclease chimeras. Targeted tumoricidal activ- sarcoma cells and receptor-mediated cytotoxicity of tumor cells
ity in vitro and in vivo. J Biol Chem 1992; 267: 19572–8. to chimeric protein between interleukin 4 and Pseudomonas
191. Pirker R, FitzGerald DJ, Willingham MC, Pastan I. Enhancement exotoxin. Cancer Res 1991; 51: 3011–7.
of the activity of immunotoxins made with either ricin A chain or 207. Puri RK, Leland P, Obiri NI et al. Targeting of interleukin 13
Pseudomonas exotoxin in human ovarian and epidermoid carci- receptor on human renal cell carcinoma cells by a recombinant
noma cell lines. Cancer Res 1988; 48: 3919–23. chimeric protein composed of interleukin-13 and a truncated
192. Francis JW, Brown RH, Figueredo D et al. Enhancement of diph- form of Pseudomonas exotoxin A (PE38QQR). Blood 1996;
theria toxin potency by replacement of the receptor binding domain 87: 4333–9.
with tetanus toxin C-fragment: a potential vector for delivering het- 208. Wen Z, Tao X, Lakkis F, Kiyokawa T, Murphy JR. Diphtheria
erologous proteins to neurons. J Neurochem 2000; 74: 2528–36. toxin-related alpha-melanocyte-stimulating hormone fusion
193. Fisher CE, Sutherland JA, Krause JE, Murphy JR, Leeman SE, toxin. J Biol Chem 1991; 266: 12289–93.
vanderSpek JC. Genetic construction and properties of a diphthe- 209. Murphy JR, Bishai W, Borowski M, Miyanohara A, Boyd J,
ria toxin-related substance P fusion protein: in vitro destruction Nagle S. Genetic construction, expression and melanoma selec-
of cells bearing substance P receptors. Proc Natl Acad Sci USA tive cytotoxicity of a diphtheria toxin-related alpha-melanocyte-
1996; 93: 7341–5. stimulating hormone fusion protein. Proc Natl Acad Sci USA
194. Masood R, Lunardi-Iskandar Y, Jean LF et al. Inhibition of AIDS- 1986; 83: 8358–62.
associated Kaposi’s sarcoma cell growth by DAB389-interleukin 210. vanderSpek JC, Sutherland JA, Zang H, Battey JF, Jensen RT,
6. AIDS Res Human Retroviruses 1994; 10: 969–75. Murphy JR. Diphtheria toxin-related fusion protein DAB389GRP.
195. Siegall CB, Kreitman RJ, FitzGerald DJ, Pastan I. Anti tumor Cancer Res 1997; 57: 290–4.
effects of interleukin 6-Pseudomonas exotoxin chimeric mole- 211. Negro A, Skaper SD. Synthesis and cytotoxic profile of a diphthe-
cules against the human hepatocellular carcinoma, PLC/PRF/5 in ria toxin neurotrophin 4 chimera. J Neurochem 1997; 68: 554–63.
mice. Cancer Res 1991; 51: 2831–6. 212. Aullo P, Alcam J, Popoff MR, Klatzmann DR, Murphy JR,
196. Arom N, Masood R, Zheng T, Cai J, Smith DL, Gill PS. Vascular Boquet PA. A recombinant diphtheria toxin related human CD4
endothelial growth factor chimeric toxin is highly active against fusion protein specifically kills HIV infected cells which express
endothelial cells. Cancer Res 1999; 59: 183–8. gp120 but selects fusion toxin resistant cells which carry HIV.
197. Ramakrishnan S, Olson TA, Bautch VL, Mohanraj D. Vascular EMBO J 1992; 11: 575–83.
endothelial growth factor-toxin conjugate specifically inhibits 213. Davey RT, Boenning CM, Herpin BR et al. Use of recombinant
KDR/flk-l-positive endothelial cell proliferation in vitro and soluble CD4 Pseudomonas exotoxin, a novel immunotoxin, for
angiogenesis in vivo. Cancer Res 1996; 56: 1324–30. treatment of persons infected with human immunodeficiency
198. Veenendaal LM, Jin H, Ran S et al. In vitro and in vivo studies of virus. J Infect Dis 1994; 170: 1180–8.
a VEGF121/rGelonin chimeric fusion toxin targeting the neovas- 214. Landgraf R, Pegram M, Slamon DJ, Eisenberg D. Cytotoxicity
culature of solid tumors. Proc Natl Acad Sci USA 2002; 99: and specificity of directed toxins composed of diphtheria toxin
7866–71. and the EGF-like domain of heregulin beta 1. Biochemistry 1998;
199. Marsh JW. Antibody-mediated routing of diphtheria toxin in 37: 3220–8.
murine cells results in a highly efficacious immumotaxin. J Biol 215. Gilliland DG, Collier RJ, Moehring JM, Moehring TJ. Chimeric
Chem 1988; 263: 15993–9. toxins: toxic, disulfide-linked conjugate of conconavalin A with
200. Sweeney ER Foss FM, Murphy JR, vanderSpek JC. Interleukin 7 fragment A from diphtheria toxin. Proc Natl Acad Sci USA 1978;
(IL-7) receptor-specific cell killing by DAB389IL-7: a novel 75: 5319–23.
Arthur E. Frankel et al. 437

216. Yamaguchi T, Kato R, Beppu M et al. Preparation of concanava- 234. Pederzolli C, Belmonte G, Dalla Serra M, Macek P, Menestrina
lin A ricin A-chain conjugate and its biologic activity against G. Biochemical and cytotoxic properties of conjugates of trans-
various cultured cells. J Natl Cancer Inst 1979; 62: 1387–95. ferrin with equinatoxin II, a cytolysin from a sea anemone.
217. Bacha P, Murphy JR, Reichlin S. Thyrotropin-releasing hormone Bioconj Chem 1995; 6: 166–73.
diphtheria toxin-related polypeptide conjugates. J Biol Chem 235. Bera TK, Kennedy PE, Berger EA, Barbas CF, Pastan I. Specific
1983; 258: 1565–70. killing of HIV-infected lymphocytes by a recombinant immuno-
218. Youle RJ, Neville DM. Receptor-mediated transport of the hybrid toxin directed against the HIV-1 envelope glycoprotetn. Mol Med
protein ricin diphtheria toxin fragment A with subsequent ADP- 1998; 4: 384–91.
ribosylation of intracellular elongation factor II. J Biol Chem 236. Pincus SH, Wehrly K, Cole R et al. In vitro effects of anti-HIV
1979; 254: 11089–96. immunotoxins directed against multiple epitopes on HIV type 1
219. vanderSpek JC, Sutherland J, Sampson E, Murphy JR. Genetic envelope glycoprotein 160. AIDS Res Human Retroviruses 1996;
construction and characterization of the diphtheria toxin-related 12: 1041–51.
interleukin 15 fusion protein DAB389sIL15. Protein Eng 1995; 237. Bjorn MJ, Groetsema G. Immunotoxins to the murine transferrin
8: 1317–21. receptor: intracavitary therapy of mice bearing syngeneic perito-
220. Bernhard MI, Foon KA, Oeltmann TN et al. Guinea pig line 10 neal tumors. Cancer Res 1987; 47: 6639–45.
hepatocarcinoma model: characterization of monoclonal antibody 238. Ghetie MA, Morarn I, Margineanu M, Ghetie V. Protein A vec-
and in vivo effect of unconjugated antibody and antibody conju- torized toxins II. Preparation and ‘in vitro’ cytotoxic effect of
gated to diphtheria toxin A chain. Cancer Res 1983; 43: 4420–8. protein A-ricin A chain conjugate on antibody coated human
221. Uchida T, Yamaizumi M, Mekada E et al. Reconstitution of tumour cells. Mol Immunol 1988; 25: 473–7.
hybrid toxin from fragment A of diphtheria toxin and a subunit of 239. Erice A, Balfour HH, Myers DE et al. Anti-human immunodefi-
Wistaria floribunda lectin. J Biol Chem 1978; 253: 6307–10. ciency virus type 1 activity of an anti-CD4 immunoconjugate
222. Chang TM, Dazord A, Neville DM. Artificial hybrid protein con- containing pokeweed antiviral protein. Antimicrob Agents
taining a toxic protein fragment and a cell membrane receptor- Chemother 1993; 37: 835–8.
binding moiety in a disulfide conjugate. J Biol Chem 1977; 252: 240. Wawrzynczak EJ, Watson GJ, Cumber AJ et al. Blocked and non-
1515–22. blocked ricin immunotoxins against the CD4 antigen exhibit
223. Chadwick DE, Williams DP, Niho Y, Murphy JR, Minden MD. higher cytotoxic potency than a ricin A chain immunotoxin
Cytotoxicity of a recombinant diphtheria toxin granulocyte col- potentiated with ricin B chain or with a ricin B chain immuno-
ony-stimulating factor fusion protein on human leukemic blast toxin. Cancer Immunol Immunother 1991; 32: 289–95.
cells. Leuk Lymph 1993; 11: 249–62. 241. Woo BH, Lee IT, Na DH, Lee KC. Sepharose-unbinding ricin E
224. Tojo A, Oshima Y. Targeted toxin therapy in the treatment of leu- as a source for ricin A chain immunotoxin. J Immunol Meth
kemia. Jpn J Clin Hematol 1995; 36: 578–81. 2001; 249: 91–8.
225. Bound TF, Wang QC, Reisfeld RA, Kaplan NO. Monoclonal 242. Pai LH, Bookman MA, Ozols RE et al. Clinical evaluation of intra-
antibody and an antibody toxin conjugate to a cell surface proteo- peritoneal Pseudomonas exotoxin immunoconjugate OVB3-PE in
glycan of melanoma cells suppress in vivo tumor growth. Proc patients with ovarian cancer. J Clin Oncol 1991; 9: 2095–103.
Nad Acad Sci USA 1983; 80: 529–33. 243. Bjorn MJ, Manger R, Sivam G, Morgan AC, Torok-Storb B.
226. Monaco ME, Mack J, Dugan MD, Ceriani R. An antibody toxin Selective elimination of breast cancer cells from human bone
conjugate directed against a human mammary cancer antigen. marrow using an antibody Pseudomonas exotoxin A conjugate.
Ann NY Acad Sci 1986; 464: 389–99. Cancer Res 1990; 50: 5992–6.
227. Gilliland DG, Steplewski Z, Collier RJ, Mitchell KF, Chang TH, 244. Rajagopal V, Kreitman RJ. Recombinant toxins that bind to the
Koprowski H. Antibody-directed cytotoxic agents: use of mono- urokinase receptor are cytotoxic without requiring binding to the
clonal antibody to direct the action of toxin A chains to colorectal alpha2-macroglobulin receptor. J Biol Chem 2000; 275: 7566–73.
carcinoma cells. Proc Natl Acad Sci USA 1980; 77: 4539–43. 245. Cavallaro U, del Vecchio A, Lappi DA, Soria MR. A conjugate
228. Hertler AA, Schlossman DM, Borowitz MJ, Blythman HE, between human urokinase and saporin, a type-1 ribosome-inacti-
Casellas P, Frankel AE. An anti-CD5 immunotoxin for chronic vating protein, is selectively cytotoxic to mokinase receptor-
lymphocytic leukemia: enhancement of cytotoxicity with human expressing cells. J Biol Chem 1993; 268: 23186–90.
serum albumen-monenstn. Int J Cancer 1989; 43: 215–9. 246. Vallera DA, Li C, Jin N, Panoskaltsis-Mortari A, Hall WA.
229. Villemez CL, Carlo PL. Preparation of an immunotoxin for Targeting overexpressed urokinase-type plasminogen activator
Acanthamoeba castellanii. Biochem Rophys Res Common 1984; receptor (uPAR) on human glioblastoma with the diphtheria toxin
125: 25–9. fusion protein DTAT in nude mice. J Natl Cancer Inst 2002; 94:
230. Moolten FL, Capparall NJ, Zajdel SH, Cooperband SR. Antitumor 597–606.
effects of antibody-diphtheria toxin conjugates. II. Immunotherapy 247. Li C, Hall WA, Vallera DA. Diphtheria toxin 390/IL13 fusion
with conjugates directed against tumor antigens induced by simian protein selectively targets receptor expressing human glioblastoma
virus 40. J Natl Cancer Inst 1975; 55: 473–7. growing in nude mice. Protein Eng 2002; 15: 419–27.
231. Mannhalter JW, Gilliland DG, Collier RJ. A hybrid toxin contain- 248. Yang D, Krum CT, Payne J et al. Recombinant heregulin
ing fragment A from diphtheria toxin linked to the B protomer of Pseudomonas exotoxin fusion proteins: interactions with the
cholera toxin. Biochim Biophys Acta 1980; 626: 443–50. heregulin receptors and antitumor activity in vivo. Clin Cancer
232. Gilliland DG, Collier RJ. A model system involving anti-conconava- Res 1998; 4: 993–1004.
lin A for antibody targeting of diphtheria toxin fragment Al. 249. Gawlak SL, Pastan I, Siegall CB. Basic fibroblast growth factor
Cancer Res 1980; 40: 3564–9. Pseudomonas exotoxin chimeric proteins; comparisons with
233. Gilliland DG, Collier RJ. Characterization of hybrid molecules acidic fibroblast growth factor Pseudomonas exotoxin. Bioconj
containing fragment A from diphtheria toxin linked to concanav- Chem 1993; 4: 483–9.
alin A or the binding subunit of ricin toxin. J Biol Chem 1981; 250. Brinkmanm U, Gallo M, Brinkmann E, Kunwar S, Pastan I. A
256: 12731–9. recombinant immunotoxin that is active on prostate cancer cells
438 Immunotoxins

and that is composed of the Fv region of monoclonal antibody 266. Pai LH, Wittes R, Setser A, Willingham MC, Pastan I. Treatment
PR1 and a truncated form of Pseudomonas exotoxin. Proc Nad of advanced tumors with immunotoxin LMB-1: an antibody
Acad Sci USA 1993; 90: 547–51. linked to Pseudomonas exotoxin. Nature Med 1996; 2: 350–3.
251. Mickisch GH, Pai LH, Siegsmund M, Campain J, Gottesman 267. Benhar I, Pastan I. Characterization of B1(Fv)FE38 and B1(dsFv)
MM, Pastan I. Pseudomonas exotoxin conjugated to monoclonal PE38: single-chain and disulfide-stabilized Fv immunotoxins
antibody MRK16 specifically kills multidrug resistant cells in with increased activity that cause complete remissions of estab-
cultured renal carcinomas and in MDR-transgenic mice. J Urol lished human carcinoma xenografts in nude mice. Clin Cancer
1993; 149: 174–8. Res 1995; 1: 1023–9.
252. Merwin JR, Lynch MJ, Madri JA, Pastan I, Siegall CB. Acidic 268. Debinski W, Pastan I. Recombinant F(ab’) C242 Pseudomonas
fibroblast growth factor Pseudomonas exotoxin chimeric protein exotoxin, but not the whole antibody-based immunotoxin, causes
elicits antiangiogenic effects on endothelial cells. Cancer Res regression of a human colorectal tumor xenograft. Clin Cancer
1992; 52: 4995–5001. Res 1995; 1: 1015–22.
253. Kreitman RJ, Schneider WP Queen C et al. Mik-beta 1(Fv)-PE40, 269. Wolff EA, Hellstrom I, Chace DE Hellstrom KE, Siegall CB.
a recombinant immunotoxin cytotoxic toward cells bearing the Anti-tumour activity of a melanoma-specific immunotoxin,
beta-chain of the IL-2 receptor. J Immunol 1992; 149: 2810–5. ME20-LysPE40. Ther Immunol 1995; 2: 137–45.
254. Godal A, Kumle B, Pihl A, Juell S, Fodstad O. Immunotoxins 270. Francisco JA, Gilliland LK, Stebbins MR, Norris NA, Ledbetter
directed against the high-molecular weight melanoma associated JA, Siegall CB. Activity of a single-chain immunotoxin that
antigen. Identification of potent antibody toxin conjugates. Int J selectively kills lymphoma and other B-lineage cells expressing
Cancer 1992; 52: 631–5. the CD40 antigen. Cancer Res 1995; 55: 3099–104.
255. Debinski W, Karlsson B, Lindholm L et al. Monoclonal antibody 271. Nechushtan A, Yarkoni S, Marianovsky I, Lorberbaum-Galski H.
C242 Pseudomonas exotoxin A. A specific and potent immuno- Adenocarcinoma cells are targeted by the new GnRH-PE66 chi-
toxin with antitumor activity on a human colon cancer xenograft meric toxin through specific gonadotropin releasing hormone
in nude mice. J Clin Invest 1992; 90: 405–11. binding sites. J Biol Chem 1997; 272: 11597–603.
256. Arom N, Klimpel KR, Singh Y, Leppla SH. Fusions of anthrax 272. Zimmermann S, Wels W, Froesch BA, Gerstmayer B, Stahel RA,
toxin lethal factor to the ADP-ribosylation domain of Pseudomonas Zangemeister-Wittke U. A novel immunotoxin recognizing the
exotoxin A are potent cytotoxins which are translocated to the epithelial glycoprotein-2 has potent antitumoral activity on che-
cytosol of mammalian cells. J Biol Chem 1992; 267: 15542–8. motherapy-resistant lung cancer. Cancer Immumol Immunother
257. Prior TI, Helman LJ, FitzGerald DJ, Pastan I. Cytotoxic activity 1997; 44: 1–9.
of a recombinant fusion protein between insulin-like growth fac- 273. Fishman A, Lorberbaum-Galski H. Targeted elimination of cells
tor I and Pseudomonas exotoxin. Cancer Res 1991; 51: 174–80. expressing the high-affinity receptor for IgE (Fc) epsilon RI by a
258. Pastan IH, Archer GE, McLendon RE et al. Intrathecal adminis- Pseudomonas exotoxin-based chimeric protein. Eur J Immunol
tration of single-chain immunotoxin, LMB-7 [B3(Fv)-PE38], 1997; 27: 486–94.
produces cures of carcinomatous meningitis in a rat model. Proc 274. Mori T, Shoemaker RH, McMahon JR Gulakowski RJ, Gustafson
Natl Acad Sci USA 1995; 92: 2765–9. KR, Boyd MR. Construction and enhanced cytotoxicity of a
259. Hall WA, Myklebust A, Godal A, Nesland JM, Fodstad O. In vivo [cyanovirin-N]-[Pseudomonas exotoxin] conjugate against
efficacy of intrathecal transferrin Pseudmonas exotoxin A immu- human immumodeficiency virus-infected cells. Biochem Biophys
notoxin against LOX melanoma. Neurosurgery 1994; 34: 649–55. Res Common 1997; 239: 884–8.
260. Kreitman RJ, Chang CN, Hudson DV, Queen C, Bailon P, Pastan I. 275. Nishida K, Seto M, Takahashi T et al. In vitro effects of a recombi-
Anti Tac(Fab)-PE40, a recombinant double-chain immunotoxin nant toxin, mSCF-PE40, targeting c-kit receptors ectopically
which kills interleukin-2-receptor-bearing cells and induces com- expressed in small cell lung cancers. Cancer Lett 1997; 113: 153–8.
plete remission in an in vivo tumor model. Int J Cancer 1994; 57: 276. Chowdhury PS, Viner JL, Beers R, Pastan I. Isolation by a high-
856–64. affinity stable single-chain Fv specific for mesothelin from DNA-
261. Leamon CP Pastan I, Low PS. Cytotoxicity of folate Pseudomonas immunized mice by phage display and construction of a recombinant
exotoxin conjugates towards tumor cells. Contribution of translo- immunotoxin with anti-tumor activity. Proc Natl Acad Sci USA
cation domain. J Biol Chem 1993; 268:,4847–54. 1998; 95: 669–74.
262. Lorimer IA, Keppler-Hafkemeyer A, Beers RA, Pegram CN, 277. Siegall CB, Bacus SS, Cohen BD et al. HER4 expression cor-
Bigner DD, Pastan I. Recombinant immunotoxins specific for a relates with cytotoxicity directed by a heregulin-toxin fusion
mutant epidermal growth factor receptor: targeting with a single protein. J Biol Chem 1995; 270: 7625–30.
chain antibody variable domain isolated by phage display. Proc 278. Haggerty HG, Warner WA, Comereski CR et al. BR96sFvPE40
Natl Acad Sci USA 1996; 93: 14815–20. immunotoxin: nonclinical safety assessment. Toxicol Pathol
263. Klimka A, Barth S, Drillich S et al. A deletion mutant of 1999; 27: 87–94.
Pseudomonas exotoxin-A fused to recombinant human inter- 279. Kreitman RJ, Wilson WH, White JD et al. Phase I trial of recom-
leukin-9 (rhIL-9-ETA’) shows specific cytotoxicity against binant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) in patients with
IL-9-receptor-expressing cell lines. Cytokines Mol Ther 1996; hematologic malignancies. J Clin Oncol 2000; 18: 1622–36.
2: 139–46. 280. Rozemuller H, Chowdhury PS, Pastan I, Kreitman RJ. Isolation
264. Baldwin RL, Kobrin MS, Fran T, Pastan I, Korc M. Cytotoxic of new anti-CD30 scFvs from DNA-immunized mice by phage
effects of TGF-alpha Pseudomonas exotoxin A fusion protein in display and biologic activity of recombinant immunotoxins pro-
human pancreatic carcinoma cells. Pancreas 1996; 13: 16–21. duced by fusion with truncated Pseudomonas exotoxin. Int J
265. Reiter Y, Wright AF, Tonge DW, Pastan I. Recombinant single- Cancer 2001; 92: 861–70.
chain and disulfide-stabilized Fv-immumotoxins that cause com- 281. Wang L, Liu B, Schmidt M, Lu Y, Wets W, Fan Z. Antitumor
plete regression of a human colon cancer xenograft in nude mice. effect of an HER2-specific antibody toxin fusion protein on
Int J Cancer 1996; 67: 113–23. human prostate cancer cells. Prostate 2001; 47: 21–8.
Arthur E. Frankel et al. 439

282. Bruhl H, Cihak J, Stangassinger M, Schlondorff D, Mack M. heregulin receptors and antitumor activity in vivo. Clin Cancer
Depletion of CCR5-expressing cells with bispecific antibodies Res 1998; 4: 993–1004.
and chemokine toxins: a new strategy in the treatment of chronic 300. Mixan B, Cohen BD, Bacus SS, Fell HP, Siegall CB. Betacellulin
inflammatory diseases and HIV. J Immunol 2001; 166: 2420–6. Pseudomonas toxin fusion proteins bind but are not cytotoxic to
283. Engebraaten O, Sivam G, Juell S, Fodstad O. Systemic immuno- cells expressing HER4; correlation of EGFR for cytotoxic activ-
toxin treatment inhibits formation of human breast cancer metastasis ity. Oncogene 1998; 16: 1209–15.
and tumor growth in nude rats. Int J Cancer 2000; 88: 970–6. 301. Brenner T, Steinberger I, Soffer D, Beraud E, Ben-Nun A,
284. Hassan R, Viner JL, Wang QC, Margulies I, Kreitman RJ, Pastan I. Lorberboum-Galski H. A novel antigen toxin chimeric protein:
Anti-tumor activity of Kl-LysPE38QQR, an immunotoxin targeting myelin basic protein Pseudomonas exotoxin (MBP-PE40) for
mesothelin, a cell-surface antigen overexpressed in ovarian cancer treatment of experimental autaimmune encephalomyelitis.
and malignant mesothelioma. J Immunother 2000; 23: 473–9. Immunol Lett 1999; 68: 403–10.
285. Barth S, Huhn M, Matthew B et al. Recombinant anti-CD25 302. Gregg EO, Bridges SH, Youle RJ et al. Whole ricin and recombi-
immunotoxin RFT5(SCFV)-ETA’ demonstrates successful elimi- nant ricin A chain idiotype-specific immunotoxins for therapy of
nation of disseminated human Hodgkin lymphoma in SCID mice. the guinea pig L2C B cell leukemia. J Immunol 1987; 138:
Int J Cancer 2000; 86: 718–24. 4502–8.
286. Liu S, Netzel-Arnett S, Birkedal-Hansen H, Leppla SH. Tumor 303. Boltansky H, Slater J, Youle R, Isersky C, Kalmar M. IgE-
cell-selective cytotoxicity of matrix metalloproteinase-activated immunotoxins. II. IgE-ricin A-chain. Immunopharmacology
anthrax toxin. Cancer Res 2000; 60: 6061–7. 1987; 14: 47–62.
287. Shinohara H, Fan D, Ozawa S et al. Site-specific expression of 304. Levin IN, Griffin TW, Childs LR, Davis S, Haagensen DE.
transferrin receptor by human colon cancer cells directly corre- Comparison of multiple anti-CEA immunotoxins active against
lates with eradication by anti-transferrin recombinant immuno- human adenocarcinoma cells. Cancer Immunol Immunother
toxin. Int J Oncol 2000; 17: 643–51. 1987; 24: 202–6.
288. Kreitman RJ, Margulies I, Steder-Stevenson M, Wang QC, 305. Rennie DP, McGregor AM, Wright J, Weetman AP, Hall R,
FitzGerald DJ, Pastan I. Cytotoxic activity of disulfide stabilized Thorpe P. An immunotoxin of ricin A chain conjugated to thyro-
recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) toward globulin selectively suppresses the antithyroglobulin autoanti-
fresh malignant cells from patients with B cell leukemias. Clin body response. Lancet 1983; 2: 1338–40.
Cancer Res 2000; 6: 1476–87. 306. Wiels J, Junqua S, Dujardin P, Le Pecq JR Tursz T. Properties of
289. Proby CM, Ota T, Suzuki H et al. Development of chimeric immunotoxins against a glycolipid antigen associated with
molecules for recognition and targeting of antigen-specific B Burkitt’s lymphoma. Cancer Res 1984; 44: 129–33.
cells in pemphigus vulgaris. Br J Dermatol 2000; 142: 321–30. 307. Swam G, Pearson JW, Bohn W, Oldham RK, Sadoff JC, Morgan
290. Bera TK, Viner J, Brinkmanm E, Pastan I. Pharmacokinetics and AC. Immunotoxins to a human melanoma-associated antigen:
antitumor activity of a bivalent disulfide-stabilized Fv immuno- comparison of geloninwith ricin and other A chain conjugates.
toxin with improved antigen binding to erbB2. Cancer Res 1999; Cancer Res 1987; 47: 3169–73.
59: 4018–22. 308. Street NE, Fulton RJ, Sanders VM, Vitetta ES. Inhibition of
291. Schmidt M, Maurer-Gebhard M, Groner B, Kohler G, Brochmann- helper function of murine T cells with Fab’-anti-L3T4 ricin A
Santas G, Wels W. Suppression of metastasis formation by a chain immunotoxin. J Immunol 1987; 139: 1734–8.
recombinant single chain antibody toxin targeted to full-length and 309. Boltansky H, Dyer J, Esworthy S, Kalmar M. IgE-immunotoxins.
oncogenic variant EGF receptors. Oncogene 1999; 18: 1711–21. I. IgE-intact ricin. Immunopharmacology 1987; 14: 35–45.
292. Skrepnik N, Zieske AW, Bravo JC, Gillespie AT, Hunt JD. 310. Thiesen HJ, Juhl H, Arndt R. Selective killing of human bladder
Recombinant oncotoxin AR209 (anti-P185 erbB-2) diminishes cancer cells by combined treatment with A and B chain ricin anti-
human prostate carcinoma xenografts. J Urol 1999; 161: 984–9. body conjugates. Cancer Research 1987; 47: 419–23.
293. Ben-Yehudah A, Yarkoni S, Nechushtan A, Belostotsky R, 311. Morgan AC, Bordonaro J, Pearson JW, Sivam G. Immunotoxins
Lorberboum-Galski H. Linker-based GnRH-PE chimeric proteins to a human melanoma-associated antigen: resistance to poke-
inhibit cancer growth in nude mice. Med Oncol 1999; 16: 38–45. weed antiviral protein conjugates in vitro. J Natl Cancer Inst
294. Klimka A, Barth S, Matthey B et al. An anti-CD30 single chain 1987; 78: 1101–6.
Fv selected by phage display and fused to Pseudomonas exotoxin 312. Uckun FM, Azemove SM, Myers DE, Vallera DA. AntiCD2(T,
A (Ki-4(scFv)-ETA) is a potent immunotoxin against a Hodgkin- p50) intact ricin immunotoxins for GVHD-prophylaxis in alloge-
derived cell line. Br J Cancer 1999; 80: 1214–22. neic bone marrow transplantation. Leuk Res 1986; 10: 145–53.
295. Seetharam S, Nodzenski E, Beckett MA et al. Modulation of apoptotic 313. Roth JA, Ames RS, Byers V, Lee HM, Scannon PJ. Monoclonal
response of a radiation-resistant human carcinoma by Pseudomonas antibody 45–2D9 conjugated to the A chain of ricin is specifically
exotoxin chimeric protein. Cancer Res 1998; 58: 3215–20. toxic to c-Ha-ras-transfected NIH 3T3 cells expressing gp74. J
296. Akamatsu Y, Murphy JC, Nolan KF et al. A single-chain immuno- Immunol 1986; 136: 2305–10.
toxin against carcinoembryonic antigen that suppresses growth of 314. Kronke M, Schlick E, Waldmann TA, Vitetta ES, Greene WC.
colorectal carcinoma cells. Clin Cancer Res 1998; 4: 2825–32. Selective killing of human T-lymphotropic virus-infected leukemic
297. Bera TK, Pastan I. Comparison of recombinant immunotoxins T-cells by monoclonal anti-interleukin 2 receptor antibody ricin
against LeY antigen expressing tumor cells: influence of affinity, A chain conjugates: potentiation by ammonium chloride and
size, and stability. Bioconj Chem 1998; 9: 736–43. monensin. Cancer Res 1986; 46: 3295–8.
298. Essand M, Pastan I. Anti-prostate immunotoxins: cytotoxicity of 315. Thorpe PE, Brown AN, Bremner JA, Foxwell BM, Stirpe F. An
E4 antibody Pseudomonas exotoxin constructs. Intl J Cancer immunotoxin composed of monoclonal anti-Thy 1.1 antibody
1998; 77: 123–7. and a ribosome-inactivating protein from Saponaria officinalis:
299. Yang D, Krum CT, Payner J et al. Recombinant heregulin potent antitumor effects in vitro and in vivo. J Natl Cancer Inst
Pseudomonas exotoxin fusion proteins: interactions with the 1985; 75: 151–9.
440 Immunotoxins

316. Bjorn MJ, Ring D, Frankel AE. Evaluation of monoclonal anti- 334. Ettenson D, Sheldon K, Marks A, Houston LL, Baumal R.
bodies for the development of breast cancer immunotoxins. Comparison of growth inhibition of a human ovarian adenocarci-
Cancer Res 1985; 45: 1214–21. noma cell line by free monoclonal antibodies and their corre-
317. Preijers FW, de Witte T, Wessels JM et al. Autologous transplan- sponding antibody-recombinant ricin A chain immunotoxins.
tation of bone marrow purged in vitro with antiCD7-(WT1-)ricin Anticancer Res 1988; 8: 833–8.
A immunotoxin in T-cell lymphoblastic leukemia and lymphoma. 335. Tazzari PL, Barbieri L, Gobbi M et al. An immunotoxin contain-
Blood 1989; 74: 1152–8. ing a rat IgM monoclonal antibody (Campath 1) and saporin 6:
318. Till MA, Zolla-Pazner S, Gorny MK, Patton JS, Uhr JW, Vitetta ES. effect on T lymphocytes and hematopoietic cells. Cancer
Human immunodeficiency virus-infected T cells and monocytes are Immunol Immunother 1988; 26: 231–6.
killed by monoclonal human anti-gp41 antibodies coupled to ricin A 336. Matsushita S, Koito A, Maeda Y, Hattori T, Takatsuki K. Selective
chain. Proc Nat1 Acad Sci USA 1989; 86: 1987–91. killing of HIV-infected cells by anti-gpl20 immunotoxins. AIDS
319. Byers VS, Rodvien R, Grant K et al. Phase I study of monoclonal Res Human Retroviruses 1990; 6: 193–203.
antibody ricin A chain immunotoxin XomaZyme-791 in patients 337. Ghetie V, Till MA, Uhr JW, Vitetta ES. Large scale preparation of
with metastatic colon cancer. Cancer Res 1989; 49: 6153–60. an immunoconjugate constructed with human recombinant CD4
320. Dinota A, Barbieri L, Gobbi M et al. An immunotoxin containing and deglycosylated ricin A chain. J Immunol Methods 1990; 126:
momordin suitable for bone marrow purging in multiple myeloma 135–41.
patients. Br J Cancer 1989; 60: 315 19. 338. Engert A, Burrows F, Jung W et al. Evaluation of ricin A chain-
321. Barbieri L, Dinota A, Gobbi M et al. Immunotoxins containing containing immunotoxins directed against the CD30 antigen as
saporin 6 and monoclonal antibodies recognizing plasma cell- potential reagents for the treatment of Hodgkin’s disease. Cancer
associated antigens: effects on target cells and on normal myeloid Res 1990; 50: 84–8.
precursors (CFU-GM). Eur J Haematol 1989; 42: 238–45. 339. Schmidberger, H, King, L, Lasky, LC, Vallera, DA. Antitumor
322. Conde FP, Orlandi R, Canavan S et al. The Aspergillus toxin activity of L6 ricin immunotoxin against the H2981-T3 lung
restrictocin is a suitable cytotoxic agent for generation of immu- adenocarcinoma cell line in vitro and in vivo. Cancer Res 1990;
noconjugates with monoclonal antibodies directed against human 50: 3249–56.
carcinoma cells. Eur J Biochem 1989; 178: 795–802. 340. May, RD, Finkelman, FD, Wheeler, HT, Uhr JW, Vitetta, ES.
323. Avila AD, Mateo de Acosta C, Lage A. A new immunotoxin built Evaluation of ricin A chain-containing immunotoxins directed
by linking a hemolytic toxin to a monoclonal antibody specific against different epitopes on the delta-chain of cell surface-
for immature T lymphocytes. Int J Cancer 1988; 42: 568–71. associated IgD on marine B cells. J Immunol 1990; 144:
324. Lou Y, Seon BK. Marked difference in the in vivo antitumor effi- 3637–42.
cacy between two immunotaxins targeted to different epitopes of 341. Wawrzynczak EJ, Derbyshire EI, Henry RV et al. Selective cyto-
common acute lymphoblastic leukemia antigen (CD 10). toxic effects of a ricin A chain immunotoxin made with the
Mechanisms involved in the differential activities of immunotox- monoclonal antibody SWA11 recognising a human small cell
ins. J Immunol 1990; 145: 1974–82. lung cancer antigen. Br J Cancer 1990; 62: 410–4.
325. Colombatti M, Bisconti M, Dell’Arciprete L, Gerosa MA, 342. Marches R, Mota G, Margineanu M et al. Treatment of murine
Tridente G. Sensitivity of human glioma cells to cytotoxic het- EL4 leukemia in ascitic form with anti-Thy1.2 specific immuno-
eroconjugates. Int J Cancer 1988; 42: 441–8. toxins. Neoplasma 1990; 37: 573–8.
326. Press OW, Martin PJ, Thorpe PE, Vitetta ES. Ricin A-chain con- 343. Wu M, Tang SL, Zang RJ, Yu H. Selective killing of tumor cells in
taining immunotoxins directed against different epitopes on the vitro by an immunotoxin composed of ricin and monoclonal anti-
CD2 molecule differ in their ability to kill normal and malignant body against Ia antigen. Int J Immunopharm 1990; 12: 235–9.
T cells. J Immunol 1988; 141: 4410–7. 344. Santana, JM, Teixeira, AR. Effect of immumotoxins against
327. Manske JM, Buchsbaum DJ, Hanna DE, Vallera DA. Cytotoxic Trypanosoma cruzi. Am J Trop Med Hygiene 1989; 41: 177–82.
effects of anti-CD5 radioimmunotoxins on human tumors in vitro 345. Shumakov VI, Maisjuk IG, Agapov II et al. In vitro efficacy of
and in a nude mouse model. Cancer Res 1988; 48: 7107–14. conjugates of anti-CD45 monoclonal antibodies with plant toxin
328. Derocq JM, Casellas P, Laurent G, Ravel S, Vidal H, Jansen F. A-chains. Transplant Proc 1998; 30: 971–3.
Comparison of the cytotoxic potency of T101 Fab, F(ab)2 and 346. La Russa VF, Griffin JD, Kessler SW et al. Effects of anti-CD33
whole IgG immunotoxins. J Immunol 1988; 141: 2837–43. blocked ricin immumotoxin on the capacity of CD34+ human
329. Preijers FW, Tax WJ, Wessels JM, Capel PJ, De Witte T, Haanen marrow cells to establish in vitro hematopoiesis in long-term
C. Different susceptibilities of normal T cells and T cell lines to marrow cultures. Exp Haematol 1992; 20: 442–8.
immunotoxins. Scand J Immunol 1988; 27: 533–40. 347. Burlet A, Chapleur-Chateau M, Haumont-Pellegri B et al. Long-
330. Kanellos J, MacKenzie IF, Pietersz GA. In vivo studies of whole term reduction of vasopression excretion induced by the central
ricin monoclonal antibody immunoconjugates for the treatment injection of an immunoconjugate (antibody to vasopressin linked
of marine tumours. Immunol Cell Rot 1988; 66: 403–15. to ricin A chain). Neuroscience 1992; 50: 965–73.
331. Izquierdo M, Balboa MA, Figuera A, Lopez-Botet M. Selective T 348. Bolognesi A, Tazzari PL, Olivieri F et al. Evaluation of immumo-
cell subset depletion with anti-CD4 and antiCD8 intact ricin toxins containing single-chain ribosome-inactivating proteins
immunotoxins. Clin Exp Immunol 1988; 74: 300–4. and an anti-CD22 monoclonal antibody (OM124): in vitro and in
332. Shen GL, Li JL, Ghetie MA et al. Evaluation of four CD22 vivo studies. Br J Haematol 1998; 101: 179–88.
antibodies as ricin A chain-containing immunotoxins for the in 349. Wang QC, Ying WB, Xie H, Zhang ZC, Yang ZH, Ling LQ.
vivo therapy of human B-cell leukemias and lymphomas. Int J Triochsanthin monoclonal antibody conjugate specifically cyto-
Cancer 1988; 42: 792–7. toxic to human hepatoma cells in vitro. Cancer Res 1991; 51:
333. Cattel L, Delprino L, Brusa P, Dosio F, Comoglio PM, Prat M. 3353–5.
Comparison of blocked and non-blocked ricin antibody immuno- 350. Derbyshire PJ, Wawrzynczak EJ. Monoclonal antibodies recog-
toxins against human gastric carcinoma and colorectal adenocarci- nizing the cluster 2 antigen associated with human small cell
noma cell lines. Cancer Immunol Immunother 1988; 27: 233–40. lung cancer mediate the toxic effects of ricin A chain in an indirect
Arthur E. Frankel et al. 441

assay of immumotoxin cytotoxicity. Br J Cancer 1991; 369. Duke-Cohan JS, Morimoto C, Schlossman SF. Depletion of the
14(Suppl): 74–7. helper/inducer (memory) T cell subset using a bispecific antibody
351. Zong W, Dai B. Preparation of SOKTI-RTA immunotoxin and its toxin conjugate directed against CD4 and CD29. Transplantation
specific cytotoxic effect on human T lymphocytes. J West China 1993; 56: 1188–96.
Univ Med Sci 1991; 22: 240–4. 370. Faguet GB, Agee JF. Four ricin chain A-based immunotoxins
352. Engert A, Brown A, Thorpe P. Resistance of myeloid leukemia directed against the common chronic lymphocytic leukemia anti-
cell lines to ricin A-chain immumotoxins. Leuk Res 1991; 15: gen: in vitro characterization. Blood 1993; 82: 536–43.
1079–86. 371. Zangemeister-Wittke U, Collinson AR, Fisch I et al. Antitumor
353. Rosenblum MG, Murray JL, Cheung L, Rifkin R, Salmon S, activity of a blocked ricin immunotoxin with specificity against
Bartholomew R. A specific and potent immunotoxin composed of the cluster-5A antigen associated with human small-cell lung
antibody ZME-018 and the plant toxin gelonin. Mol Biother cancer. Int J Cancer 1993; 54: 1028–35.
1991; 3: 6–13. 372. Crews JR, Maier LA, YH et al. A combination of two immuno-
354. Wawrzynczak EJ, Henry RV, Cumber AJ, Parnell GD, Derbyshire toxins exerts synergistic cytodoxic activity against human breast
EJ, Ulbrich N. Biochemical, cytodoxic and pharmacokinetic cancer cell liens. Int J Cancer 1992; 51: 772–9.
properties of an immunotoxin composed of a mouse monoclonal 373. Tazzari PL, Bolognesi A, de Totero D et al. Ber-H2 (antiCD30)-
antibody Fib75 and the ribosome inactivating protein alpha-sarcin saporin immunotoxin: a new tool for the treatment of Hodgkin’s
from Aspergillus giganteus. Eur J Biochem 1991; 196: 203–9. disease and CD30+ lymphoma: in vitro evaluation. Br J Haematol
355. Pincus SH, Cole RL, Hersh EM et al. In vitro efficacy of anti- 1992; 81: 203–11.
HIV immunotoxins targeted by various antibodies to the enve- 374. Paprocka M, Wiedlocha A, Walzel H, Radzikowski C. The activ-
lope protein. J Immunol 1991; 146: 4315–24. ity of two immunodoxins composed of monoclonal antibody
356. Shitara K, Hanai N, Kusano A et al. Application of anti-sialyl Lea MoAb-16 and A-chain of ricin (MoAb-16-RTA) or A-chain of
monoclonal antibody KM231, for immunotherapy of cancer. mistletoe lectin I (MoAb-16-MLIA). Arch Immumol Ther Exp
Anticancer Res 1991; 11: 2003–13. 1992; 40: 223–7.
357. Ito T, Qiu H, Collins JA, Brill AB, Johnson DK, Griffin TW. 375. Derbyshire EJ, Wawrzynczak EJ. An anti-mucin immunotoxin
Preclinical assessments of 90Y-labeled C110 anti-carcinoembry- BrE-3-ricin A chain is potently and selectively toxic to human
onic antigen immumotoxin: a therapeutic immunoconjugate for small-cell lung cancer. Int J Cancer 1992; 52: 624–30.
human colon cancer. Cancer Res 1991; 51: 255–60. 376. Derbyshire EJ, Stahel RA, Wawrzynczak EJ. Potentiation of a
358. Li S, Zhang XY, Chen XT et al. Targeted anti-tumor agents and weakly active ricin A chain immumodoxin recognizing the
their cytotoxicity on gastric cancer cells in vitro. Chinese Med J neural cell adhesion molecule. Clin Exp Immunol 1992; 89:
1990; 103: 376–9. 336–40.
359. Paprocka, M, Wiedlocha, A, Radzikowski, C. Mouse L1210V 377. Zhao C. Preparation of an anti-human immunotoxin against blad-
leukemia as a model to analyse efficiency of leukemic cell elimi- der carcinoma and its in vitro targeting cytotoxicity. Chin J Oncol
nation by immunotoxin, antibody and complement, and cytostatic 1992; 14: 109–11.
agents. In Vivo 1990; 4: 209–13. 378. Shindo T, Ueda H, Makishima F, Suzuki E, Nishimura H.
360. Ebert RE, Spryn LA. Immunotoxin construction with a ribosome- Efficient selection of mu m-mutants from mu m-expressing
inactivating protein from barley. Bioconj Chem 1990; 1: 331–6. myeloma cells by treatment with ricin A-conjugated anti-mu anti-
361. Chiron M, Jaffrezou JP, Carayon P et al. Induction and increase body. Somat Cell Mol Genet 1992; 18: 553–8.
of HLA-DR antigen expression by immune interferon on ML-3 379. Collinson AR, Lambert JM, LiuY et al. Anti-CD6-blocked ricin:
cell line enhances the anti-HLA-DR immunotoxin activity. Clin an anti-pan T-cell immumotoxin. Int J Immunopharm 1994; 16:
Exp Immumol 1990; 82: 214–20. 37–49.
362. Zenner HP. A monoclonal immunotoxin against laryngeal carci- 380. Woo BH, Lee IT, Lee KC. Ricin A immumotoxins of IgG and Fab
noma cells. Otolaryngol Polska 1990; 44: 214–5. of anti-CALLA monoclonal antibody: effect of water soluble
363. Huang YW, Burrows FJ, Vitetta ES. Cytodoxicity of a novel anti- long-chain SPDP on conjugate yield, immunoselectivity and
ICAM-1 immunotoxin on human myeloma cell lines. Hybridoma cytotoxicity. Arch Pharm Res 1994; 17: 452–7.
1993; 12: 661–75. 381. Menzaghi F, Heinrichs SC, Merlo-Pich E, Tilders FJ, Koob GF.
364. Duke-Cohan JS, Morimoto C, Schlossman SF. Targeting of an Involvement of hypothalamic corticotropin-releasing factor neu-
activated T-cell subset using a bispecific antibody toxin conjugate rons in behavioral responses to novelty in rats. Neurosci Lett
directed against CD4 and CD26. Blood 1993; 82: 2224–34. 1994; 168: 139–42.
365. Efferth T, Volm M. Modulation of P-glycoprotein-mediated mul- 382. Engert A, Gottstein C, Winkler U et al. Experimental treatment of
tidrug resistance by monoclonal antibodies, immunotoxins or human Hodgkin’s disease with ricin A-chain immumotoxins.
antisense oligodeoxynucleotides in kidney carcinoma and normal Leuk Lymph 1994; 13: 441–8.
kidney cells. Oncology 1993; 50: 303–8. 383. Zangemeister-Wittke U, Collinson AR, Frosch B, Waibel R,
366. Zangemeister-Wittke U, Lehmann HP, Waibel R, Wawrzynczak Schenker T, Stahel RA. Immumotoxins recognizing a new epitope
EJ, Stahel RA. Action of a CD24-specific deglycosylated ricin on the neural cell adhesion molecule have potent cytotoxic effects
A-chain immumotoxin in conventional and novel models of against small cell lung cancer. Br J Cancer 1994; 69: 32–9.
small-cell-lung-cancer xenograft. Int J Cancer 1993; 53: 521–8. 384. Deonarain MP, Epenetos AA. Targeting phosphodiesterases as a
367. Calvete JA, Newell DR, Charlton CJ, Wright AF. Pharmacokinetic strategy for killing tumor cells. Cell Biophys 1994; 24: 49–57.
studies in mice with ICI D0490, a novel recombinant ricin 385. Burrows FJ, Overholser JP, Thorpe PE. Potent antitumor effects
A-chain immumotoxin. Br J Cancer 1993; 67: 1310–5. of an antitumor endothelial cell immunotoxin in a marine vascu-
368. Tonevitksy AG, Agapov II, Ershova GV, Sarma T, Toptygin AY, lar targeting model. Cell Biophys 1994; 24: 15–25.
Mechetner EB. Cytodoxic effect of ricin A-chain conjugates con- 386. Goldmacher VS, Bourret LA, Levine BA et al. Anti-CD38
taining monoclonal antibodies against human erythroid cells. Int blocked ricin: an immunotoxin for the treatment of multiple
J Immunopharm 1993; 15: 229–35. myeloma. Blood 1994; 84: 301–25.
442 Immunotoxins

387. Tonevitsky AG, Toptygin AI, Agapov II et al. Chimeric toxin of mAb 35 on extraocular muscles of rabbits. Invest Ophthal Vis Sci
the A-subunit of viscumin and the B-subunit of ricin. Molek Biol 2000; 41: 3402–9.
1994; 28: 574–9. 405. Seon BK, Matsuno F, Haruta Y, Kondo M, Barcos M. Long-lasting
388. Usui H, Hakomori S. Evaluation of ricin A chain-containing complete inhibition of human solid tumors in SCID mice by target-
immunotoxins directed against glycolipid and glycoprotein on ing endothelial cells of tumor vasculature with anti-human endo-
mouse lymphoma cells. Acta Med Okayama 1994; 48: 305–9. glin immunotoxin. Clin Cancer Res 1997; 3: 1031–44.
389. Thompson PA, McAtee R, Infante AJ, Currier P, Beninati W, 406. Wang D, Li Q, Hudson W, Berven E, Uckan F, Kersey JH.
Krolick KA. V beta-specific immunotoxin selectively kills ace- Generation and characterization of an anti-CD19 singlechain Fv
tylcholine receptor-reactive T lymphocytes from mice with immunotoxin composed of C-terminal disulfide linked dgRTA.
experimental myasthenia gravis. Int Immunol 1994; 6: 1807–15. Bioconj Chem 1997; 8: 878–84.
390. Waurzyniak B, Schneider EA, Tumer N et al. In vivo toxicity, 407. Zewe M, Rybak SM, Dubel S et al. Cloning and cytotoxicity of a
pharmacokinetics, and antileukemic activity of TXU (antiCD7)- human pancreatic RNase immunofusion. Immunotechnology
pokeweed antiviral protein immunotoxin. Clin Cancer Res 1997; 1997; 3: 127–36.
3: 881–90. 408. Pauza ME, Doumbia SO, Pennell CA. Construction and charac-
391. Zalcberg JR, Pietersz G, Toohey B et al. A phase I/II study of the terization of human CD7-specific single-chain Fv immunotoxins.
intralesional injection of ricin monoclonal antibody conjugates in J Immunol 1997; 158: 3259–69.
patients with hepatic metastases. Eur J Cancer 1994; 304: 1227–31. 409. Yoshida M, Rybak RJ, Choi Y et al. Development of a severe
392. Gottstein C, Schon G, Tawadros S et al. Anti-disialoganglioside combined immunodeficiency (SCID) mouse model consisting of
ricin A-chain immunotoxins show potent antitumor effects in vitro highly disseminated human B-cell leukemia/lymphoma, cure of
and in a disseminated human neuroblastoma severe combined the tumors by systemic administration of immumotoxin, and
immunodeficiency mouse model. Cancer Res 1994; 54: 6186–93. development/application of a clonotype specific polymerase chain
393. Tazzari PL, Bolognesi A, De Totero D et al. Immunotoxins con- reaction-based assay. Cancer Res 1997; 57: 678–85.
taining saporin linked to different CD2 monoclonal antibodies: in 410. Shimizu K, Nagai K, Nakagawa H. An immumotoxin, anti-VIP
vitro evaluation. Br J Haematol 1994; 86: 97–105. antibody ricin A chain conjugate eliminates neurons in the hypo-
394. Kung AH, Cavagnaro JA, Makin A, White MA, Kong KN. thalamic suprachiasmatic nucleus selectively and abolishes the
Toxicologic evaluations of an immumotoxin, H65-RTA. Fund circadian rhythm of water intake. Brain Res Bull 1996; 41:
Appl Toxicol 1995; 26: 75–84. 369–78.
395. Chignola R, Pasti M, Candiani C et al. Escape mechanisms of 411. Barnett BB, Smee DF, Malek SM, Sidwell RW. Selective cyto-
human leukemic cells to long-term immunotoxin treatment in an toxicity of ricin A chain immunotoxins towards murine cytomeg-
in vitro experimental model. Int J Cancer 1995; 61: 535–41. alovirus-infected cells. Antimicrob Agents Chemother 1996; 40:
396. Lustgarten J, WaksT, Eshhar Z. Prolonged inhibition of IgE pro- 470–2.
duction in mice following treatment with an IgE-specific immu- 412. Surolia N, Misquith S. Cell surface receptor directed targeting of
motoxin. Mol Immunol 1996; 33: 245–51. toxin to human malaria parasite, Plasmodium falciparum. FEBS
397. Lazzaro GE, Meyer BF, Willis JL, Erber WN, Herrmann RP, Lett 1996; 396: 57–61.
Davies JM.The synthesis of a peanut agglutinin ricinA chain con- 413. Dore JM, Gras F, Wijdenes J. Expression and activity of a recom-
jugate: potential as an in vitro purging agent for autologous bone binant chimeric protein composed of pokeweed antiviral protein
marrow in multiple myeloma. Exp Hematol 1995; 23: 1347–52. and of human interleukin-2. FEBS Lett 1997; 402: 50–2.
398. Schnell R, Linnartz C, Katouzi AA et al. Development of new ricin 414. Scott CF, Goldmacher VS, Lambert JM, Jackson JV, McIntyre
A-chain immumotoxins with potent anti-tumor effects against GD. An immunotoxin composed of a monoclonal antitransferrin
human Hodkgin cells in vitro and disseminated Hodgkin tumor in receptor antibody linked by a disulfide bond to the ribosome-
SCID mice using high-affinity monoclonal antibodies directed inactivating protein gelonin: potent in vitro and in vivo effects
against the CD30 antigen. Int J Cancer 1995; 63: 238–44. against human tumors. J Natl Cancer Inst 1987; 79: 1163–72.
399. Conry RM, Khazaeli MB, Saleh MN et al. Phase I trial of an anti- 415. Longo DL, Duffey PL, Gribben JG et al. Combination chemo-
CD19 deglycosylated ricin A chain immunotoxin in non-Hodgkin’s therapy followed by an immunotoxin (anti-B4-blocked ricin) in
lymphoma: effect of an intensive schedule of administration. J patients with indolent lymphoma: results of a phase II study.
Immumother 1995; 18: 231–41. Cancer J Sci Am 2000; 6: 146–50.
400. Lynch TJ, Lambert TM, Coral F et al. Immunotoxin therapy of 416. Tazzari PL, de Totern D, Bolognesi A et al. An Epstein Barr
small-cell lung cancer: a phase I study of N901-blocked ricin. J virus-infected lymphoblastoid cell line (3430B) that grows in
Clin Oncol 1997; 15: 723–34. SCID-mice with the morphologic features of a CD30+ anaplastic
401. Vallera DA, Taylor PA, Panoskaltsis-Mortari A, Blazar BR. large cell lymphoma, and is sensitive to anti-CD30 immunotox-
Therapy for ongoing graft-versus-host disease induced across the ins. Haematologica 1999; 84: 988–95.
major or minor histocompatibility barrier in mice with anti-CD3 417. Sharma S, Podder SK, Karande AA. Comparative studies on
F(ab’)2-ricin toxin A chain immunotoxin. Blood 1995; 86: kinetics of inhibition of protein synthesis in intact cells by ricin
4367–75. and a conjugate of ricin B-chain with momordin. Mol Cell
402. Chignola R, Anselmi C, Dalla Serra M et al. Self-potentiation of Biochem 1999; 200: 133–41.
ligand toxin conjugates containing ricin A chain fused with viral 418. Matsuno F, Haratu Y, Kondo M, Tsai H, Barcos M, Seon BK.
structures. J Biol Chem 1995; 270: 23345–51. Induction of lasting complete regression of preformed distinct
403. Shimizu K, Park KC, Tamura K et al. Internalization with high solid tumors by targeting the tumor vasculature using two new
targeting potential of mouse monoclonal antibody ONS-M21 rec- anti-endoglin monoclonal antibodies. Clin Cancer Res 1999; 5:
ognizing human malignant glioma antigen. Cancer Lett 1998; 371–82.
127: 171–6. 419. Ohtomo T, Kawata H, Sekimori Y et al. A humanized single-
404. Christiansen SP, Sandnas A, Prill R, Youle RJ, McLoon LK. chain Fv fragment with high targeting potential against human
Acute effects of the skeletal muscle-specific immunotoxin ricin- malignant gliomas. Anticancer Res 1998; 18: 4311–5.
Arthur E. Frankel et al. 443

420. Clinchy B, Vitetta ES. The use of an anti-CD3 immunotaxin to mediated function using CRF coupled to saporin. Endocrinology
prevent the development of lymphoproliferative disease in SCID/ 2000; 141: 498–504.
PBL mice. J Immunol Meth 1998; 218: 141–53. 437. Chandler LA, Sosnowski BA, McDonald JR et al. Targeting
421. Ippoliti R, Ginobbi P, Lendaro F et al. The effect of monensin and tumor cells via EGF receptors: selective toxicity of an HBEGF-
chlunquine on the endocytosis and toxicity of chimeric toxins. toxin fusion protein. Int J Cancer 1998; 78: 106–11.
Cell Mol Life Sci 1998; 54: 866–75. 438. Ippoliti R, Lendaro E, Bellelli A et al. A saporin-insulin conju-
422. Boyer CM, Pusztai L, Wiener JR et al. Relative cytodoxic activity gate: synthesis and biochemical characterization. Nat Toxins
of immunotoxins reactive with different epitopes on the extracel- 1996; 4: 156–62.
lular domain of the c-erbB-2 (HER-2/neu) gene product p185. Int 439. Vooijs WC, Post J, Wijdenes J et al. Efficacy and toxicity of plasma
J Cancer 1999; 82: 525–31. cell reactive monoclonal antibodies B-B2 and B-B4 and their
423. Weinberg AD, Bourdette DN, Sullivan TJ et al. Selective deple- immunotoxins. Cancer Immunol Immunother 1996; 42: 319–28.
tion of myelin-reactive T cells with the anti-OX-40 antibody 440. Dosio F, Brusa P, Crosasso P, Fruttero C, Cattel L, Bolognesi A.
ameliorates autoimmume encephalomyelitis. Nature Med 1996; Synthesis of different immunotaxins composed by ribosome
2: 183–98. inactivating proteins non-covalendy bound to monoclonal anti-
424. Casellas P, Brown JP, Gros O et al. Human melanoma cells can be body. Farmaco 1996; 51: 477–82.
killed in vitro by an immunotoxin specific for melanoma-associ- 441. Flavell DJ, Flavell SU, Boehm DA et al. Preclinical studies with
ated antigen p97. Int J Cancer 1982; 30: 437–43. the anti-CD19 saporin immunotoxin BU12-saporin for the treat-
425. Masui H, Kamrath H, Apell G, Houston LL, Mendelsohn J. ment of human B cell tumours. Br J Cancer 1995; 72: 1373–9.
Cytodoxicity against human tumor cells mediated by the conjugate 442. Cazzola M, Bergamaschi G, Dezza L et al. Cytotoxic activity of
of anti-epidermal growth factor receptor monoclonal antibody to an anti-transferrin receptor immunotoxin on normal and leukemic
recombinant ricin A chain. Cancer Res 1989; 49: 3482–8. human hematopoietic progenitors. Cancer Res 1991; 51: 536–41.
426. Rybak SM, Saxena SK, Ackerman EJ, Youle RJ. Cytodoxic 443. Blank M, Manosroi J, Tamer Y et al. Suppression of experimental
potential of ribonuclease and ribonuclease hybrid proteins. J Biol systemic erythematosus (SLE) with specific anti-idiotypic anti-
Chem 1991; 266: 21202–7. body saporin conjugate. Clin Exp Immunol 1994; 98: 434–41.
427. Frankel AE, Laver JH, Willingham MC, Burns LI, Kersey JH, 444. Tecce R, Digiesi G, Savarese A, Trizio D, Natali PG. Characterization
Vallera DA. Therapy of patients with T-cell lymphomas and leu- of cytotoxic activity of saporin antigp185/HER-2 immunotoxins.
kemias using an anti-CD7 monoclonal antibody ricin A chain Int J Cancer 1993; 55: 122–7.
immunotoxin. Leuk Lymph 1997; 26: 287–98. 445. Bonardi MA, French RR, Amlot P, Gromo G, Modena D, Glennie
428. Terenzi A, Bolognesi A, Pasqualucci L et al. Anti-CD30 (BERH2) MJ. Delivery of saporin to human B-cell lymphoma using bispe-
immunotoxins containing the type-1 ribosome inactivating pro- cific antibody: targeting via CD22 but not CD19, CD37, or
teins momordin and PAP-S (pokeweed antiviral protein from immunoglobulin results in efficient killing. Cancer Res 1993; 53:
seeds) display powerful antitumour activity against CD30+ tumour 3015–21.
cells in vitro and in SCID mice. Br J Haematol 1996; 92: 872–9. 446. Poccia F, Piselli P, Di Cesare S et al. Recognition and killing of
429. Battalli MG, Polito L, Bolognesi A, Lafleur L, Fradet Y, Stirpe F. tumour cells expressing heat shock protein 65 kD with immuno-
Toxicity of ribosome-inactivating proteins containing immuno- toxins containing saporin. Br J Cancer 1992; 66: 427–32.
toxins to a human bladder carcinoma cell line. Int J Cancer 1996; 447. Tazzari PL, Bolognesi A, De Totero D et al. B-B10 (antiCD25)-
65: 485–90. saporin immunotoxin a possible tool in graft versus host disease
430. Myers DE, Uckun FM. An anti-CD72 immunotoxin against ther- treatment. Transplant 1992; 54: 351–6.
apy-refractory B-lineage acute lymphoblastic leukemia. Leuk 448. Tecce R, Nicotra MR, Fraioli R, Cuomo M, Trizio D, Natali PG.
Lymph 1995; 18: 119–22. Saporin 6 conjugated to monoclonal antibody selectively kills
431. Smith CV, Sablinski T, Arn JS et al. In vivo treatment with mono- human melanoma cells. Melanoma Res 1991; 1: 115–23.
clonal antibodies directed against CD4 and CD8 antigens in min- 449. Tecce R, Fraioli R, De Frabritiis P et al. Production and charac-
iature swine. J Immunother 1994; 16: 105–14. terization of two immunotoxins specific for M5b ANLL leukaemia.
432. Ek O, Gaynon P, Zeren T, Chelstrom LM, Myers DE, Uckun FM. Int J Cancer 1991; 49: 310–6.
Treatment of human B-cell precursor leukemia in SCID mice by 450. Dinota A, Tazzari PL, Michieli M et al. In vitro bone marrow
using a combination of the anti-CD19 immunotoxin B43-PAP purging of muftidrug resistant cells with a mouse monoclonal
with the standard chemotherapeutic drugs vincristine, methyl- antibody directed against M, 170,000 glycoprotein and a
prednisolone, and L-asparaginase. Leuk Lymph 1998; 31: saporin-conjugated anti-mouse antibody. Cancer Res 1990; 50:
143–9. 4291–4.
433. Kwok KH, Law KB, Wong RN, Yung KK. Immunolesioning of 451. Siena S, Bragni M, Formosa A al. Immunotoxin mediated killing
nerve growth factor p75 receptor-containing neurons in the rat brain of clonable T-lymphocytes infiltrating an irreversibly rejected
by a novel immunotoxin: anti-p75-anti-mouse IgG-trichosanthin human renal allograft. J Biol Resp Reg Homeostatic Agents
conjugates. Brain Res 1999; 846: 154–63. 1989; 3: 84–8.
434. Flavell DJ, Boehm DA, Noss A, Warnes SL, Flavell SU. Therapy 452. Bragni M, Siena S, Formosa A et al. B-cell restricted saporin
of human T-cell acute lymphoblastic leukaemia with a combination immunotoxins: activity against b-cell lines and chronic lympho-
of anti-CD7 and anti-CD38 saporin immunotoxins is significantly cytic leukemia cells. Blood 1989; 73: 753–62.
better than therapy with each individual immunotoxin. Br J 453. Barbieri L, Bolognesi A, Dinota A et al. Selective killing of
Cancer 2001; 84: 571–8. CD4+ and CD8+ cells with immunotoxins containing saporin.
435. Davol P, Beitz JG, Mohle, M et al. Saporin toxins directed to basic Scand J Immunol 1989; 30: 369–72.
fibroblast growth factor receptors effectively target human ovarian 454. Barbieri L, Dinota A, Gobbi M et al. Immuntoxins containing
teratocarcinoma in an animal model. Cancer 1995; 76: 79–85. saporin 6 and monoclonal antibodies recognizing plasma cell-
436. Maciejewski-Lenoir D, Heinrichs SC, Liu XJ et al. Selective associated antigens: effects on target cells and on normal myeloid
impairment of corticotropin-releasing factor 1 (CRF1) receptor- precursors (CFU-GM). Eur J Haematol 1989; 42: 238–45.
444 Immunotoxins

455. Bragni M, Lappi DA, Siena, S et al. Activity of a monoclonal 472. Hirota N, Ueda M, Ozawa S, Abe O, Shimizu N. Suppression of
antibody saporin 6 conjugate against B-lymphoma cells. J Natl an epidermal growth factor receptor-hyperproducing tumor by an
Cancer Inst 1988; 80: 511–7. immunotoxin conjugate of gelonin and a monoclonal anti-epidermal
456. Glennie MJ, McBride HM, Stirpe F, Thorpe PE, Worth AT, growth factor receptor antibody. Cancer Res 1989; 49: 7106–9.
Stevenson GT. Emergence of immunoglobulin variants following 473. Reimann KA, Goldmacher VS, Lambert JM et al. In vivo admin-
treatment of a B cell leukemia with an immunotoxin composed of istration of lymphocyte-specific monoclonal antibodies in nonhuman
anti-idiotypic antibody and saporin. J Exp Med 1987; 166: 43–62. primates. IV. Cytotoxic effect of an antiT11-gelonin immuno-
457. Singh V, Curtiss R. Hormonotoxins: the role of positive charge of toxin. J Clin Invest 1988; 82: 129–38.
lysine residue on the immunological, biological, and cytotoxic 474. Tedder TF, Goldmacher VS, Lambert JM, Schlossman SF.
properties of ovine lutropin-S-S-gelonin conjugates. Mol Cell Epstein Barr virus binding induces internalization of the C3d
Biochem 1994; 130: 91–101. receptor: a novel immunotoxin delivery system. J Immunol 1986;
458. Scott CF, Goldmacher VS, Lambert JM et al. The antileukemic 137: 1387–91.
efficacy of an immunotoxin composed of a monoclonal anti- 475. Colombatti M, Bron C. Sensitivity of target cells to immunotox-
Thy-1 antibody disulfide linked to the ribosome inactivating pro- ins: possible role of cell-surface antigens. Immunology 1985; 55:
tein gelonin. Cancer Immunol Immunother 1987; 25: 31–40. 331–8.
459. Wenning LA, Murphy RM. Coupled cellular trafficking and dif- 476. Psarras K, Ueda M, Tanabe M et al. Targeting activated lympho-
fusional limitations in delivery of immunotoxins to multicell cytes with an entirely human immunotoxin analogue: human
tumor spheroids. Biotech Bioeng 1999; 62: 562–75. pancreatic RNasel-human IL-2 fusion. Cytokine 2000; 12:
460. Pagliaro LC, Liu B, Munker R et al. Humanized M195 monoclo- 786–90.
nal antibody conjugated to recombinant gelonin: an anti-CD33 477. Yoon JM, Han SH, Kown OB Kim SH, Park MH, Kim BK.
immunotoxin with antileukemic activity. Clin Cancer Res 1998; Cloning and cytotoxicity of fusion proteins of EGF and angio-
4: 1971–6. genin. Life Sci 1999; 64: 1435–45.
461. Arpicco S, Dosio F, Brusa P, Crosasso P, Cattel L. New coupling 478. Frankel AE, Ramage J, Latimer A et al. High-level expression and
reagents for the preparation of disulfide-linked conjugates with purification of the recombinant diphtheria fusion toxin DTGM for
increased stability. Bioconj Chem 1997; 8: 327–37. phase I clinical trials. Prot Expres Purif 1999; 16: 190–201.
462. Smee DF, Sidwell RW, Barnett BB. Combination of antiviral immu- 479. Hotchkiss CE, Hall PD, Cline JM et al. Toxicology and pharma-
notoxin and ganciclovir or cidofovir for the treatment of murine cokinetics of DTGM, a fusion toxin consisting of a truncated
cytomegalovirus infections. Antiviral Res 1996; 32: 165–71. diphtheria toxin (DT388) linked to human granulocyte mac-
463. O’Boyle KP, Colletti D, Mazurek C et al. Potentiation of antipro- rophage colony-stimulating factor, in cynomolgas monkeys.
liferative effects of monoclonal antibody Lym-1 and immuno- Toxicol Appl Pharm 1999; 158: 152–60.
conjugate Lym-1-gelonin on human Burkitt’s lymphoma cells 480. Feuring-Buske M, Buske C, Unterhalt M, Hiddemann W. Recent
with gamma-interferon and tumor necrosis factor. J Immunother advances in antigen-targeted therapy in non-Hodkgin’s lym-
1995; 18: 221–30. phoma. Ann Hematol 2000; 79: 167–74.
464. Barnett BB, Smee DF, Malek SM, Sidwell RW. Selective cyto- 481. Kreitman RJ, Wilson WH, Bergeron K et al. Efficacy of the anti-
toxicity towards cytomegalovirus-infected cells by immunotox- CD22 recombinant immunotoxin BL22 in chemotherapy-resistant
ins consisting of gelonin linked to anti-cytomegalovirus antibody. hairy-cell leukemia. N Engl J Med 2001; 345: 241–7.
Antiviral Res 1995; 28: 93–100. 482. Kreitman RJ, Wilson WH, Robbins D et al. Responses in refrac-
465. Misquith S, Surolia A. In vivo treatment of Heymann’s nephritis using tory hairy cell leukemia to a recombinant immunotoxin. Blood
a cytotoxic protein toxin conjugate. FEBS Lett 1995; 373: 151–4. 1999; 94: 3340–8.
466. Masuda K, Takahashi K, Nagata S, Hirano K, Takagishi Y. 483. Laske DW, Youle RJ, Oldfield EH. Tumor regression with
Immunotoxins composed of monoclonal antibody to alphafeto- regional distribution of the targeted toxin Tf-CRM107 in patients
protein and gelonin as a potent hepatoma-targeted drug delivery with malignant brain tumors. Nature Med 1997; 3: 1362–8.
system. J Drug Targeting 1994; 2: 323–31. 484. Weaver M, Laske DW. Transferrin receptor ligand-targeted toxin
467. Rosenblum MG, Zuckerman JE, Marks JW, Rotbein J, Allen conjugate (Tf-CRM107) for therapy of malignant gliomas. J
WR. A gelonin-containing immunotoxin directed against human Neurooncol 2003; 65: 3–13.
breast carcinoma. Mol Biother 1992; 4: 122–9. 485. LeMaistre CF, Saleh MN, Kuzel TM et al. Phase I trial of a ligand
468. Barnett BB, Burns NJ, Park KJ, Dawson MI, Kende M, Sidwell fusion-protein (DAB389IL-2) in lymphomas expressing the
RW. Antiviral immunotoxins: antibody-mediated delivery of receptor for interleukin-2. Blood 1998; 91: 399–405.
gelonin inhibits Pichinde virus replication in vitro. Antiviral Res 486. Olsen E, Duvic M, Frankel A et al. Pivotal phase III trial of two
1991; 15: 125–38. dose levels of denileukin diftitox for the treatment of cutaneous
469. Harris P, Reed E, King DW, Suciu-Foca N. In vitro studies of the T-cell lymphoma. J Clin Oncol 2001; 19: 376–88.
effect of Mab NDA4 linked to toxin on the proliferation of a 487. Foss F, Demierre MF, DiVenuti G. A phase-1 trial of bexarotene
human EBV-transformed lymphoblastoid B cell line and of gibbon and denileukin diftitox in patients with relapsed or refractory
MLA leukemia cell line. Cell Immunol 1991; 134: 85–95. cutaneous T-cell lymphoma. Blood 2005; 106: 454–7.
470. Mujoo K, Reisfeld RA, Cheung L, Rosenblum MG. A potent and 488. Duvic M, Cather J, Maize J, Frankel AE. DAB389IL2 diphtheria
specific immunotoxin for tumor cells expressing disialoganglio- fusion toxin produces clinical responses in tumor stage cutaneous
side GD2. Cancer Immunol Immunother 1991; 34: 198–204. T cell lymphoma. Am J Hematol 1998; 58: 87–90.
471. Cumber AJ, Henry RV, Parnell GD, Wawrzynczak EJ. Purification 489. Saleh MN, LeMaistre CF, Kuzel TM et al. Antitumor activity of
of immunotoxins containing the ribosome inactivating proteins DAB389IL-2 fusion toxin in mycosis fungoides. J Am Acad
gelonin and momordin using high performance liquid immunoaf- Dermatol 1998; 39: 63–73.
finity chromatography compared with blue sepharose CL-6B 490. Talpur R, Apisarnthanarax N, Ward S, Duvic M. Treatment of
affinity chromatography. J Immunol Meth 1990; 135: 15–24. relapsed refractory peripheral T-cell lymphoma with denileukin
Arthur E. Frankel et al. 445

diftitox (Ontak): a case report of one patient. Leuk Lymph 2002; Sezary syndrome: correlation of activity and interleukin-2 receptor
43: 121–6. expression in a phase II study. Blood 1994; 84: 1765–74.
491. Gottlieb SL, Gilleaudeau P, Johnson R et al. Response of psoria- 509. Sewell KL, Parker KC, Woodworth TG, Reuben J, Swartz W,
sis to a lymphocyte-selective toxin (DAB389IL-2) suggests a Trentham DE. DAB486IL-2 fusion toxin in refractory rheuma-
primary immune, but not keratinocyte, pathogenic basis. Nat toid arthritis. Arthritis Rheum 1993; 9: 1223–33.
Med 1995; 1: 442–7. 510. Moreland LW, Sewell KL, Trentham DE et al. Interleukin-2 diph-
492. Gottlieb AB, Bacha P, Parker K, Strand V. Use of the interleu- theria fusion protein (DAB486IL-2) in refractory rheumatoid
kin-2 (IL-2) fusion protein, DAB389IL-2, for the treatment of arthritis. Arthritis Rheum 1995; 9: 1177–86.
psoriasis. Dermatol Ther 1998; 5: 48–63. 511. Schrohenloher RE, Koopman WJ, Woodworth TG, Moreland
493. Bagel J, Garland WT, Breneman D et al. Administration of LW. Suppression of in vitro IgM rheumatoid factor produc-
DAB389IL-2 to patients with recalcitrant psoriasis: a double-blind, tion by diphtheria toxin interleukin 2 recombinant fusion protein
phase II multicenter trial. J Am Acad Dermatol 1998; 38: 938–44. (DAB486IL-2) in patients with refractory rheumatoid arthritis.
494. Uckun FM. Immunotoxins for the treatment of leukaemia. Br J J Rheumatol 1996; 23: 1845–8.
Haematol 1993; 85: 435–8. 512. LeMaistre CF, Rosen S, Frankel A et al. Phase I trial of H65-RTA
495. Sausville EA, Headlee D, Stetler-Stevenson M et al. Continuous immunoconjugate in patients with cutaneous Tcell lymphoma.
infusion of the anti-CD22 immunotoxin IgG-RFB4-SMPT-dgA Blood 1991; 78: 1173–82.
in patients with B-cell lymphoma: a phase I study. Blood 1995; 513. Stone MJ, Sausville EA, Fay JW et al. A phase I study of bolus
85: 3457–65. versus continuous infusion of the anti-CD19 immunotoxin, IgG-
496. Senderowicz AM, Vitetta F, Headlee D et al. Complete sustained HD37-dgA, in patients with B-cell lymphoma. Blood 1996; 88:
response of a refractory, post-transplantation, large B-cell lym- 1188–97.
phoma to an anti-CD22 immunotoxin. Ann Intern Med 1997; 514. Messman RA, Vitetta ES, Headlee D et al. A phase I study of
126: 882–5. combination therapy with immunotoxins IgG-HD37deglycosylated
497. Amlot PL, Stone MJ, Cunningham D et al. A phase I study of an ricin A chain (dgA) and IgG-RFB4-dgA (Combotox) in patients
anti-CD22-deglycosylated ricin A chain immunotoxin in the with refractory CD 19(+), CD22(+) B cell lymphoma. Clin
treatment of b-cell lymphomas resistant to conventional therapy. Cancer Res 2000; 6: 1302–13.
Blood 1993; 82: 2624–33. 515. Grossbard ML, Freedman AS, Ritz J et al. Serotherapy of B-cell
498. Vitetta ES, Stone M, Amlot P et al. Phase I immunotoxin trial in neoplasms with anti-B4-blocked ricin: a phase I trial of daily
patients with B-cell lymphoma. Cancer Res 1991; 51: 4052–8. bolus infusion. Blood 1992; 79: 576–85.
499. Falini B, Bolognesi A, Flenghi L et al. Response of refractory 516. Grossbard ML, Lambert JM, Goldmacher VS et al. AntiB4-
Hodgkin’s disease to monoclonal anti-CD30 immunotoxin. blocked ricin: a phase I trial of 7-day continuous infusion in
Lancet 1992; 339: 1195–6. patients with B-cell neoplasms. J Clin Oncol 1993; 11: 726–37.
500. Goldberg MR, Heimbrook DC, Russo P et al. Phase I clinical 517. Rand RW, Kreitman RJ, Patronas N, Varricchio F, Pastan I, Puri
study of the recombinant oncotoxin TP40 in superficial bladder RK. Intratumoral administration of recombinant circularly per-
cancer. Clin Cancer Res 1995; 1: 57–61. muted interleukin-4 Pseudomonas exotoxin in patients with high-
501. Tepler I, Schwartz G, Parker K et al. Phase I trial of an interleu- grade glioma. Clin Cancer Res 2000; 6: 2157–65.
kin-2 fusion toxin (DAB4861L-2) in hematologic malignancies: 518. LeMaistre CF, Deisseroth A, Foget B et al. Phase I trial of H65-
complete response in a patient with Hodgkin’s disease refractory RTA in patients with chronic lymphocytic leukemia. Blood 1990;
to chemotherapy. Cancer 1994; 73: 1276–85. 76(Suppl. 1): 295a.
502. Platanias, LC, Ratain, MJ, O’Brien, S, Larson, RA, Vardiman, 519. Weiner LM, O’Dwyer J, Kitson J et al. Phase I evaluation of an
JW, Shaw, JP, Williams, SF, Baron, JM, Parker, K, Woodworth, anti-breast carcinoma monoclonal antibody 260F9 recombinant
TG. Phase I trial of a genetically engineered interleukin-2 fusion ricin A chain immunoconjuate. Cancer Res 1989; 49: 4062–7.
toxin (DAB486L-2) as a 6 h intravenous infusion in patients with 520. Spitler LE, del Rio M, Khentigan A et al. Therapy of patients
hematologic malignancies. Leuk Lymph 1994; 14: 257–62. with malignant melanoma using a monoclonal anti-melanoma
503. LeMaistre CF, Craig FE, Meneghetti C et al. Phase I trial of a 90 antibody-ricin A chain immunotoxin. Cancer Res 1987; 47:
minute infusion of the fusion toxin DAB486IL-2 in hematological 1717–23.
cancers. Cancer Res 1993; 53: 3930–4. 521. Oratz R, Speyer JL, Wernz JC et al. Antimelanoma monoclonal
504. LeMaistre CF, Meneghetti C, Rosenblum M et al. Phase I trial of antibody ricin A chain immunoconjugate (XMMME-001-RTA)
an interleukin-2 (IL-2) fusion toxin (DAB486IL-2) in hemato- plus cyclophosphamide in the treatment of metastatic malignant
logic malignancies expressing the IL-2 receptor. Blood 1992; 79: melanoma: results of a phase II trial. J Biol Resp Mod 1990; 9:
2547–54. 345–54.
505. LeMaistre CF, Rosenblum MG, Reuben JM et al. Therapeutic 522. Spitler L, Mishak R, Scannon P. Therapy of metastatic melanoma
effects of genetically engineered toxin (DAB486IL-2) in patient with Xomazyme Mel, a murine monoclonal antibody ricin A chain
with chronic lymphocytic leukaemia. Lancet 1991; 337: 1124–5. immunotoxin. Int J Rad Appl Instrum [B] 1989; 16: 625–7.
506. Hesketh P, Caguioa P, Koh H et al. Clinical activity of a cytotoxic 523. Waldman T, Pastan I, Gansow O, Junghans R. The multichain
fusion protein in the treatment of cutaneous T-cell lymphoma. interleukin-2 receptor: a target for immunotherapy. Ann Intern
J Clin Oncol 1993; 11: 1682–90. Med 1992; 116: 148–60.
507. vanderSpek J, Cosenza L, Woodworth T, Nichols JC, Murphy JR. 524. Bookman MA, Godfrey S, Padavic K et al. Anti-transferrin
Diphtheria toxin-related cytokine fusion proteins: elongation receptor immunotoxin (IT) therapy: phase I intraperitoneal (i.p)
factor 2 as a target for the treatment of neoplastic disease. Mol trial. Proc Am Soc Clin Oncol 1990; 9: 187.
Cell Biochem 1994; 138: 151–6. 525. Laske DW, Muraszko KM, Oldfield EH et al. Intraventricular
508. Foss FM, Borkowski TA, Gilliom M et al. Chimeric fusion pro- immunotoxin therapy for leptomeningeal neoplasia. Neurosurgery
tein toxin DAB4861L-2 in advanced mycosis fungoides and the 1997; 41: 1039–51.
446 Immunotoxins

526. LynchTJ, Lambert JM, Coral F et al. Immunotoxin therapy of 543. Onda M, Nagata S, Tsutsumi Y et al. Lowering the isoelectric
small-cell lung cancer: a phase I study of N901-blocked ricin. J point of the Fv portion of the recombinant immunotoxins leads to
Clin Oncol 1997; 15: 723–34. decreased non-specific animal toxicity without affecting anti-
527. Pai-Scherf LH, Villa J, Pearson D, Watson T, Liu E, Willingham tumor activity. Cancer Research 2001; 61: 5070–7.
MC, Pastan I. Hepatotoxicity in cancer patients receiving erb-38, 544. Onda M, Willingham MC, Wang QC et al. Inhibition of TNF-
a recombinant immunotoxin that targets the erbB2 receptor. Clin alpha produced by Kupffer cells protects against the nonspecific
Cancer Res 1999; 5: 2311–5. liver toxicity of immunotoxin anti-Tac(Fv)PE38, LMB-2. J
528. van Oosterhout YVJM, van Emst L, Schattenberg AVMB et al. A Immunol 2000; 165: 7150–6.
combination of anti-CD3 and anti-CD7 ricin A-immunotoxins 545. Baluna R, Coleman F, Jones C, Ghetie V, Vitetta FS. The effect of
for the in vivo treatment of acute graft versus host disease. Blood a monoclonal antibody coupled to ricin A chain derived peptides
2000; 95: 3693–701. on endothelial cells in vitro: insights into toxin-mediated vascular
529. Engert A, Diehl V, Schnell R et al. A phase I study of an anti- damage. Exp Cell Res 2000; 258: 417–24.
CD25 ricin A-chain immunotoxin (RFT5-SMPT-dgA) in 546. Siegall CB, Liggitt D, Chace D et al. Characterization of vascular
patients with refractory Hodgkin’s lymphoma. Blood 1997; 89: leak syndrome induced by the toxin component of Pseudomonas
403–10. exotoxin-based immunotoxins and its potential inhibition with
530. Postan. I, Hassam R, Fitzgerald DJ et al. Immunotoxin therapy nonsteroidal anti-inflammatory drugs. Clin Cancer Res 1997; 3:
of cancer. Nature Rev. Cancer 2006; 6; 559-565. 339–45.
531. Frankel AE, Powell BL, Hall PD, Case LD, Kreitman RJ. Phase 547. Kaneta Y, Tsukazaki K, Kubushiro K et al. Effect of gelonin
I trial of a novel diphtheria toxin/granulocyte macrophage col- immunoconjugate with monoclonal antibody MSN-1 to endome-
ony-stimulating factor fusion protein (DT388GMCSF) for refrac- trial adenocarcinoma on antigen-producing tumor cells in vivo.
tory or relapsed acute myeloid leukemia. Clin Cancer Res 2002; Jpn J Cancer Res 1998; 89: 583–8.
8: 1004–13. 548. Singh R, Browning JL, Abi-Habib R et al. Recombinant prostate-
532. Theodoulou M, Baselga J, Scher H et al. Phase I doseescalation specific antigen proaerolysin shows selective protease sensitivity
study of the safety, tolerability, pharmacokinetics and biologic and cell cytotoxicity. Anticancer Drugs 2007; 18: 809–16.
effects of DAB389EGF in patients with solid malignancies that 549. Williams SA, Merchant RF, Garrett-Mayer E et al. A prostate-
express EGF receptors (EGFr). Proc Am Soc Clin Oncol 1995; specific antigen-activated channel-forming toxin as therapy for
14: 480. prostate disease. J Natl Cancer Inst 2007; 99: 376–85.
533. Byers VS, Henslee PJ, Kernan NA et al. Use of an anti-pan 550. Su Y, Ortiz J, Liu S et al. Systemic urokinase-activated anthrax
T-lymphocyte ricin A chain immunotoxin in steroid-resistant toxin therapy produces regressions of subcutaneous human non-
acute graft-versus-host disease. Blood 1990; 75: 1426–32. small cell lung tumor in athymic nude mice. Cancer Res 2007;
534. Martin PJ, Nelson BJ, Appelbaum FR et al. Evaluation of an 67: 3329–36.
CD5-specific immunotoxin for treatment of acute graft-versus- 551. Stish BJ, Oh S, Vallera DA. Anti-glioblastoma effect of a recom-
host disease after allogeneic marrow transplantation. Blood 1996; binant bispecific cytotoxin cotargeting human IL-13 and EGF
88: 824–30. receptors in a mouse xenograft model. J Neurooncol 2007; Dec
535. Grossbard ML, Mullion PS, Freedman AS et al. A phase II study 15 Epub.
of adjuvant therapy with anti-B4-blocked ricin after autologous 552. Stish BJ, Chen H, Shu Y et al. Increasing anticarcinoma activity
bone marrow transplantation for patients with relapsed B-cell of an anti-erbB2 recombinant immunotoxin by the addition of an
non-Hodkgin’s lymphoma. Clin Cancer Res 1999; 5: 2392–8. anti-EpCAM sFv. Clin Cancer Res 2007; 13: 3058–67.
536. Multani PS, O’Day S, Nadler LM, Grossbard ML. Phase II clini- 553. Rustamzadeh E, Vallera DA, Todhunter DA et al. Immunotoxin
cal trial of bolus infusion anti-B4 blocked ricin immunoconjugate pharmacokinetics: a comparison of the anti-glioblastoma bispe-
in patients with relapsed B-cell non-Hodgkin’s lymphoma. Clin cific fusion protein (DTAT13) to DTAT and DTIL13. J Neurooncol
Cancer Res 1998; 4: 2599–604. 2006; 77: 257–66.
537. Scadden DT, Schenkein DP, Bernstein Z et al. Immunotoxin 554. Vallera DA, Todhunter DA, Kuroki DW et al. A bispecific recom-
combined with chemotherapy for patients with AIDS-related binant immunotoxin, DT2219, targeting human CD19 and CD22
non-Hodgkin’s lymphoma. Cancer 1998; 83: 2580–7. receptors in a mouse xenograft model of B-cell leukemia/lym-
538. Grossbard ML, Fidias P, Kinsella 1 et al. Anti-B4-blocked ricin: phoma. Clin Cancer Res 2005; 11: 3879–88.
a phase II trial of 7 day continuous infusion in patients with mul- 555. Abi-Habib RJ, Liu S, Bugge TH et al. A urokinase-activated
tiple myeloma. Br J Haematol 1998; 102: 509–15. recombinant diphtheria toxin targeting the granulocyte-mac-
539. Piascik P. FDA approves fusion protein for treatment of lym- rophage colony-stimulating factor receptor is selectively cyto-
phoma. J Am Pharm Assoc 1999; 39: 571–2. toxic to human acute myeloid leukemia blasts. Blood 2004; 104:
540. Hall PD, Virella G, Willoughby T, Atchley DH, Kreitman RJ, 2143–8.
Frankel AE. Antibody response to DT-GM, a novel fusion toxin 556. Stish BJ, Chen H, Shu Y et al. A bispecific recombinant cytotoxin
consisting of a truncated diphtheria toxin (DT) linked to human (DTEGF13) targeting human interleukin-13 and epidermal growth
granulocyte macrophage colony-stimulating factor (GM), during factor receptors in a mouse xenograft model of prostate cancer.
a phase I trial of patients with relapsed or refractory acute myel- Clin Cancer Res 2007; 13: 6486–93.
oid leukemia. Clin Immunol 2001; 100: 191–7. 557. Avia AD, Calderon CF, Perez RM et al. Construction of an immu-
541. Rose DM, Pai LH, Pastan I. Identification of epitopes on a mutant notoxin by linking a monoclonal antibody against the human epi-
form of Pseudomonas exotoxin using serum from humans treated dermal growth factor receptor and a hemolytic toxin. Biol Res
with Pseudomonas exotoxin containing immunotoxins. Eur J 2007; 40: 173–183.
Immunol 1997; 27: 1459–68. 558. Benitez J, Ferreras JM, Munoz R et al. Cytotoxicity of an ebulin
542. Hertler AA, Spitler LE, Frankel AE. Humoral immune response I-anti-human CD105 immunotoxin on mouse fibroblasts (L929)
to a ricin A chain immunotoxin in patients with metastatic mela- and rat myoblasts (L6E9) cells expressing human CD105. Med
noma. Cancer Drug Delivery 1987; 4: 245–53. Chem 2005; 1: 65–70.
Arthur E. Frankel et al. 447

559. Cohen S, Cahan R, Ben-Dov E et al. Specific targeting to murine fragment variable antibody against CTLA4 and the N-terminal
myeloma cells of Cyt1Aa toxin from Bacillus thuringiensis spub- fragment of human perforin. Protein Expr Purif 2006; 48: 307–13.
species israelensis. J Biol Chem 2007; 282: 28301–8. 577. Onda M, Wang QC, Guo HF et al. In vitro and in vivo cytotoxic
560. Foster KA, Adams EJ, Durose L et al. Re-engineering the target activities of recombinant immunotoxin 8H9(Fv)-PE38 against
specificity of Clostridial neurotoxins-a route to novel therapeu- breast cancer, osteosarcoma and neuroblastoma. Cancer Res
tics. Neurotox Res 2006; 9: 101–7. 2004; 64: 1419–24.
561. Jia Y, Li H, Chen W, Li M et al. Prevention of murine autoimmune 578. Saavedra-Lozano J, Cao Y, Callison J et al. An anti-CD45RO
encephalomyelitis by in vivo expression of a novel recombinant immunotoxin kills HIV-latently infected cells from individuals
immunotoxin DT390-RANTES. Gene Ther 2006; 13: 1351–9. on HAART with little effect on CD8 memory. Proc Natl Acad Sci
562. Bang SH, Nagata S, Onda M et al. HA22(R490A) is a recombi- USA 2004; 101: 2494–9.
nant immunotoxin with increased antitumor activity without an 579. Martsev SP, Tsybovsky YI, Stremovskiy OA et al. Fusion of the anti-
increase in animal toxicity. Clin Cancer Res 2005; 11: 1545–50. ferritin antibody VL domain to barnase results in enhanced solubility
563. Marks AJ, Cooper MS, Anderson RJ et al. Selective apoptotic and altered pH stability. Protein Eng Des Sel 2004; 17: 85–93.
killing of malignant hemopoietic cells by antibody-targeted 580. Kurschus FC, Kleinschmidt M, Fellows E et al. Killing of target
delivery of an amphipathic peptide. Cancer Res 2005; 65: cells by redirected granzyme B in the absence of perforin. FEBS
2373–7. Lett 2004; 562: 87–92.
564. Pincus SH, Fang H, Wilkinson RA et al. In vivo efficacy of anti- 581. Polito L, Bolognesi A, Tazzari PL et al. the conjugate Rituximab/
glycoprotein 41, but not anti-glycoprotein 120, immunotoxins in a saporin-S6 completely inhibits clonogenic growth of CD20-
mouse model of HIV infection. J Immunol 2003; 170: 2236–41. expressing cells and produces a synergistic toxic effect with
565. Stocker M, Tur MK, Sasse S et al. Secretion of functional anti- Fludarabine. Leukemia 2004; 18: 1215–22.
CD30-angiogenin immunotoxins into the supernatant of trans- 582. De Lorenzo C, Arciello A, Cozzolino R et al. A fully human anti-
fected 293T-cells. Protein Expr Purif 2003; 28: 211–9. tumor immunoRNase selective for ErbB-2-positive carcinomas.
566. van Roon JA, van Vuuren AJ, Wijngaarden S et al. Selective Cancer Res 2004; 64: 4870–4.
elimination of synovial inflammatory macrophages in rheuma- 583. Huang X, Bennett M, Thorpe PE. Anti-tumor effects and lack of
toid arthritis by an Fcgamma receptor 1-directed immunotoxin. side effects in mice of an immunotoxin directed against human
Arthritis Rheum 2003; 48: 1229–38. and mouse prostate-specific membrane antigen. Prostate 2004;
567. Shin YK, Choi YL, Choi EY et al. Targeted cytotoxic effect of 61: 1–11.
anti-JL1 immunotoxin against a human leukemia cell line and its 584. Bruell D, Bruns CJ, Yezhelyev M et al. Recombinant anti-EGFR
clinical implications. Cancer Immunol Immunother 2003; 52: immunotoxin 425(scFv)-ETA’ demonstrates anti-tumor activity
506–12. against disseminated human pancreatic cancer in nude mice. Int J
568. Di Paolo C, Willuda J, Kubetzko S et al. A recombinant immuno- Mol Med 2005; 15: 305–13.
toxin derived from a humanized epithelial cell adhesion mole- 585. Li H, Li MY, Jiang ZH et al. Construction and prokaryotic expres-
cule-specific single-chain antibody fragment has potent and sion of a recombinant immunotoxin fused with mouse interleukin
selective anti-tumor activity. Clin Cancer Res 2003; 9: 2837–48. 18 and truncated Pseudomonas exotoxin. Sichuan Da Xue Xue
569. Rosenblum MG, Cheung LH, Liu Y et al. Design, expression, Bao Yi Xue Ban 2004; 35: 753–6.
purification, and characterization, in vitro and in vivo, of an anti- 586. Arditti FD, Rabinkov A, Miron T et al. Apoptotic killing of
melanoma single-chain Fv antibody fused to the toxin gelonin. B-chronic lymphocytic leukemia tumor cells by allicin generated
Cancer Res 2003; 63: 3995–4002. in situ using a rituximab-allinase conjugate. Mol Cancer Ther
570. Otten HG, de Gast GC, Vooijs WC et al. Preclinical evaluation of 2005; 4: 325–31.
anti-CD86 immunotoxin in rhesus monkeys: analysis of systemic 587. Cui JX, Ji JF, Lv AG et al. Construction and expression of novel
toxicity, pharmacokinetics, and effect on primary T-cell responses. immunotoxin cpIL-4 (13D)-PE38KDEL with increased activity.
Cancer Immunol Immunother 2003; 52: 569–75. Biochemistry 2005; 70: 62–8.
571. Xia HC, Li F, Zhang ZC. Purification and characterization of 588. Erickson HA, Jund MD, Pennell CA. Cytotoxicity of human RNase-
Moschatin, a novel type I ribosome-inactivating protein from the based immunotoxins requires cytosolic access and resistance to
mature seeds of pumpkin (Cucurbita moschata) and preparation ribonuclease inhibition. Protein Eng Des Sel 2006; 19: 37–45.
of its immunotoxin against human melanoma cells. Cell Res 589. Foehr ED, Lorente G, Kuo J et al. Targeting of the receptor protein
2003; 13: 369–74. tyrosine phosphatase β with a monoclonal antibody delays tumor
572. Zhang RP, Xiong CY, Yan M, Zhang ZC. In vitro inhibition of growth in a glioblastoma model. Cancer Res 2006; 66: 2271–8.
human melanoma cells by immunotoxin luffin B-Ng76. Sheng 590. Bremer E, Samplonius DF, Peipp M et al. Target cell-restricted apop-
Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai) 1998; 30: tosis induction of acute leukemic T cells by a recombinant tumor
561–4. necrosis factor-related apoptosis-inducing ligand fusion protein with
573. Tur MK, Huhn M, Thepen T et al. Recombinant CD64-specific specificity for human CD7. Cancer Res 2005; 65: 3380–8.
single chain immunotoxin exhibits specific cytotoxicity against 591. Chang CH, Sapra P, Vanama SS et al. Effective therapy of human
acute myeloid leukemia cells. Cancer Res 2003; 63: 8414–9. lymphoma xenografts with a novel recombinant ribonuclease/
574. Liu Y, Cheung LH, Marks JW et al. Recombinant single-chain anti-CD74 humanized IgG4 antibody immunotoxin. Blood 2005;
antibody fusion construct targeting human melanoma cells and 106: 4308–14.
containing tumor necrosis factor. Int J Cancer 2004; 108: 549–57. 592. Nagai T, Tanaka M, Tsuneyoshi Y et al. In vitro and in vivo effi-
575. Palmisano GL, Tazzari PL, Cozzi E et al. Expression of CTLA-4 cacy of a recombinant immunotoxin against folate receptor beta
in non-human primate lymphocytes and its use as a potential target on the activation and proliferation of rheumatoid arthritis syn-
for specific immunotoxin-mediated apoptosis: results of in vitro ovial cells. Arthritis Rheum 2006; 54: 3126–34.
studies. Clin Exp Immunol 2004; 135: 259–66. 593. Stocker M, Klockenbring T, Huhn M et al. Antigen-specific tar-
576. Wan L, Zeng L, Chen L et al. Expression, purification, and refold- geting and elimination of EBV-transformed B cells by allergen
ing of a novel immunotoxin containing humanized single-chain toxins. J Allergy Clin Immunol 2005; 116: 910–5.
448 Immunotoxins

594. Hossann M, Li Z, Shi Y et al. Novel immunotoxin: a fusion pro- 611. Schwemmlein M, Stieglmaier J, Kellner C et al. A CD19-specific
tein consisting of gelonin and an acetylcholine receptor fragment single-chain immunotoxin mediates potent apoptosis of B-lineage
as a potential immunotherapeutic agent for the treatment of leukemic cells. Leukemia 2007; 21: 1405–12.
Myasthenia gravis. Protein Expr Purif 2006; 46: 73–84. 612. Wang H, Song S, Kou G et al. Treatment of hepatocellular carci-
595. Wolf P, Gierschner D, Buhler P et al. A recombinant PSMA- noma in a mouse xenograft model with an immunotoxin which is
specific single-chain immunotoxin has potent and selective toxic- engineered to eliminate vascular leak syndrome. Cancer Immunol
ity against prostate cancer cells. Cancer Immunol Immunother Immunother 2007; 56: 1775–83.
2006; 55: 1367–73. 613. Kim GB, Wang Z, Liu YY et al. A fold-back single-chain dia-
596. Bolognesi A, Polito L, Farini V et al. CD38 as a target of IB4 mAb body format enhances the bioactivity of an anti-monkey CD3
carrying saporin-S6: design of an immunotoxin for ex vivo deple- recombinant diphtheria toxin-based immunotoxin. Protein Eng
tion of hematological CD38+ neoplasia. J Biol Regul Homeost Des Sel 2007; 20: 425–32.
Agents 2005; 19: 145–52. 614. Mazor Y, Noy R, Wels WS et al. chFRP5-ZZ-PE38, a large IgG-
597. Schwemmlein M, Peipp M, Barbin K et al. A CD33-specific single- toxin immunoconjugate outperforms the corresponding smaller
chain immunotoxin mediates potent apoptosis of cultured human FRP5(Fv)-ETA immunotoxin in eradicating ErbB2-expressing
myeloid leukaemia cells. Br J Haematol 2006; 133: 141–51. tumor xenografts. Cancer Lett 2007; 257: 124–135.
598. van Vuuren AJ, van Roon JA, Walraven V et al. CD64-directed 615. Du X, Nagata S, Ise T et al. FCRL1 on chronic lymphocytic leu-
immunotoxin inhibits arthritis in a novel CD64 transgenic rat kemia, hair cell leukemia and B-cell non-Hodgkin’s lymphoma
model. J Immunol 2006; 176: 5833–8. as a target of immunotoxins. Blood 2007; Sept 25 Epub.
599. Zhang H, Zhang S, Zhang H et al. Cytotoxicity of a novel fibro- 616. Filpula D, Yang K, Basu A et al. Releasable PEGylation of meso-
blast growth factor receptor targeted immunotoxin on a human thelin targeted immunotoxin SS1P achieves single dosage com-
ovarian teratocarcinoma cell line. Cancer Biother Radiopharm plete regression of a human carcinoma in mice. Bioconj Chem
2006; 21: 321–32. 2007; 18: 773–84.
600. Fuchs H, Bachran C, Li T et al. A cleavable molecular adapter 617. Kreitman RJ, Squires DR, Stetler-Stevenson M et al. Phase I trial
reduces side effects and concomitantly enhances efficacy in of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in
tumor treatment by targeted toxins in mice. J Control Release patients with B-cell malignancies. J Clin Oncol 2005; 23:
2007; 117: 342–50. 6719–29.
601. Hess PR, Barnes C, Woolard MD et al. Selective deletion of 618. Powell DJ, Felipe-Silva A, Merino MJ et al. Administration of a
antigen-specific CD8+ T cells by MHC class I tetramers coupled CD25-directed immunotoxin, LMB-2, to patients with metastatic
to the type I ribosome-inactivating protein saporin. Blood 2007; melanoma induces a selective partial reduction in regulatory T
109: 3300–7. cells in vivo. J Immunol 2007; 179: 4919–28.
602. Xi Y, Yuan Z, Zhang H et al. Molecular construction and 619. Dang NH, Pro B, Hagemeister FB et al. Phase II trial of denileukin
characterization of a novel exotoxin fusion protein that selec- diftitox for relapsed/refractory T-cell non-Hodgkin’s lymphoma.
tively blocks the B7:CD28 costimulatory signal system. J Br J Haematol 2007; 136: 439–47.
Immunother 2006; 29: 586–95. 620. Frankel AE, Surendranathan A, Black JH et al. Phase II clinical
603. Singh R, Samant U, Hyland S et al. Target-specific cytotoxic studies ofdenileukin diftitox diphtheria toxin fusion protein in
activity of recombinant immunotoxin scFv(MUC1)-ETA on patients with previously treated chronic lymphocytic leukemia.
breast carcinoma cells and primary breast tumors. Mol Cancer Cancer 2006; 106: 2158–64.
Ther 2007; 6: 562–9. 621. Dang NH, Hagemeister FB, Pro B et al. Phase II study of denileu-
604. Chen W, Li H, Jia Y et al. In vivo administration of plasmid kin diftitox for relapsed/refractory B-cell non-Hodgkin’s lym-
DNA encoding recombinant immunotoxin DT390-IP-10 attenu- phoma. J Clin Oncol 2004; 22: 4095–102.
ates experimental autoimmune encephalomyelitis. J Autoimmun 622. Quintas-Cardama A, Kantarjian H, Verstovsek S. Treatment of
2007; 28: 30–40. systemic mastocytosis with denileukin diftitox. Am J Hematol
605. Du X, Ho M, Pastan I. New immunotoxins targeting CD123, a 2007; 82: 1124.
stem cell antigen on acute myeloid leukemia cells. J Immunother 623. Kerl K, Prins C, Cerroni L, French LE. Regression of extranodal
2007; 30: 607–13. natural killer/T-cell lymphoma nasal type with denileukin diftitox
606. Baatar D, Olkhanud P, Newton D et al. CCR4-expressing T cell (Ontak) and bexarotene (Targretin): report of a case. Br J
tumors can be specifically controlled via delivery of toxins to Dermatol 2006; 154: 988–91.
chemokine receptors. J Immunol 2007; 179: 1996–2004. 624. Hathaway T, Subtil A, Kuo P et al. Efficacy of denileukin diftitox in
607. Munoz R, Arias Y, Ferreras JM et al. Targeting a marker of the subcutaneous panniculitis-like T-cell lymphoma. Clin Lymphoma
tumour neovasculature using a novel anti-human CD105- Myeloma 2007; 7: 541–5.
immunotoxin containing the non-toxic type 2 ribosome-inacti- 625. Evens AM, Ziegler SL, Gupta R et al. Sustained hematologic and
vating protein nigrin b. Cancer Lett 2007; 256: 73–80. central nervous system remission with single-agent denileukin
608. Ochiai H, Archer GE, Herndon JE et al. EGFRvIII-targeted diftitox in refractory adult T-cell leukemia/lymphoma. Clin
immunotoxin induces antitumor immunity that is inhibited in the Lymphoma Myeloma 2007; 7: 472–4.
absence of CD4+ and CD8+ T cells. Cancer Immunol Immunother 626. Ho VT, Zahrieh D, Hochberg E et al. Safety and efficacy of
2008; 57: 115–21. denileukin diftitox in patients with steroid-refractory acute graft-
609. Braschoss S, Hirsch B, Dubel S et al. New anti-CD30 human pan- versus-host disease after allogeneic hematopoietic stem cell
creatic ribonuclease-based immunotoxin reveals strong and transplantation. Blood 2004; 104: 1224–6.
specific cytotoxicity in vivo. Leuk Lymphoma 2007; 48: 1179–86. 627. Talpur R, Jones DM, Alencar AJ et al. CD25 expression is cor-
610. Bak SP, Walters JJ, Takeya M et al. Scavenger receptor-A-targeted related with histological grade and response to denileukin diftitox
leukocyte depletion inhibits peritoneal ovarian tumor progres- in cutaneous T-cell lymphoma. J Invest Dermatol 2006; 126:
sion. Cancer Res 2007; 67: 4783–9. 575–83.
Arthur E. Frankel et al. 449

628. Ghori F, Polder KD, Pinter-Brown LC et al. Thyrotoxicosis after 638. Hassan R, Broaddus VC, Wilson S et al. Anti Mesothelin immu-
denileukin diftitox therapy in patients with mycosis fungoides. J notoxin SS1P in combination with gemcitabine results in
Clin Endocrinol Metab 2006; 91: 2205–8. increased activity against mesothelin-expressing tumor xeno-
629. Park M, Liu GT, Piltz-Seymour J et al. Vision loss following grafts. Clin Cancer Res 2007; 13: 7166–71.
denileukin diftitox treatment: a case report of possible posterior 639. Zhang Y, Xiang L, Hassan R et al. Immunotoxin and Taxol syn-
ischemic optic neuropathy. Leuk Lymphoma 2007; 48: 808–11. ergy results from a decrease in shed mesothelin levels in the
630. Dang NH, Fayad L, McLaughlin P et al. Phase II trial of the combi- extracellular space of tumors. Proc Natl Acad Sci USA 2007;
nation of denileukin diftitox and rituximab for relapsed/refractory 104: 17099–104.
B-cell non-Hodgkin’s lymphoma. Br J Haematol 2007; 138: 502–5. 640. Coffield S, Boyer A, Culp L et al. Intraprostatic treatment of
631. DiVenuti G, Nawgiri R, Foss F. Denileukin diftitox and hyper- patients with locally recurrent prostate cancer with the PSA-
CVAD in the treatment of human T-cell lymphotropic virus activated protoxin PRX302. ASCO GU Meeting, Feb 14–16
1-associated acute T-cell leukemia-lymphoma. Clin Lymphoma 2008, Orlando, FL. Abstract no. 20172.
2003; 4: 176–8. 641. Kunwar S, Prados MD, Chang SM et al. Direct intracerebral
632. Litzinger MT, Fernando R, Curiel TJ et al. IL-2 immunotoxin delivery of cintredekin besudotox (IL13-PE38QQR) in
denileukin diftitox reduces regulatory T cells and enhances vac- recurrent malignant glioma: a report by the Cintredekin
cine-mediated T-cell immunity. Blood 2007; 110: 3192–201. Besudotox Intraparenchymal Study Group. J Clin Oncol 2007;
633. Mahnke K, Schonfeld K, Fondel S et al. Depletion of CD4+ 25: 837–44.
CD25+ human regulatory T cells in vivo: kinetics of Treg deple- 642. Sampson JH, Raghavan R, Brady ML et al. Clinical utility of a
tion and alterations in immune functions in vivo and in vitro. Int patient-specific algorithm for simulating intracerebral drug infu-
J Cancer 2007; 120: 2723–33. sions. Neuro Oncol 2007; 9: 343–53.
634. Rainov NG, Soling A. Technology evaluation: TransMID, KS 643. Frankel AE, Liu JS, Rizzieri DA et al. Phase I clinical study of
Biomedix/Nycomed/Sosei/PharmaEngine. Curr Opin Mol Ther diphtheria toxin-interleukin 3 fusion protein in patients with
2005; 7: 483–92. acute myeloid leukemia and myelodysplasia. Leuk Lymphoma,
635. Weber FW, Floeth F, Asher A et al. Local convection enhanced in press.
delivery of IL4-Pseudomonas exotoxin (NBI-3001) for treatment 644. Liu TF, Urieto JO, Moore JE et al. Diphtheria toxin fused to vari-
of patients with recurrent malignant glioma. Acta Neurochir ant interleukin-3 provides enhanced binding to the interleukin-3
Suppl 2003; 88: 93–103. receptor and more potent leukemia cell cytotoxicity. Exp Hematol
636. Garland L, Gitlitz B, Ebbinghaus S et al. Phase I trial of intrave- 2004; 32: 277–81.
nous IL-4 pseudomonas exotoxin protein (NBI-3001) in patients 645. Azemar M, Djahansouzi S, Jager E et al. Regression of cutaneous
with advanced solid tumors that express the IL-4 receptor. J tumor lesions in patients intratumorally injected with a recombi-
Immunother 2005; 28: 376–81. nant single-chain antibody-toxin targeted to ErbB2/HER2. Breast
637. Hassan R, Bullock S, Premkumar A et al. Phase I study of SS1P, Cancer Res Treat 2003; 82: 155–64.
a recombinant anti-mesothelin immunotoxin given as a bolus I.V. 646. von Minckwitz G, Harder S, Hovelmann S et al. Phase I clinical
infusion to patients with mesothelin-expressing mesothelioma, study of the recombinant antibody toxin scFv(FRP5)-ETA spe-
ovarian, and pancreatic cancers. Clin Cancer Res 2007; 13: cific for the ErbB2/HER2 receptor in patients with advanced
5144–9. solid malignancies. Breast Cancer Res 2005; 7:R617–26.
12 Drug Immunoconjugates
MALEK SAFA, KENNETH A. FOON, AND ROBERT K. OLDHAM

Monoclonal antibodies and their immunoconjugates kinds of genotypic inferences, these authors and others
represent one of the first practical methods for the selec- have demonstrated beyond doubt that heterogeneity is
tive treatment of cancer [49, 96]. Monoclonal antibody characteristic of both animal and human tumors (Fig. 1).
technology now allows for the generation of antibodies Recently, there have been references to the implications of
or “cocktails” of antibodies that have some selectivity this tumor-cell heterogeneity for treatment, and proposals
for cancer tissue as compared with the normal tissue of have been made to approach the problem clinically [58,
origin. These antibodies can be tested as unconjugated 59, 61–63, 74, 89].
antibody alone or in conjunction with effector cells. The There are two types of tumor-cell heterogeneity [73].
“signal strength” of the antibody may be made more There are differences between patients, with heterogene-
powerful by conjugating antibody to drugs, toxins, bio- ity being apparent among tumors of the same histological
logicals, and radioisotopes with different mechanisms class. This is macroheterogeneity. Tumors such as breast
of action and different levels of potency. This chapter cancer can be very similar by histological examination,
will focus on the use of drug immunoconjugates for and yet lethality can occur in 7 months or 17 years.
cancer treatment. Clinical observations have made it clear that what is seen
under the microscope bears little relationship to the
behavior of the cancer in the patient. This sort of “pheno-
typic analysis” has led to further investigations, and it is
The Problem of Heterogeneity: now recognized by most cancer biologists that consider-
Antibody-Based Therapeutics as able differences exist among patients with respect to the
phenotypic characteristics of the cancer. In addition, there
a Solution is the problem of heterogeneity within each tumor in each
The problem of tumor-cell heterogeneity (Fig. 1) and patient; microheterogeneity. Thus, many studies have
the implications for therapy have been reviewed in demonstrated that multiple clones may exist within the
Chapter 2. Curiously, although tumor-cell heterogeneity primary tumor and that these clones may have different
has been broadly recognized for decades, clinicians metastatic capabilities, giving rise to heterogeneity even
continue to take the simplistic view that treatment need among different deposits in a single patient.
not reflect a specific approach to heterogeneity [90, Many still believe that even with heterogeneity
113–115]. Thus, treatment is still designed as if there are between individuals and within each patient’s cancer,
singular underlying common principles useful in cancer underlying common features still exist and will allow a
therapy. Single modalities or fixed combinations aimed general treatment to be developed that might be useful
at eradicating cancer without a strategy designed to for all cancer, or all of a histological type of cancer. This
approach the problem of tumor-cell heterogeneity still view supports the “drug development paradigm,” wherein
dominate cancer research and treatment [60, 67, 68, 71, drugs are developed as broadly active agents to be tested
74, 89]. in different histological types of cancer (see Chapter 3).
Current data on tumor-cell heterogeneity and the bio- Another view is that each cancer and its antigenic phe-
logic basis of that heterogeneity is covered in Chapter 2 notype (and behavior) are unique. This view, currently
and have been previously reviewed [29]. The basic ten- held by a minority of investigators, would suggest the
ant of these studies is that each primary tumor is com- need to individualize treatment for each patient and may
posed of a smaller or larger number of clones, each of even require individual therapeutic manipulations for
which has its own genotypic and phenotypic character- a single patient over the clinical course of his or her
istics. Metastasis tends to occur from single cells or disease based on these differences [65, 68, 71–74].
clumps of cells from within the primary tumor. By virtue Acceptance of this hypothesis would dramatically change
of a series of phenotypic analyses, as well as certain cancer treatment and would require a laboratory-clinic

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 451
© Springer Science + Business Media B.V. 2009
452 Drug Immunoconjugates

Figure 1. Illustration of tumor cell heterogeneity

interface of a type not often used in cancer therapeutics. intravenous injection [62, 63, 93]. Biodistribution
With the advent of the “new biology” using genetically studies utilizing antibody conjugated to tracer quantities
engineered biologicals and monoclonal antibodies, one of isotope have demonstrated excellent tumor imaging,
has diverse mechanisms for the generation of biological but a considerable amount of the antibody is distributed
responses, which may match the diversity implicit in to the liver, spleen, and other organs of the reticuloen-
tumor cell populations. For the first time, there is hope dothelial system [2, 12, 13, 35, 43, 46, 113]. The infre-
that one can truly approach the heterogeneity of each quency of response to unconjugated antibody and the
patient’s cancer and the other biologic differences of each ability of conjugates to induce regression of bulky cancer
patient in a rational manner [36, 58, 59, 60, 62, 63, 65, 74]. in animal models support the concept of immunoconju-
gate therapy.
When considering the development of a conjugate–
monoclonal antibody complex for drug targeting, the
Rationale for following factors are of importance:
Immunoconjugates 1. The recognition site for the monoclonal antibody should be
located on the surface of the cell.
Most of the early monoclonal antibody clinical trials
2. The antibodies should have sufficient tumor tissue specificity.
focused on the use of unconjugated single antibodies 3. The extent of localization of the antibody at the target site.
in order to determine toxicity, tolerance, the localization 4. Biodistribution of the conjugate–antibody conjugate in the
of antibody in solid tumor deposits, and the distribution body relative to that of the parent antibody.
of antibody in normal and neoplastic tissues [21, 31, 5. Stability of the conjugate–antibody complex in blood.
6. The toxicity of the conjugate.
53–55, 81, 83, 93–95, 96]. Although the clinical responses
7. The conjugate must be non-immunogenic and biodegradable.
to unconjugated antibody have not been consistent, there 8. The conjugate must be released upon interaction between
is unequivocal evidence that antibody does selectively localize the carrier molecule and the cell and the toxic component
in tumor deposits and on individual cancer cells after must be active at the surface or be internalized into the cell.
Malek Safa et al. 453

Table 1. Optimization of monoclonal antibody therapy


Rationale for Antitumor
ANTIBODY SPECIFICITY
Cocktails Immunoperoxidase
Immunofluorescence
There already exist many different monoclonal antibod- Radiolocalization
ies to be assessed in clinical trials in patients with solid ANTIGEN CHARACTERIZATION
tumors [18, 19, 65, 74]. A large number of antibodies for Biochemical nature
Topography and density
leukemia, lymphoma, melanoma, lung, breast, and
Epitopes
colon cancer have been described and characterized. Heterogeneity
Antibody preparations are available as immunoglobulin ANTIBODY–ANTIGEN INTERACTION
M (IgM), IgA, and various subclasses of IgG. There are Turnover of antibody bound to tumor cells
several monoclonal antibodies which have been engi- Degree of antibody internalization
Antigen affinity
neered to be predominantly human. These are: Rituximab Antigen levels in serum
(Rituxan) directed against CD-20 antigen, Campath ANTIBODY DELIVERY
(Alemtuzumab) against CD52, Trastuzumab (Herceptin) Dose
against HER-2/neu receptor, Bevacizumab (Avastin) Regimen
against VEGF, Cetuximab (Erbitux) and Panitumumab Route
Pharmacokinetics
(Vectibix) against EGFR receptor. These antibodies Comparison of various cytotoxic agents conjugated to the
have been approved and are in clinical use for the treat- same antibody
ment of non-Hodgkin’s lymphoma, CLL, breast, lung
and colon cancer [52, 74, 112]. In addition, the use of
murine monoclonal antibody MAb-17–1A has resulted Given a wide choice of antibody preparations and the
in improved survival of patients with Dukes’ C colorec- ability to prepare them for the clinic, the next consider-
tal cancer [82]. A large variety of new monoclonal anti- ation is, how does one select preparations and patients
bodies, both murine and human, continue to be for clinical trials? It seems unlikely that firm rules can
discovered and developed. The recent success in the be made on the selection of antibody preparations for all
humanization process has further improved the bio- solid tumors. Obviously, the distribution of the tumor,
availability and reduced the immunogenicity of these its vascularity, its sensitivity to drugs and radiation, and
antibodies. Furthermore, the technological evolution the quantitative expression of antigens on the tumor cell
from murine-based therapeutic monoclonal antibodies surfaces are all significant factors. Using conjugates of
to chimeric (part murine part human protein such as isotopes, drugs, biologicals, and toxins, the issue may
cetuximab), humanized (e.g. trastuzumab) and fully be essentially one of biodistribution within the tumor
humanized antibodies (bevacizumab, panitumumab) bed and access of the toxic agent to the tumor cell within
has led to reduction in immune-mediated clearance and each tumor nodule. Thus, it is apparent that while cer-
hypersensitivity, improved the safety and feasibility of tain principles in the use of monoclonal antibody for
repeated administration and thereby created a viable human solid tumors may be important, each tumor type
therapeutic strategy. Thus, it is apparent that the limita- may have to be individually evaluated for the optimal
tion for the use of monoclonal antibody preparations in use of a monoclonal antibody preparation [73, 74].
treatment will not be due to a shortage of antibody prep- Antibodies presumably must reach the tumor bed to
arations [58, 74]. be effective [58, 62, 63]. One general principle has been
Although the “perfect” antibody for use as an unconju- that antibody fragments may more quickly diffuse from
gated antibody or as a targeting agent has not been iden- the vascular compartment to the tumor bed. Data have
tified for any human cancer, there are a variety of selective indicated that the more antibody one infuses into the
antibodies available for clinical investigation. Methods vascular compartment, the more antibody one delivers
exist to manufacture high-purity (>99%), homogeneous to the tumor cell bed [58, 62, 63]. Although access to the
preparations of monoclonal antibodies. Characterization tumor bed is clearly quite important, retention within
as to antibody isotype, level of purity, degree of the tumor bed may be equally or more important. The
contamination by other substances, stability, and other preparations of antibody fragments may diffuse more
pertinent physical/chemical characteristics is possible. quickly into the tumor nodule [2], but larger molecules
Thus, no insurmountable obstacles exist with respect to may be retained for a longer time within that same nod-
testing a wide variety of monoclonal antibodies in ule [58]. Therefore, at the level of these elementary
patients (Table 1) [74]. principles, there is much to be learned about the use of
454 Drug Immunoconjugates

CB4F10.11+AC4C2 99%

CB4F10.11+AC4C3 99%

MC6G10+140.72 97%

MC6G10+6PL4 93%

MC6G10+AC4C2 87%

MC6G10+AC4C6 92%

MC6G10+AC4C3 74%

MC6G10+DC3.24 40%

MC6G10+CD4F10.15 96.6%

MC6G10+CD4F10.11 49.2%

Figure 2. Antibody preparations to a single patient’s melanoma as seen on flow cytometry

BREAST TUMOR

C
O BR3
U 97% MPC 105 BR1 + BR3
N 100% MPC 185
T

BR-1
94% MPC 98

1 200
LOG GREEN FLUORESCENCE (VIABLE)

Figure 3. A two-antibody cocktail for breast cancer

these antibody preparations in clinical trials, and these to ten antibodies per patient to analyze heterogeneity
clinical trials should not yet be subject to hard and fast [4, 50, 73]. As shown in Fig. 2, the flow cytometry patterns
rules [58, 74]. On the basis of early assessments of the for each antibody may differ considerably when tested
heterogeneity of malignant melanoma [45], including against the patient’s cancer. By additive testing of the
studies of both melanoma cell lines and fresh biopsies individual antibodies, a cocktail of antibodies, consisting
of melanoma nodules, it became obvious that consider- of two to five components, can be created to cover all of
able antigenic heterogeneity exists. When the data were the cells in the tumor as assessed by flow cytometry and
analyzed in detail with a panel of antimelanoma mono- immunoperioxidase staining (Fig. 3).
clonal antibodies, each patient’s tumor had its own pattern Our data support the concept that tumor-cell heterogene-
of reactivity. As an extension of this approach, we have ity calls for the generation of antibodies and/or the use
generated antibodies for individual patients, making up of typing panels to prepare antibody cocktails containing
Malek Safa et al. 455

multiple components in an attempt to deliver antibody and its antibody after injection, is critical to an understanding
conjugated toxic substance to all the replicating cells in the of the pharmacokinetics in these antibody trials [3, 5, 73].
cancer [73]. This approach should substantially improve the Further, it is important to measure antiglobulin levels
ability to target toxic substances selectively to the cancer of initially and after treatment in conducting these clinical
an individual patient. Further analysis is needed with respect trials, since such antiglobulin responses may ultimately
to heterogeneity within the individual. Our preliminary data be important in the biodistribution, toxicity and efficacy
appear to indicate that this microheterogeneity is not strik- of an infused antibody [20, 62, 91–94].
ing, and it may be possible to cover it with a sufficiently
complex cocktail. However, the possibility of somatic
mutation and/or selective pressure in treatment leading to
further tumor-cell diversity in patients with metastatic dis-
Chemotherapeutic Drug
ease is real [23]. In lymphoma, there is evidence to indicate Immunoconjugates
a multiclonality; while it is not completely clear, some of
The rationale for conjugating antibody to chemothera-
this multiclonality may result from selective pressures
peutic drugs is fairly straightforward. These drugs have
induced by treatment with anti-idiotypic antibody [33, 54].
been used for many years in clinical studies. The spec-
trum of activity and the toxicology of these agents are
well understood. In addition, there is considerable evidence
Clinical-Laboratory Integration to support the belief that dose–response curves exist
with these drugs in the killing of tumor cells. Their lack
for Drug Immunoconjugate Trials of selectivity for cancer and the major problem of nor-
There are certain important principles in the design and mal tissue toxicity are well known. All of these factors
execution of clinical trials using monoclonal antibodies constitute a firm basis for attempting antibody delivery
[62–64, 72, 74]. To learn from these early trials it is to target the chemotherapeutic agent to the tumor site.
necessary to have an active and competent laboratory Surprisingly, very few clinical studies have been done
available for in vitro assessments. It is essential to dem- using antibody-targeted chemotherapy. Perhaps the reason
onstrate that the antibody reaches the tumor nodule. For relates to the lower specific activity seen in vitro with these
this, the technique of immunohistology using biopsy conjugates. Compared with immunotoxins, drug conju-
specimens subsequent to the infusion of antibody has gates have been less active in preclinical studies, and this
been quite valuable [36, 62, 92–94]. The distribution of finding has discouraged investigators with respect to the
antibody within the tumor nodule in contrast to surround- possible in vivo activity of such preparations [23, 33].
ing normal tissues can be assessed. This technique also gives
information on heterogeneity of antigen expression.
In vitro overlay with the treatment antibody can assess
Pre-Clinical Studies
saturation, and the use of other antibodies can define Preclinical studies using chemotherapeutic agents conju-
heterogeneity of antigen expression. Isotope-labeled gated to antibody have been reported for a variety of
antibody preparations may also be used to evaluate the drugs [10, 19, 22, 33, 40, 44, 47, 57, 79, 97, 100, 101,
biodistribution of antibody [12, 13, 35, 43, 46]. 102]. These studies have indicated that drug conjugates
In addition to these techniques using “fixed” tissue, can have in vitro and in vivo activity. The drugs most
cytofluorometry can be used to visualize individual cells often used have been vinca alkaloids, methotrexate,
with respect to antigen distribution, antibody localization, daunorubicin, and doxorubicin. In one study methotrex-
and saturation. When the tumor nodule is disaggregated ate was coupled to an IgM monoclonal antibody targeted
after in vivo treatment, one can determine with great preci- to a variety of tumors, including mouse teratocarcinoma
sion the level of antibody localization on individual cells [9]. Deposition of immunoconjugate in viable tumor and
and, by adding exogenous antibody, the degree of satura- lack of binding to antigen negative normal tissue was
tion on these individual cells [62, 92–94]. Thus, we have demonstrated in an in vivo animal model. Also, metho-
techniques to assess the delivery of antibody conjugates to trexate conjugated to monoclonal antibody has been com-
the tumor, as well as the degree of saturation of membrane pared with methotrexate-IgG (nonspecific antibody) and
antigens. In addition, actual determinations of heterogene- free methotrexate against EL4 lymphoma [34]. The
ity within individual tumor biopsies and between patients methotrexate-antibody conjugate was three and seven
can easily be made from these studies [73]. times more effective against EL4 lymphoma than metho-
The ability to measure antigen and antigen–antibody trexate-IgG and free methotrexate, respectively. Similarly,
complexes before and after treatment, and circulating paclitaxel–antibody conjugates afford selective toxicity
456 Drug Immunoconjugates

and are more cytotoxic in vitro than equimolar concentra- cells of antibody-phospholipase C (PLC) immunoconju-
tions of free paclitaxel or free paclitaxel plus free anti- gates. Then, liposomes containing daunorubicin are intro-
body [37]. In an in vivo model of xenografted tumors, duced which are specifically lysed at the tumor site by
systemic administration of paclitaxel–antibody conju- PLC to release their cytotoxic contents in the vicinity of
gates prevented tumor growth and prolonged survival of the tumor cells. For tumor xenografts in mice, the anti-
mice better than free drugs [37]. In another study, amin- body-PLC/liposome approach resulted in an inhibition of
opterin which is a more potent antifolate than methotrex- tumor growth without eradication of tumors [14]. In
ate, was coupled to a monoclonal antibody [84]. These another report [85], idarubicin, an analogue of daunorubi-
investigators demonstrated that administering leucovorin cin, was coupled to a CD19 antibody. A human pre-B-cell
48–72 h following a sublethal dose of the aminopterin– acute lymphoblastic leukemia cell line (NALM-6) was
antibody conjugate resulted in maintenance of the anti- implanted into nude mice and this immunoconjugate
tumor efficacy of the immunoconjugate and a significant was demonstrated to have substantial anti-tumor activity,
reduction in toxicity. Vindesine (a vinca alkaloid) has was non-toxic and was stable in vivo. It was proposed as
been conjugated with different anti-tumor antibodies, an excellent immunoconjugate for clinical trials.
such as anti-CEA, melanoma, osteosarcoma and others Another anti-tumor antibiotic agent, DU-257 which is
[78, 86–88]. Studies with vindesine-anti-CEA conjugates a duocarmycin derivative, was conjugated to a monoclonal
[88] indicate that they increase the therapeutic index of antibody (KM231) specifically reactive to GD3 antigen
vindesine by decreasing the toxicity and increasing the which is highly expressed on the surface of many malig-
specificity to tumor. Furthermore, evaluation of the con- nant tumors. The conjugate showed significant growth
jugate against non-CEA producing colon carcinoma inhibition of a human colorectal carcinoma cell line
xenograft and CEA-producing xenograft showed that sig- (SW1116) [106].
nificant retardation in the growth of tumor was observed Recently, two new antibody–drug conjugates have
only in CEA-producing tumor [101]. The anthracycline been developed as potential therapies for colon cancer
family of antitumor antibiotics, most notably doxorubicin and lung cancer. The antibodies were hMN-14 (labetu-
(DOX) and daunorubicin, has been used extensively for zumab), a non-internalizing humanized antibody that
drug targeting applications (102). BR96-DOX, which is binds to the carcinoembryonic antigen (CEA) expressed
chimeric with human IgG1, binds to Ley-related tumor- by many solid cancers, and hRS7, a rapidly internalizing
associated antigen expressed on most human carcinomas humanized antibody targeting epithelial glycoprotein-1
and on normal cells of the gastrointestinal tract of humans, (EGP-1) expressed on high amounts in certain human
dogs, and rats [107]. BR96-DOX induced cures of human cancers. The drug selected for conjugation was SN-38,
lung, breast and colon carcinomas in athymic mice and the active metabolite of irinotecan, a chemotherapeutic
rats [38, 107, 108], and syngeneic colon tumors in immu- agent used for the treatment of colorectal, lung, and other
nocompetent rats [100, 101]. In another study [116], the cancers. SN-38 cannot be systemically administered to
monoclonal antibody MSN-1 (IgM), which reacts with patients with cancer due to its toxicity and poor solubility.
endometrial adenocarcinomas, was combined with DOX, In an animal model of colon cancer with lung metastases,
and the complex showed significant antitumor activity in therapy with labetuzumab-SN-38 increased median sur-
athymic mice bearing endometrial carcinoma cell tumors. vival time two-fold to 73.5 days compared to non-treated
In one study, IMMU-110, an anti-CD74-DOX immuno- and non-targeting conjugate controls. Similar results
conjugate was cytotoxic to non-Burkitt and in Burkitt were observed in animals bearing human non-small cell
non-Hodgkin lymphoma cell lines. In addition, IMMU- lung cancer treated with hRS-SN-38 [28].
110 was therapeutic in drug-sensitive and drug-resistant In a different approach [17] investigators studied
non-Hodgkin lymphoma animal models, suggesting that maytansinoids. These drugs are 100–1,000 times more
antibody targeting can bypass the multi-drug resistant cytotoxic than common anticancer drugs. These investiga-
(MDR) drug efflux system that prevents free doxorubicin tors felt that the limitation of drug immunoconjugates is
from being therapeutic [11]. Anti-CD74-DOX immuno- that they act stoichiometrically, requiring much higher
conjugate (IMMU-110) is cytotoxic in non-Hodgkin’s intracellular concentrations to achieve the comparable
lymphoma models and overcomes MDR [11]. In another cytotoxicity as compared to a protein toxin where one mol-
study, IMMU-110 showed significant activity against ecule in the cytoplasm leads to cell death. Maytansine was
multiple myeloma cell lines and in SCID mouse models coupled to a monoclonal antibody via disulfide containing
of disseminated multiple myeloma [103]. A two-step linkers that are cleaved intracellularly to release the drug.
method has been developed for targeting cytotoxic drugs They demonstrated antigen specific cytotoxicity of cul-
into tumor cells [14]. It first involves the binding to tumor tured human cells and minimal systemic toxicity in mice
Malek Safa et al. 457

with an excellent pharmacokinetic profile. In one study, the mice treated with rituximab in combination with CMC-
immunoconjugate of b-76-8, a monoclonal antibody 544 had the longest median survival as compared to con-
against HM1.24/BST-2 (CD317) which is a cell surface trol mice [39]. Targeting CD20 and CD22 with rituximab
antigen overexpressed on multiple myeloma cells, with the in combination with CMC-544 results in improved anti-
analog of maytansine was shown to be active in a xenograft tumor activity against non-Hodgkin’s lymphoma pre-
model of human multiple myeloma [76]. Similarly, clinical models. In another study, antibody–drug
SAR3419, a novel humanized anti-CD19 antibody (huB4) conjugates to CD79 (anti-CD79-MCCDM1 and anti-
conjugated to a maytansine derivative (DM4) produced CD79b-MC-MMAF) showed activity in non-hodgkin
100% cures in two non-Hodgkin lymphoma cell lines [1]. lymphoma cell lines [80].
These studies and many others in preclinical models using
antibody conjugated to chemotherapeutic agents lend sup-
port to the idea that drug-conjugated antibodies may allow
Clinical Trials
better targeting of chemotherapeutic agents to the tumor One of the first reports using drug-labeled antibody used
site [7, 16, 19, 23, 30, 41, 42, 64–68, 77, 105, 109]. antibody 791T/36 conjugated to methotrexate (MTX) [7].
Src proto-oncogene family protein tyrosine kinases Initial studies with this conjugate demonstrated that meth-
(PTKs) play a key role in cell function and attempts have otrexate conjugation did not alter the pharmacokinetics
been made to develop agents that specifically inhibit Src- or tumor localization indices for the antibody labeled with
131
family PTKs. In one report [110], a general PTK inhibi- I [25]. In a different study [8], 16 patients with primary
tor, Genistein, that inhibits all members of the Src PTK colorectal cancer were injected intravenously with I131-
family was used to target to cancer cells. Genistein is an labeled 791T-36-MTX and imaged using a gamma cam-
isoflavone derived from fermentation broth of era after 48–72 h. Assays of tissue radioactivity showed
Pseudomonas. It is also a natural occurring tyrosine high tumor uptake of drug-antibody conjugate in 13 of 15
kinase inhibitor present in soybeans. Genistein inhibits tumor specimens. Also, the monoclonal antibody KS1/4,
purified Lck kinase from human lymphoid cells at micro- which targets an antigen expressed on epithelial malig-
molar concentrations. These investigators conjugated nancies and some normal epithelial cells were conjugated
Genistein to an anti-CD19 monoclonal antibody. An anti- to methotrexate [26]. Eleven patients with advanced
CD19 antibody was chosen because the CD19 receptor is non-small cell carcinoma of the lung were treated up to a
not shed from cells, it is internalized upon association maximum tolerated antibody dose of 1,750 mg/m2 with a
with antibody and is physically and functionally associ- cumulative dose of methotrexate of 40 mg/m2. Toxicities
ated with the Src protooncogene family PTKs including in this study included fever, anorexia, nausea, vomiting,
Lck. Human acute lymphoblastic leukemia of the pre-B diarrhea, abdominal pain, guaiac positive stools and
cell variety was treated in a severe combined immunode- hypoalbuminemia. Two patients had episodes of aseptic
ficient mouse model with this immunoconjugate. These meningitis. One patient had a greater than 50% decrease
investigators demonstrated that the immunoconjugate of two lung nodules. Post-treatment tumor biopsies
bound with high affinity to the leukemia cells, inhibited demonstrated binding of the immunoconjugate to tumor
selectively the CD19-associated tyrosine kinases, and epithelial cells. Some of the gastrointestinal toxicities
triggered rapid apoptotic cell death. Furthermore, at less may also have been secondary to binding of the immuno-
than one tenth the maximum tolerated dose, greater than conjugate to a similar antigen on gastrointestinal epithe-
99.99% of the leukemia cells were killed, which led to lial cells.
100% long-term event-free survival in these animals. In Mylotarg (gemtuzumab ozogamicin, previously known
another study [55], LL2, an anti-CD22 monoclonal anti- as CMA-676) is an FDA approved antibody-targeted
body against B-cell lymphoma, was covalently linked to chemotherapy agent consisting of the humanized murine
the amphibian ribonuclease, onconase, a member of the CD33 antibody to which the calicheamicin γ1 derivative
pancreatic Rnase A superfamily. The LL2-onconase con- is attached [111]. It is assumed that binding of Mylotarg
jugate showed significant antitumor activity against dis- to the CD33 antigen results in internalization followed
seminated Daudi lymphoma in mice with severe combined by the release of the potent antitumor antibiotic cali-
immunodeficiency disease. Furthermore, the life span of cheamicin and subsequent induction of cell death. In one
these mice was increased 135% as compared to controls. study [111], 122 patients with relapsed acute myeloge-
Similarly, CMC-544, an IgG4 CD-22-humanized tar- nous leukemia (AML) were treated with Mylotarg as a
geted immunoconjugate of the antitumor antibiotic cali- single infusion at a dose of 9 mg/m2. Each patient received
cheamicin, showed activity against a panel of non-Hodgkin two Mylotarg doses with at least 14 days between the
lymphoma cell lines. In the same study, lymphoma-bearing doses. Within 3–6 h after infusion, near complete satura-
458 Drug Immunoconjugates

tion of CD33 antigen sited by Mylotarg was reached for administered over a 3-week period without alopecia or
AML blasts. Furthermore, Mylotarg induced dose- severe bone marrow depression. In several of these
dependent apoptosis in myeloid cells in vitro. In a multi- patients, antibody delivery was confirmed by biopsy of
center trial [98], 142 patients with CD-33 positive AML skin and analysis of cytologically positive pleural fluid.
in first relapse were treated with two doses of Mylotarg. Persistence of antibody conjugates for up to 1 week
Thirty percent of patients achieved remission with a post-treatment was noted with doxorubicin doses of
favorable safety profile. Based on these results, Mylotarg 50 mg borne on approximately 300 mg of antibody.
was approved for the treatment of relapsed CD-33 posi- These doxorubicin–monoclonal antibody cocktail
tive acute myeloid leukemia. immunoconjugates were used in 24 patients. Mild toxic
In another study, 48 patients with follicular (FL) or reactions were seen in 17/24 (fever, chills, pruritis, and
diffuse large B-cell lymphoma (DLBCL) were treated skin rash) after the administration of these drug immu-
with CMC-544 (antiCD22 conjugated to calicheamicin). noconjugates. In several patients there was limited anti-
The objective response rate was 69% and 33% for patients genic drift among the sequential biopsies within the
with FL and DLBCL, respectively [27]. In another dose same patient over time. Five minor responses were seen
escalation study of AVE9633, an antiCD33-Maytansinoid with these conjugates. Two patients with breast cancer
immunoconjugate, antileukemia activity was observed in had isolated improvement in skin ulcerations, and three
patients with relapsed or refractory CD-33 positive additional patients with other types of cancers had minor
acute myeloid leukemia [48]. In a phase I study, huN9- responses. None of these responses were sufficient to
01-DM1 (BB-10901) which is a humanized monoclonal reach a partial response (50% reduction in mean tumor
antibody–maytansinoid immunoconjugate that binds diameters) by standard oncological criteria.
with high affinity to CD56, was shown to be safe and Toward the end of these studies and at the higher
clinically active in patients with relapsed or refractory doses of doxorubicin, toxicities were noted in these
CD56 positive multiple myeloma [15]. In addition, in a patients. Urine color turned red, and alopecia with bone
phase II trial, huN901-DM1 showed activity in patients marrow suppression was noted. On reexamination of the
with CD-56 positive relapsed small cell cancer [51]. doxorubicin procured from Adria Laboratories, it was
Other studies further support the possibility of clini- noted that the preparation method for the doxorubicin
cal trials with drug immunoconjugates [18, 19, 25, 30, had changed. A stabilizer (methylparaben) had been
32–34, 104]. The rationale for such studies has been added to the preparation (RDF-Adriamycin), which
previously described [61–67, 72–74] and the dose- likely changed the stability of the doxorubicin immuno-
delivery curves for tissue penetration and antigen satu- conjugates. This change occurred near the end of the
ration have been fully described [62, 69, 73–75]. Issues study, and the clinical studies were terminated to deter-
of antibody specificity, antigen heterogeneity, optimal mine a better method for manufacturing doxorubicin
linkers, drug selection, and specific activity of the immunoconjugates in light of the changed chemistry
immunoconjugate all remain under active investigation. [23, 24, 56, 69, 70, 73].
We have utilized cocktails of antibodies custom tai- A subsequent study was carried out using mitomycin-
lored to individual patient tumors [68, 71, 73, 75]. These C immunoconjugates [75]. Nineteen patients were
studies have utilized a biopsy of the cancer from each treated with mitomycin-C conjugated to similar cock-
patient and either the generation of new antibodies to tails of monoclonal antibodies. Thrombocytopenia at
that patient’s tumor or the use of existing panels of anti- the 60 mg (mitomycin) dose in this protocol was dose-
bodies to type the patient’s tumor [3, 50]. From these limiting. The anti-mouse antibody titers were lower in
two methods, cocktails of two to six antibodies were the mitomycin-C compared to the doxorubicin-treated
generated and conjugated to doxorubicin [56, 68–71] or patients [5]. No responses were seen with mitomycin-C
mitomycin-C [75]. The immunoglobulin-to-doxorubicin immunoconjugates, although several patients had less
ratios were in the range of 1:40 to 1:60. This level of tumor-related pain after treatment [73].
coupling did not alter the antibody’s in vitro immunore- The role of drug resistance in cancer treatment is
activity, and excellent in vitro cytotoxicity was observed another issue to be considered. The current widely held
on antigen-positive cell lines compared to antigen-nega- opinion is that tumor cells, once resistant, cannot effec-
tive controls. The spectrum of clinical toxicities was tively be treated with the same drug. A variety of
strikingly different for doxorubicin conjugated to anti- approaches using mechanisms to overcome membrane
body as compared with free doxorubicin [68, 70]. More resistance have been suggested. However, antibody-
than 1 g of doxorubicin on up to 5 g of antibody was mediated delivery may be another such mechanism, and
Malek Safa et al. 459

it is unclear whether doxorubicin-resistant tumor cells cells in ways that will avoid the undesirable side effects
will be resistant to doxorubicin conjugates if delivered in on normal tissues. Given these considerations, it is prob-
appropriate quantities. In vitro and animal model data ably no longer necessary to distinguish between drugs
have confirmed sensitivity to doxorubicin conjugate and toxins. Perhaps we should speak of strong and weak
where the cell line was resistant to free drug [19, 22–24]. drugs (chemicals), in that toxin molecules are simply
These and many other issues will be approachable through biological chemicals with a higher degree of toxicity
the use of drug–antibody conjugates. Other studies com- than the chemicals we commonly identify as drugs. This
paring drug and toxin conjugates are of interest in regard may, however, only partially be the case, since most tox-
to the issue of specific activity [23, 25]. While the immu- ins are complex biologicals and most chemotherapeutic
notoxins had a higher specific cytolytic capability in vitro, drugs are relatively simple chemicals. Such differences in
and may be most useful where target cells express low size and structure may profoundly affect in vivo trafficking
levels of antigens, drug conjugates may be preferable and immunogenicity.
when target cells express a high density of antigens or With the advent of monoclonal antibodies and the
when large doses of conjugate are needed to penetrate possibility of delivering toxic substances selectively to
tumors. In these situations, higher-specific-activity conju- cancer cells, many toxic chemicals previously rejected
gates (toxins) may be too toxic for clinical use. as chemotherapeutic agents may now be recognized for
use in association with antibody in an immunoconjugate
[99]. Pro-drug strategies and methods of in vivo activation
may add further selectivity [6]. Thus, cancer treatment
Conclusions can now enter a new era, since selective delivery offers
Our studies, as well as many similar studies, clearly the hope of increased specific activity against the cancer
indicate the potential for the new selective approach to with less toxicity to the patient. With FDA approval of
cancer treatment. Selective and effective new anti-can- Mylotarg, clinical proof of principle for drug immuno-
cer approaches are needed in the systemic treatment of conjugates was achieved [74]. We are now in an era
human malignancies. Many very toxic molecules are where costs [58, 68, 73], not technology, will now be the
well known to medical science (Table 2). Of key impor- limiting factor in developmental therapeutics [59].
tance is the delivery of these toxic substances to tumor

Table 2. Immunoconjugates with cytotoxic agents


References
1. Aboukameel A., Goustin A.S., Al-Khatib A., et al. Blood (ASH
PROTEIN TOXINS AND CYTOTOXINS Meeting Abstracts) 2007;110: Abstract 2339.
Abrin 2. Andrew AM, Pim MV, Perkins AC, Baldwin RW. Comparative
Ricin imaging and biodistribution studies with an anti-CEA monoclonal
Calicheamicin antibody and its F(ab)2 and Fab fragments in mice with colon
Diphtheria carcinoma xenografts. Eur J Nucl Med 1986;12:168–175.
Duocarmycins 3. Avner B, Avner BP, Gaydos B, et al. Characterization of a method
Gelonin using viable human target cells as the solid phase in a cell concen-
Hematoporphyrin tration fluorescence immunoassay (CCFIA) for screening of
Ribonuclease monoclonal antibodies and hybridoma supernatants. J Immunol
Purothionine Methods 1988;113:123–135.
Pseudomonas exotoxin 4. Avner BP, Liao SK, Avner B, et al. Therapeutic murine monoclonal
Alpha-amanitin antibodies developed for individual cancer patients. J Biol
CHEMOTHERAPEUTIC DRUGS Response Modif 1989;8(1):25–36.
Vindesine 5. Avner B, Swindell L, Sharp E, et al. Evaluation and clinical relevance
Methotrexate of patient immune responses to intravenous therapy with murine
Daunorubicin monoclonal antibodies conjugated to adriamycin. Mol Biother
Doxorubicin 1991;3(1):14–21.
Idarubicin 6. Bagshawe KD. Antibody directed enzymes activate prodrugs at
Mitomycin-C tumor site. Order SE, ed. Antibody Immunoconj. Radiopharm.
Paclitaxel 1990;3(1):60.
Various new drugs with high toxicity profiles 7. Ballantyne KC, Perkins AC, Pimm MV, et al. Localization of
BIOLOGIC AGENTS monoclonal antibody-drug conjugate 791T/36-methotrexate in
Interferon colorectal cancer. STS Abstr 1986;No.88.
Tumor necrosis factor 8. Ballantyne KC, Perkins AC, Pimm MV, Hardcastle JD, et al.
Cobra venom factor Biodistribution of a monoclonal antibody-methotrexate conjugate
460 Drug Immunoconjugates

(791T/36-MTX) in patients with colorectal cancer. Int J Cancer 28. FDA NEWS Oct. 29, 2007; Vol. 5 No. 212.
Suppl 1988;2:103–108. 29. Fidler IJ, Poste G. The cellular heterogeniety of malignant neo-
9. Ballou B, Jaffe R, Persiani S, et al. Tissue localization of metho- plasms: implications for adjuvant chemotherapy. Semin Oncol
trexate-monoclonal-IgM immunoconjugates: anti-SSEA-1 and 1985;12:207–221.
MOPC 104E in mouse teratocarcinomas and normal tissues. 30. Foon KA, Bernhard MI, Oldham RK. Monoclonal antibody ther-
Cancer Immunol Immunother 1992;35:251–256. apy: assessment by animal tumor models. J Biol Response Modif
10. Belles-Isles M, Page M. In vitro activity of daunomycin anti-alpha- 1982;1:277–304.
fetoprotein conjugate on mouse hepatoma cell. Cancer (Phila) 31. Foon KA, Bunn PA, Schroff RW, et al. Monoclonal antibody ther-
1980;41:841–845. apy of chronic lymphocytic leukemia and cutaneous T-cell lym-
11. Blumenthal R, Stein R, Michell R, et al. Anti-CD74-doxorubicin phoma: preliminary observations. In: Boss BD, Langman RE,
immunoconuugate (IMMU-110) is cytotoxic in non-Hodgki’s Trowbridge IS, Dudlbecco R, eds. Monoclonal Antibody and
lymphoma (NHL) models and overcomes MDR. J Clin Oncol 2006 Cancer. New York: Academic, 1983:39–52.
ASCO Annual Meeting Proceedings Part 1, Vol. 24, June 20. 32. Ford CH, Newman CE, Johnson JR, et al. Localization and toxicity
12. Carrasquillo JA, Abrams PG, Schroff R, et al. Effect of antibody study of a vindesine-anti-CEA conjugate in patients with advanced
dose on the imaging and biodistribution of indium-111 9.2.27 cancer. Br J Cancer 1983;47:35–42.
anti-melanoma monoclonal antibody. J Nucl Med 1988;29(1): 33. Ghose T, Blair AH. Antibody linked cytotoxic agents in the treat-
39–47. ment of cancer: current status and future prospects. J Natl Cancer
13. Carrasquillo JA, Bunn PA, Kennan AM, et al. Radioimmunodetection Inst 1978;61:657–676.
of cutaneous T-cell lymphoma with 111In-T101 monoclonal antibody. 34. Ghose T, Blair AH, Uadia P, et al. Antibodies as carriers of cancer
N Engl J Med 1986;315:673–680. chemotherapeutic agents. Ann NY Acad Sci 1985;446:213–227.
14. Carter G, White P, Fernie M, Carr FJ, et al. Enhanced antitumour 35. Goldenberg DM, DeLand FH. History and status of tumor imaging
effect of liposomal daunorubicin using antibody-phospholipase C with radiolabled antibodies. J Biol Response Modif 1982;1:121–136.
conjugates or fusion protein. Int J Oncol 13:819–825, 1998. 36. Goodman GE, Beauimer P, Hellstrom I, et al. Phase I trial of murine
15. Chanan-Khan A., Jagannath S., Fram R., et al. Phase I study of monoclonal antibodies in patients with advanced melanoma. J Clin
huN901-DM1 (BB-10901) in patients with relapsed and Oncol 1984;3:340–352.
relapsed/refractory CD-56 positive multiple myeloma. Blood 37. Guillemard V, Saragovi HU. Taxane-antibody conjugates afford
Nov.2007;110:11. potent cytotoxicity, enhanced solubility, and tumor target selectiv-
16. Chari RJ, Gross JL, Goldmacher VS, et al. Conjugates of mono- ity. Cancer Res. 2001;61:694–699.
clonal antibodies and cytotoxic macrolide drugs: potent, target 38. Hellstrom I, Hellstrom KE, Siegall CB, Trail PA. Immunoconjugates
specific antibody-drug conjugates. Antibody Immunoconj and immunotoxins for therapy of carcinomas. In August JT,
Radiopharm 1990;3(1):64. Anders MW, Murad F, Coyle JT, eds., Advances in Pharmacology,
17. Chari VJ, Martell BA, Gross JL, et al. Immunoconjugates contain- Vol. 33, edition 1. San Diego, CA: Academic; 1995:349–388.
ing novel maytansinoids: promising anticancer drugs. Cancer Res 39. Hernandez-Ilizaliturri F., Devineni S., Cruczman M., et al.
1992;52:127–131. Targeting CD20 and CD22 with rituximab in combination with
18. Dillman RO. Monoclonal antibodies in the treatment of cancer. CMC-544 results in improved anti-tumor activity against non-
Crit Rev Hematol Oncol 1984;1:357–386. Hodgkin’s lymphoma pre-clinical models. Blood (ASH Meeting
19. Dillman RO. Monoclonal antibodies for treating cancer. Ann Abstracts) 2005;106:Abstract 1473.
Intern Med 1989;111:592–603. 40. Hurwitz E, Levy R, Maron, et al. The covalent binding of dauno-
20. Dillman RO. Human antimouse and antiglobulin responses to mycin and Adriamycin to antibodies with retention of both drug
monoclonal antibodies. Antibody Immunocon Radiopharm antibody activities. Cancer Res 1975;3:1175–1181.
1990;3(1):1–16. 41. Hwang KM, Foon KA, Cheung PH, et al. Selective antitumor effect
21. Dillman RO, Shawler DL, Sobol RE. Murine monoclonal antibody on L-10 hepato-carcinoma cells of a potent immunoconjugate
therapy in two patients with chronic lymphocytic leukemia. Blood composed of the A chain for abrin and monoclonal antibody to a
1982;59:1036–1046. hepatoma-associated antigen. Cancer Res 1984;44:4578–4586.
22. Dillman RO, Shawler DL, Johnson DE, et al. Preclinical trials with 42. Hwang KM, Foon KA, Cheung PH, et al. Selective antitumor
combinations and conjugates of T101 and doxorubicin. Cancer effect of a potent immunoconjugate composed of the A chain of
Res 1986;46:4886–4891. abrin and monoclonal antibody to a hepatola associate antigen.
23. Dillman RO, Johnson DE, Shawler DL. Comparisons of drug and Cancer Res 1984b;44:4578–4586.
toxin immunoconjugates. Antibody Immunocon Radiopharm 43. Hwang KM, Keenan AM, Frincke J, et al. Dynamic interaction of
1988;1:65–77. 111 indium-labeled monoclonal antibodies with surface of solid
24. Dillman RO, Johnson DE, Shawler DL, Koziol JA. Superiority of tumors visualized in vivo by external scintigraphy. J Nat Cancer
an acid-labile daunorubicin-monoclonal antibody immunoconju- Inst 1986;76:849–855.
gate compared to free drug. Cancer Res 1988;48:6097–6102. 44. Johnson JR, Ford CMG, Newman E, et al. A vindesine-anti-CEA
25. Embleton MJ. Drug targeting by monoclonal antibodies. Br J conjugate cytotoxic for human cancer cell in vitro. Br J Cancer
Cancer 1987;55:227–231. 1981;44:472–477.
26. Elias DJ, Kline LE, Robbins BA, et al. Monoclonal antibody 45. Krizan Z, Murray JL, Hersh EM, et al. Increased labeling of human
KS1/4-methotrexate immunoconjugate studies in non-small cell melanoma cells in vitro using combinations of monoclonal anti-
lung carcinoma. Am J Resp Crit Care Med 1994;150:1114–1122. bodies recognizing separate cell surface antibenic determinants.
27. Fayad L., Patel H., Advani A., et al. Clinical activity of the immu- Cancer Res 1985;45:4904–4909.
noconjugate CMC-544 in B-cell malignancies: preliminary report 46. Larson SM, Brown JP, Wright PW, et al. Imaging of melanoma
of the expanded maximum tolerated dose (MTD) cohort of a phase with I-labeled monoclonal antibodies. J Nucl Med 1983;24:
1 study. Blood (ASH Annual Meeting Abstracts) 2006;108: 123–129.
Abstract 2711.
Malek Safa et al. 461

47. Lee FH, Hwang KM. Antibodies as specific carriers for chemo- 66. Oldham RK. Immunoconjugates: drugs and toxins. In: Oldham
therapeutic agents. Cancer Chemother Pharmacol 1979;3:17–25. RK, ed., Principles of Cancer Biotherapy. New York: Raven Press,
48. Legrand O., Vidriales M., Marie J.P., et al. An open label dose 1987:319–335.
escalation study of AVE9633 administered as a single agent by 67. Oldham RK. Monoclonal antibody therapy. In: Chiao JW, ed.,
intravenous infusion weekly for 2 weeks in 4-week cycle to Biological Response Modifiers and Cancer Research, Vol. 40.
patients with relapsed or refractory CD-33 positive acute myeloid New York: Marcel Dekker, 1988:3–16.
leukemia. Blood (ASH Annual Meeting Abstracts) 2007;110: 68. Oldham RK, Lewis M, Orr DW, et al. Individually specified drug
Abstract 1850. immunoconjugates in cancer treatment. Imperial Cancer Research
49. Levy R. Biologicals for cancer treatment: monoclonal antibodies. Conference, England, 1990.
Hosp Pract 1985;November 15:67–92. 69. Oldham RK. Who pays for new drugs? Nature 1988;332(28):795.
50. Liao SK, Meranda C, Avner BP, et al. Immunohistochemical phe- 70. Oldham RK, Lewis M, Orr DW, et al. Adriamycin custom-tailored
notyping of human solid tumors with monoclonal antibodies in immunoconjugates in the treatment of human malignancies. Mol
devising biotherapeutic strategies. Cancer Immunol Immunother Biother 1988;1(2):103–113.
1989;28:77–86. 71. Oldham RK, Lewis M, Orr DW, et al. Individually specified drug
51. McCann J., Fossella F.M., Fram R., et al. Phase II trial of huN9- immunoconjugates in cancer treatment. In: Ceriani RL, ed., Breast
01-DM1 in patients with relapsed small cell lung cancer and Cancer Immunodiagnosis and Immunotherapy Proceeding. Proceedings
CD56-positive small cell carcinoma. JCO, 2007 ASCO Annual of the 12th World Congress of Obstetrics and Gynecology, 1990.
Meeting Proceedings Part I, Vol. 25, No. 18s, 2007: 18084 72. Oldham RK. Monoclonal antibodies. In: Nathanson L, ed. Management
52. McLauglin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric of Advanced Melanoma. Contemporary Issues in Clinical Oncology.
anti-CD20 monoclonal antibody therapy for relapsed indolent New York: Churchill Livingstone, 1986:195–207.
lymphoma: half of patients respond to a four-dose treatment program. 73. Oldham RK. Custom tailored drug immunoconjugates in cancer
J Clin Oncol 16:2825–2833, 1998. therapy. Mol Biother 1991;3(3):148–162.
53. Miller RA, Levy R. Response of cutaneous T-cell lymphoma to ther- 74. Oldham RD, Dillman RO. Monoclonal antibodies in cancer therapy;
apy with hybridoma monoclonal antibody. Lancet 1981;2:226–230. twenty-five years of progress, JCO 2008;26(11)1774-1777.
54. Miller RA, Maloney DG, Warnke R, et al. Treatment of B-cell 75. Orr DW, Oldham RK, Lewis M, et al. Phase I trial of mitomycin-c
lymphoma with monoclonal anti-idiotype antibody. N Engl J Med immunoconjugate cocktails in human malignancies. Mol Biother
1982;306:517–522. 1989;1(4):229–240.
55. Newton DL, Hansen HJ, Mikulski SM, Rybak SM, et al. Potent 76. Ozaki S.,Tomura T., Matsumoto T. et al. Anti-myeloma activity of
and specific antitumor effects of an anti-CD22-targeted cytotoxic the Maytanisoid immunoconjugate of internalizing human mono-
ribonuclease: potential for the treatment of non-Hodgkin clonal antibody specific for HM1.24/BST2 (CD317). Blood (ASH
lymphoma. Blood. 2001;97:528–535. Meeting Abstracts), Nov 2005;106:3388.
56. Ogden JR, Leung K, Kundra SA, et al. Immunoconjugates of dox- 77. Pavanasasivam G, Pearson JW, Bohn W, et al. Immunotoxins to a
orubicin and murine antihuman breast carcinoma monoclonal anti- human melanoma asociated antigen: comparison of gelonin with ricin
bodies prepared via an n-hydroxysuccinimide active ester and other A-chain conjugates. Cancer Res 1987;47:3169–3173.
intermediate of cis-aconityl-doxorubicin: preparation and in vitro 78. Philpott GW, Gass EH, Panker CW. Affinity cytotoxicity with an
cytotoxicity. Mol Biother 1989;1(3):170–174. alcohol dehydrogenase-antibody conjugate and allyl alcohol.
57. Ohkawa K, Hibi N, Tsukada Y. Evaluation of a conjugate of puri- Cancer Res 1979;39:2084–2087.
fied antibodies against human AFP-dextran-daunorubicin to 79. Pietersz GA, Smyth MJ, Kanellos J. Preclinical and clinical studies
human AFP-producing yolk sac tumor cell lines. Cancer Immunol with a variety of immunoconjugates. Antibody Immunocon
Immunother 1986;22:81–86. Radiopharm 1988;1:79–103.
58. Oldham RK. Monoclonal antibodies in cancer therapy. J Clin 80. Polson A., Yu S.F., Ebens A., et al. Antibody-drug conjugates
Oncol 1983;1:582–590. targeted to CD79 for the treatment of non-Hodgkin lymphoma.
59. Oldham RK. Biologicals: new horizons in pharmaceutical devel- Blood, 15 July 2007;110(2):616–623.
opment. J Biol Response Modif 1983;2:199–206. 81. Raso V, Raso J, Basala M, Schlossman S. Monoclonal antibody-
60. Oldham RK. Biologicals and biological response modifiers: fourth ricin A chain conjugate selectivity cytotoxic for cells bearing the
modality of cancer treatment. Cancer Treat Rep 1984;68:221–232. common acute lymphoblastic leukemia antigen. Cancer Res
61. Oldham RK. Therapeutic monoclonal antibodies: effects of tumor 1980;42:457–464.
cell heterogeniety. In: Present Status of Nontoxic Concepts in 82. Riethmuller G, Holz E, Schlimok G, Hirche H, et al. Monoclonal
Cancer Therapy, Cancer Treatment Symposium (Germany), S antibody therapy for resected Duke’s C colorectal cancer: seven-
Karger, 1986. year outcome of a multicenter randomized trial. J Clin Oncol
62. Oldham RK, Foon KA, Morgan AC, et al. Monoclonal antibody 1998,16:1788–1794.
therapy of malignant melanoma: in vivo localization in cutaneous 83. Ritz J, Schlossman SF. Utilization of monoclonal antibodies in
metastasis after intravenous administration. J Clin Oncol treatment of leukemia and lymphoma. Blood 1982;59:1–11.
1984;2:1235–1242. 84. Rowland AJ and Pietersz GA. Reduction in the toxicity of aminop-
63. Oldham RK, Morgan AC, Woodhouse CS, et al. Monoclonal anti- terin-monoclonal-antibody conjugates by leucovorin. Cancer
bodies in the treatment of cancer: preliminary observations and Immunol Immunother 1994;39:135–139.
future prospects. Med Oncol Tumor Pharmocol 1984;1(2):51–62. 85. Rowland AJ, Pietersz GA and McKenzie IFC. Preclinical investi-
64. Oldham RK. Antibody-drug and antibody toxin conjugates. In: gation of the antitumour effects of anti-CD19-idarubicin immuno-
Reif AE, Mitchell MS, eds., Immunity to Cancer. New York: conjugates. Cancer Immunol Immunother 1993;37:195–202.
Academic, 1985:575–586. 86. Rowland GF, Corvalan JRF, Axton CA, et al. Suppression of
65. Oldham RK. Monoclonal antibodies: does sufficient selectivity to growth of human colorectal tumor in nude mice by vindesine-
cancer cells exist for therapeutic application? J Biol Response monoclonal antibody CEA conjugates. Protides Biol Fluids
Modif 1987;6:227–234. 1984;31:783–786.
462 Drug Immunoconjugates

87. Rowland GF, Axton CA, Baldwin RW, et al. Antitumor proper- 101. Smith TW. Antitumor properties of vindesine-monoclonal anti-
ties of vindesine-monoclonal antibody conjugates. Cancer Immun body conjugates. Cancer Immunol Immunother 1985;19:1–7.
Immunotherapy 1985;19:1–7. 102. Stastny JJ and Das Gupta TK. The use of daunomycin-antibody
88. Rowland GF, Simmonds RG, Grove VA, Smith W, et al. Drug immunoconjugates in managing soft tissue sarcomas: nude
localization and growth inhibition studies of vindesine-monoclo- mouse xenograft model. Cancer Res 53:5740–5744, 1993.
nal anti-CEA conjugates in human xenograft. Cancer Immun 103. Stein, R. ASCO Annual Meeting Proceedings, 2004;22(14S):6535.
Immunother 1986;21:183–187. 104. Takahashi T, Yamaguchi T, Noguchi A, et al. Clinical trial of mono-
89. Schilsky RL. Tumor cell heterogeniety: implications for clinical clonal antibody-drug conjugate, A7-NCS, for 70 patients with col-
practice. Semin Oncol 1985;12:203–206. orectal cancer. Antibody Immunocon Radiopharm 1990;3(1):60.
90. Schnipper LE. Clinical implications of tumor cell heterogeniety. 105. Thorpe PE, Ross WCJ. The preparation and cytotoxic properties
N Engl Med 1986;314:1423–1431. of antibody-toxin conjugated. Immunol Rev 1982;62:119.
91. Schroff RW, Farrell MM, Klein RA, et al. T65 antigen modula- 106. Toshiyuki S, Nagamura S, Saito H, Yamasaki M, et al. Synthesis
tion in a phase I monoclonal antibody trial with chronic lympho- of a novel duocarmycin derivative DU-257 and its application to
cytic leukemia patients. J Immunol 1984;133:1641–1648. immunoconjugate using poly(ethylene glycol)-dipeptidyl linker
92. Schroff RW, Foon KA, Beatty SM, et al. Human anti-murine capable of tumor specific activation. Bioorgan Medicin Chem 8
immunoglobulin responses in patients receiving monoclonal anti- (2000);2175–2184.
body therapy. Cancer Res 1985;45:879–885. 107. Trail PA, Willner D, Lasch SJ, Hellstrom KE, et al. Cure of xeno-
93. Schroff RW, Morgan AC, Woodhouse CS, et al. Monoclonal anti- grafted human carcinomas by BR96-doxorubicin immunoconju-
body therapy in malignant melanoma: factors effecting in vivo gates. Science 1993,261: 212–215.
localization. J Biol Response Modif 1987. 108. Trail PA, Willner D, Hellstrom KE. Site-directed delivery of anthra-
94. Schroff RW, Woodhouse CS, Foon KA, et al. Intratumor localiza- cyclines for cancer therapy. Drug Dev Res 1995,34:196–209.
tion of monoclonal antibody in patients with melanoma treated 109. Tsukada Y, Hurwitz E, Kashi R, et al. Chemotherapy by intrave-
with antibody to a 250Kd melanoma associated antigen. JNCI nous administration of conjugates of daunomycin with monoclo-
1985;74:299–306. nal conventional anti-rat-alpha-fetoprotein antibodies. Proc Natl
95. Sears HF, Herlym D, Steplewski Z, Koprowski H. Effects of Acad Sci USA 1982;79:7896–7899.
monoclonal antibody immunotherapy in patients with gastroin- 110. Uckun FM, Evans WE, Forsyth CJ, et al. Biotherapy of B-cell
testinal adenocarcinoma. J Biol Response Modif 1984;3: precursor leukemia by targeting genistein to CD19-associated
138–150. tyrosine kinases. Science 267:886–891, 1995.
96. Sears HF, Mattis J, Herlyn D, et al. Phase-1 clinical trial of mono- 111. van der Velden VHJ, te Marvelde JG, Hoogeveen PG, van
clonal antibody in treatment of gastrointestinal tumors. Lancet Dongen JJM, et al. Targeting of the CD33-calicheamicin immu-
1982;1:762–765. noconjugate Mylotarg (CMA-676) in acute myeloid leukemia: in
97. Shawler DL, Johnson DE, Sweet MD, et al. Preclinical trials vivo and in vitro saturation and internalization by leukemic and
using an immunoconjugate of T101 and methotrexate in an athymic normal myeloid cells. Blood. 2001;97:3197–3204.
mouse/human T-cell tumor model. J Biol Response Modif 112. Vogel C, Cobleigh MA, Tripathy D, Stewart SJ, et al. First-line,
1988;7:608–618. single-agent Herceptin (trastuzumab) in metastatic breast cancer:
98. Sievers EL, Larson RA, Stadtmauer EA, Appelbaum FR, et al. a preliminary report. Eur J Cancer Jan 2001;37 Suppl 1:S 25–29.
Efficacy and Safety of Gemtuzumab Ozogamicin in Patients 113. Vogel C-W, ed. Immunoconjugates: Antibody Conjugates in
With CD33-Positive Acute Myeloid Leukemia in First Relapse. Radioimaging and Therapy of Cancer. New York: Oxford
J Clin Oncol 19:3244–3254, 2001. University Press, 1987.
99. Sivam G, Comezoglu FT, Vrudhula VM, et al. Immunoconjugates 114. Von Hoff DD. Implications of tumor cell heterogeneity for in
of a small molecule protein synthesis inhibitor (trichothecene) – vitro drug sensitivity testing. Semin Oncol 1985;12:327–331.
an update. Antibody Immunocon Radiopharm 1990;3(1):63. 115. Yarbro JW. Introduction: tumor heterogeniety and the new
100. Sjogren HO, Isaksson M, Willner D, Trail PA, et al. Antitumor biology. Semin Oncol 1985;12:201–202.
activity of carcinoma- reactive BR96-doxorubicin conjugate 116. Yoshibumi K, Tsukazaki K, Kubushiro K, Nozawa S, et al.
against human carcinomas in athymic mice and rats and synge- Selective cytotoxicity of adriamycin immunoconjugate antibody
neic rat carcinomas in immunocompetent rats. Cancer Res MSN-1 to endometrial adenocarcinoma in vitro and in vivo.
1997;57:4530–4536. Oncol Reports 2000;7:1099–1106.
13 Targeted radionuclide therapy of cancer
JOHN M. PAGEL, OTTO C. BOERMAN, HAZEL B. BREITZ AND RUBY F. MEREDITH

This chapter is an update of the former Chapter 13 Their fast blood clearance could lead to high tumor-
“Radiolabeled Monoclonal Antibodies for Management to-background ratios shortly after administration of the
of Metastatic Cancer”. The emphasis on the update is radiopeptide.
use of antibody as well as non-antibody radionuclide Considerable expansion has been accomplished with
conjugates in treatment of cancer. use of pretargeted RIT and other methods to enhance
tumor-to-normal organ tissue ratios such as the use of
alpha emitting radionuclides. This chapter will focus on
Introduction these clinical therapeutic aspects of targeted radionu-
clides for diagnosis and therapy of tumors. The current
Radiolabeled monoclonal antibodies and peptide based state of clinical use of radiolabeled antibodies and radi-
radiopharmaceuticals have been evaluated as vehicles to opeptides, as well as the state of development of new
selectively target radioactivity directly to tumor cells for compounds and future innovations are reviewed.
more than a decade. Success in achieving significant Discussion of pre-clinical studies will be limited to
remissions in patients with hematologic malignancies those that offer insights into future direction for clinical
such as Non-Hodgkin Lymphoma (NHL) and acute study. The radioisotopes appropriate for diagnosis and
myeloid leukemia (AML) has encouraged investigators therapy and the radiolabeling methods that are currently
to continue working to optimize radioimmunotherapy in use are briefly described.
(RIT) approaches. RIT has thus become the third stan-
dard modality of treatment in hematologic malignan-
cies, while promising results have been obtained in
Radionuclides for Radioimmunotherapy
some solid tumors. Until the past few years, these thera- The choice of radionuclide with which to label antibodies
pies have focused almost entirely on using beta emitting or other targeting entities is governed by several consid-
radionuclides, carried directly by antibodies. In NHL, erations [5, 6]. These include the clinical indication as
for example, response rates have varied from 50% to >80%, well as physical properties such as mode of decay,
with best responses seen using non-myeloablative doses energy, and abundance of the emissions and half-life, as
of therapeutic radionuclide as first-line treatment in well as chemical properties affecting protein attachment
radiosensitive tumors [1–4]. RIT has also been employed and in vivo handling; and finally production aspects
with success as a boost to the radiation dose in combina- including specific activity, availability at needed scale, and
tion with other treatments, particularly in association cost. Recognition of the advantages and disadvantages
with stem cell rescue, as adjuvant therapy, or for treating of the available choices is important since different
small volume disease. strategies are required to maximize their potential.
Despite the enormous potential for targeted therapy, Dose rate is a specific factor modifying therapeutic
problems were identified soon after the early clinical efficacy, particularly for beta emitting radionuclides.
RIT trials were completed. Initially it was thought that Traditional external beam radiation is given at a much
any tumor could be targeted efficiently with monoclonal higher rate than internally administered radionuclides.
antibodies. However, the long circulatory half-life of the In RIT, radiation is delivered in the range of 10–30 cGy/h
blood-borne radiolabeled antibody causes prolonged and continuously decreasing because of decay.
high background activity levels leading to non-specific Generally, effectiveness of cell killing goes down as the
normal organ exposure to radiation. After the detection of dose rate lowers because more time is available for
peptide receptor expression by tumors, peptide analogues repair of sublethal damage [7]. Considering the dose
began to be investigated for specific tumor targeting. rate effect, some have suggested that 20–30% more dose
Peptides used for tumor targeting show several advantages is needed to sterilize tumors compared to fractionated
over antibodies: peptides are small and show rapid diffu- external beam treatment [7, 8], although a review of RIT
sion into (target) tissues resulting in rapid pharmacokinetics. studies in animal xenografts by Wessels suggests dose

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 463
© Springer Science + Business Media B.V. 2009
464 Targeted radionuclide therapy of cancer

effects from RIT are comparable to external beam [9]. ratios increased with use of these smaller constructs, but
An inverse effect resulting in enhancement of low-dose absolute tumor uptake and retention was decreased. In
rate effects has been observed where cells accumulate in the case of Fab and Fab’ fragments, increased kidney
the radiosensitive G2M stage of the cell cycle, which localization also occurred because the efficiency of fil-
may contribute to efficacy of the low-dose rate radiation tration for proteins of this size increases and tubular
of RIT. The type of malignancy may also be a factor as reabsorption occurs. Improved penetration and retention
lymphomas have been especially sensitive to inverse in tumor has been further explained with the use of a
dose rate effects [10]. variety of novel molecular antibody forms with different
The physical half-life of the radionuclide, as well as sizes in both solid and hematologic malignancies. These
the biological half-life for tumor uptake, retention, and include chimeric and humanized whole antibody forms,
elimination from normal tissues of the carrying vehicle CH2 deletion constructs, single-chain forms, diabodies,
must be considered for radionuclide selection. In par- minibodies, and fusion proteins [6, 13–18]. The CH2
ticular, radionuclide half-life considerations include deletion constructs are derived from the constant region
retention time for antibody or another targeting agent of about 125 kD, while single chain Fv of 25 kD are from
such as a peptide in tumor in order to deliver a dose the variable region. Diabodies are divalent forms of Fvs
commensurate with the fraction of injected activity that (scFv dimer) whereas minibodies are scFv-CH3 dimers of
localizes to target tissue. Thus, with conventionally 80 kD [13, 14, 19]. Evaluation of these immunotargeting
radiolabeled antibodies or peptides (i.e., injected with forms has indicated gains in tumor-to-normal tissue
radionuclide bound to targeting agent, the half-life must ratios, but limitations still exist in kidney and liver uptake
be long enough for tumor uptake as well as tumor irra- as well as tumor retention.
diation during time of useful tumor to normal tissue Types of emissions considered are beta particles
ratios). As examples, 131I (8 day t1/2), 186Re (3.7 day t1/2) (electrons emitted with a wide range of energies), alpha
and 90Y (2.7 day t1/2) are suitable for whole antibody decay (in which helium + two particles are emitted),
pharmacokinetics where maximum tumor uptake of and low energy Auger electron emissions, a byproduct
intact antibodies requires 24–48 h, but tumor retention of electron capture decay. While the particulate property
can persist for several days [11, 12]. Conversely 188Re of the radiation decay mode determines the therapeutic
(17 h t½) would be more compatible with short-lived potential, gamma ray emission often associated with the
small molecule targeting. radiation decay, provides the ability to image the biodis-
Another general limitation of radiotherapy is that the tribution in vivo, thus indicating tumor localization and
maximum rate of decay, hence therapeutic efficacy, non-target uptake and retention. Ideally, gamma radia-
occurs at the time of injection. For any process that tion should be of low abundance such that contribution
involves slow accumulation in tumor, typical for anti- to non-target organ irradiation is minimized.
body protein, being a large molecule, much of the radia-
tion decay affects non-tumor tissue before selective
tumor to non-tumor ratios are achieved. Therefore, much
Beta Particle Decay
effort has gone into engineering of various antibody and A wide range of beta emitter energies of emission and
peptide forms over the last several years to further over- half-life are available for RIT (Table 1). Adelstein [20],
come targeting limitations. Intact IgG antibodies are Howell [21], Humm [22], and Wheldon [23] have consid-
approximately 150 kD molecular weight and thus are ered the application of beta emitters with respect to emis-
relatively large molecules for their role as targeting vehi- sion energy and penetration, number of cell reversals, and
cles. The whole antibody characteristics of slow disap- size of tumor targets. Beta emitters have the unique
pearance from the blood and the slow tumor uptake advantage of exerting their cytotoxicity by a crossfire
kinetics have limited their ability to achieve high tumor-
to-normal tissue and blood ratios, and have resulted in Table 1. Selected beta emitting radionuclides for RIT
high radiation exposure of the radiosensitive marrow
Beta emitting Path-length Energy delivered
cells. Early efforts to improve on the pharmacokinetic isotopes t1/2 (mm) (MeV)
limitations of whole antibodies included removing the
Iodine-131 8.1 days 0.8 0.6
Fc portion to yield F(ab’)2 fragments of 100 kD size. Yttrium-90 64 h 2.7 2.3
These have been further split into Fab’ (50 kD), or in Rhenium-188 17 h 2.4 2.1
some cases, Fab fragments of similar size. In general, Copper-67 62 days 0.05–2.1 0.6
blood disappearance rates, and tumor-to-normal tissue Lutetium-177 6.7 days 0.04–1.8 0.5
John M. Pagel et al. 465

effect as only occasional beta particles will achieve lethal Only 27 212Bi atoms and 4 alpha-particle tracks (“hits”) of
212
double strand DNA breaks. When a sufficient concentra- Bi were required for a log of target cell killing. Satisfying
tion of emission occurs in a tissue volume, the probability the requirement of homogeneous targeting of alpha emit-
of lethal hits increases, predominantly from sources ters is more difficult with solid tumors which are often
bound to other cells. This crossfire killing property obvi- poorly vascularized and may be access-limited by high
ates the need for targeting every cancer cell in contrast to interstitial pressure due to poor lymphatic drainage [25].
antibody targeted delivery of drug or toxin conjugates. Toxicity to normal tissue via antibody cross-reactivity can
The crossfire effect is efficient for tumor masses larger in be high over a short range due to the potency of alpha
diameter than the average beta path length. radiation and lack of repair potential of double-stranded
Relatively few beta emitters have been studied in clini- DNA lesions. As expected, good efficacy has been seen in
cal trials for RIT. These include the low energy emitter micrometastatic models, and in established solid tumors
131
I that has a maximum energy of 0.61 MeV and rhenium little more than a delay in tumor growth has been observed.
(186Re) with a moderate energy of 1.07 MeV. The higher Alpha particle radiotherapy studies have been reviewed in
energy beta emitter that is currently most widely used detail by Hassfjell and Brechbiel [26].
radionuclide in clinical trials and clinical practice is 90Y
(2.3 MeV). Comparing isotopes, Humm [22] estimated
54% absorption of energy from particles of 131I in a 1 mm
Emerging Radionuclides for use in RIT
cluster but only 10% absorption of 90Y in this small vol- Additional pre-clinical investigations have focused on a
ume. Further analysis by Wheldon and coworkers suggest wider range of beta and alpha emitting radionuclides with
that moderate or lower energy emitters should be used for different energies of emission and half-lives to improve
clusters of up to [107] cells while an emitter such as 90Y the efficacy of RIT. Two promising beta emitting radio-
should be used for tumors of [108] cells or greater [23]. nuclides are 177Lu (0.5 MeV) and 67Cu (0.6 MeV).
Lutetium-177 has a moderate beta energy that is similar
to 131I and possesses a 6.7 day half-life. An advantage for
Alpha Particle Decay the use of 177Lu is that this lanthanide has similar chela-
There has been significant interest in targeting alpha tion properties to 90Y, suggesting that use of standard
radiation for RIT over the past several years (Table 2). chelating agents will lead to stable delivery of 177Lu to
With the large He particle emitted, alpha decay results targeted cells. Moreover, 177Lu can be produced in high
in high linear energy transfer (LET) or energy delivery specific activities relatively inexpensively, making the
over a distance of only several cell diameters. This use of this agent in clinical trials probable for the near
results in the advantages of high potency and lack of future. Two copper radionuclides have emerged with con-
oxygen dependence with correspondingly no shoulder siderable promise for RIT of hematologic malignancies.
on the cell survival curve. The short path length, how- Copper-67 has a t1/2 greater than 2 days and emits beta
ever, results in the need for much more homogeneous particles with reasonable energy for therapeutic benefit.
targeting for complete tumor cell kill. Alpha emitters Copper-64 is also a beta emitting isotope, but in addition
64
that have been studied for antibody mediated tumor Cu emits positrons, making this radionuclide appealing
targeting include 212Bi (t1/2 1.06 h), 213Bi (t1/2 0.76 h), 211At for use with positron emission tomography to allow for
(t1/2 7.2 h), and 225Ac (t1/2 10 days). accurate estimations of the absorbed doses of therapeutic
67
As these are short-lived radionuclides, applications Cu. Readily accessible quantities of these copper radio-
have been mainly in leukemia and lymphoma and non- metals have been limited, however, and must be more
systemic administration for other tumors such as intraperi- widely available at affordable cost before these agents
toneal injection. Macklis and coworkers evaluated physical gain widespread application in RIT protocols.
characteristics of 212Bi labeled antibody in lymphoma [24].

Table 2. Selected alpha emitting radionuclides for RIT Antibody-Based


Alpha emitting Path-length Energy delivered
isotopes t1/2 (μm) (MeV) Radiopharmaceuticals
Bismuth-213 46 min 84 6.0 Extensive work exploiting the safety and efficacy of
Actinium-225 10 days 50–80 8
radiolabeled monoclonal antibodies that target specific
surface antigens on malignant cells has been incorpo-
Astatine-211 7.2 h 60 6 rated into a new generation of novel treatment approaches
466 Targeted radionuclide therapy of cancer

for cancer. The characteristics of the antibodies that need to be evaluated at doses requiring marrow rescue to
must be considered for RIT are the same as those for achieve optimal results [31].
immunotherapy, and are listed in Table 3. Bierman et al. reported that 30 mCi 90Y-polyclonal
Beierwaltes’ successful treatment of metastatic mela- anti-ferritin combined with high-dose chemotherapy in
noma in a single patient with 131I-labeled polyclonal anti- patients with Hodgkin disease undergoing bone marrow
body raised to the patient’s own tumor was the first transplantation did not cause any additional adverse
indication that RIT might be feasible [27]. Order et al. effects [32]. The marrow irradiation from the 90Y did
reported initial trials of 131I and 90Y-labeled antiferritin not cause increased toxicity nor did it interfere with
polyclonal antibodies [28–30]. These studies showed that re-engraftment in this study. Although no definite
131
I-labeled antiferritin polyclonal antiserum could pro- improvement in outcome could be attributed to the RIT,
duce regressions of bulky hepatomas [29]. Four of 24 the lack of additional toxicities with the additional low-
patients had partial responses (PR) and one had a com- dose rate irradiation suggested that further similar studies
plete response (CR). Single doses as high as 150 mCi were in patients with better performance status were warranted.
given with acceptable hematologic toxicity. Subsequent Overall those early radiolocalization studies per-
studies combined cytotoxic agents (5-FU and adriamycin) formed with polyclonal antibodies, however, suggested
and external beam radiation with lower doses (50 mCi in that more homogeneous antibodies were required in
divided doses) of antibody-labeled radiation and achieved order to be clinically useful. Hybridoma technology
a response rate of 48% (7% CR; 41% PR). Monoclonal permitted careful selection of monoclonal antibodies
anti-ferritin was found to be inferior to polyclonal antisera directed to particular tumors, such that antibodies with
because of high localization in the liver, and 90Y-labeled the highest localization and binding potential and with
anti-ferritin to be superior to 131I-anti-ferritin. the lowest cross-reactivity with normal tissue are identi-
Responses following up to 30 mCi 90Y-anti-ferritin fied. The majority of the clinical trials have utilized
antibodies were also seen in patients with Hodgkin radiolabeled murine monoclonal antibodies either with
disease when used in conjunction with chemotherapy [30]. or without other concomitant therapy, combined with
131 90
A response rate of 62% was reported in 27 patients with I, Y and less frequently, 186Re as the radiolabel.
advanced Hodgkin lymphoma with 90Y-anti-ferritin.
Marrow toxicity at 30 mCi limited further dose escalation,
although multiple infusions were administered in some
Radioimmunotherapy of Hematologic
patients. In a separate approach polyclonal radiolabeled Tumors
anti-ferritin antibody was delivered by dose fractionation.
Lymphoma
At low doses, this did not improve response rates, yet may
The most impressive results of RIT to date have been
achieved in NHL. Several monoclonal antibodies conju-
Table 3. Considerations for radiolabeled antibody studies gated with radioactivity have been evaluated in the treat-
Antibody Antigen – Location, cellular density, ment of NHL. RIT has proven to be particularly effective
modulation, circulating in the treatment of patients with NHL because of the
Affinity sensitivity of lymphocytes to radiation, the large num-
Specificity
Mass ber of target antigens on the surface of lymphocytes and
Molecular form – intact, fragments the vascular accessibility of the malignancies. The first
Form-murine, chimeric, humanized, human RIT trials for B-cell lymphomas were using 131I-labeled
Radionuclide Half-life Lym-1 antibody. Lym-1 is a novel, murine, IgG2a mono-
Emissions-type, energy, abundance clonal antibody that recognizes a 31–35 kD membrane
Chemistry antigen expressed on malignant B cells characterized as
Specific activity HLA-DR10 [33].
Preparation Percent protein bound The DeNardo group has generally taken the approach
Immunoreactivity of administering lower doses of fractionated RIT, con-
Purity
sistent with standard oncological practice using chemo-
Patient Pharmacokinetics therapy and external beam therapy, to deliver higher
Images, normal organ biodistribution,
tumor uptake overall dosages with lower toxicity. In a study using low
Radiation absorbed dose estimates dose, fractionated therapy 30 patients with B-cell malig-
Antiglobulin response nancies received either 30 or 60 mCi 131I-labeled Lym-1
Tumor response following RIT antibody delivered every 2–6 weeks up to a total dose of
John M. Pagel et al. 467

300 mCi. Acute toxicity (e.g., fever, rash) was mild and differentiation antigens, including HLA class II variant
transient. Dose limiting toxicity was thrombocytopenia, molecules, idiotypic immunoglobulins, the CD 5, 20, 21,
particularly in patients with low platelet counts at base- 22 and 37 antigens, and the IL2 receptor. Targeting radia-
line and in patients with lymphomatous marrow involve- tion to the CD19, CD20, and CD22 antigens has proven,
ment. Eleven of the 30 patients received the intended however, the most effective in clinical NHL trials. These
dose of 300 mCi (thought to be the maximum-tolerated target antigens are expressed in high density with >95% of
dose (MTD) based on marrow dose estimates). The all B-cell lymphomas expressing CD19 and CD20, and
overall response rate (ORR) was 57% for all patients 70% expressing CD22. Two anti-CD20 radiopharmaceu-
and 94% of the 18 patients who received at least ticals (131I-tositumomab and 90Y-ibritumomab tiuxetan)
180 mCi. Ten percent of patients achieved a CR lasting have now been approved for use in patients with relapsed
10–44 months [34]. or refractory indolent disease.
A second study was designed to define the MTD and
131
efficacy of at least the first two, of a maximum of four, I-Tositumomab
high doses of 131I-Lym-1 given 4 weeks apart [35]. Kaminski et al. first described the use of low-dose
131
Dosages studied were 40 to 100 mCi/m2 administered I-labeled anti-CD20 antibody (tositumomab) to treat
every 4 weeks. Twenty patients with different advanced patients with relapsed indolent NHL [39]. Patients
NHL, histologic subtypes that were resistant to standard receiving this therapy first received tositumomab labeled
therapy were treated. Dose-limiting toxicity was found with a trace amount of 131I (5 mCi) followed by a thera-
to be thrombocytopenia at an MTD of 100 mCi/m2. The peutic infusion based on a predicted estimate of the
ORR was 52% with seven patients (33%) achieving a whole body radiation absorbed dose. Each dose of radi-
CR, persisting for median duration of 14 months. All olabeled antibody was preceded by an infusion of unla-
three patients that received the MTD (100 mCi/m2 × 2) beled anti-tositumomab antibody to reduce normal,
had a CR. The median duration of survival was 19 antigen-specific binding. A Phase I dose-escalation trial
months in responding patients and 1.9 months in non- was subsequently conducted to assess the toxicity and
responders. Considering both studies, responses were efficacy of non-myeloablative doses of 131I-tositumomab
observed in 30 of 45 patients including 95% of patients [40] Patients were treated with whole-body radiation
who received more than 200 mCi of 131I Lym-1. Of note, doses escalating in 10 cGy increments from 25 to 85 cGy.
the high dose approach achieved a higher CR rate than Twenty-eight of the 34 patients enrolled completed
the low dose approach. treatment. The overall response rate was 79% and 50%
The Lym-1 antibody has also been labeled with 67Cu, achieved a CR. The median duration of response was 12
in four patients to compare the biodistribution and months for all patients and 16 months for those patients
dosimetry to 131I-Lym-1 [36]. Both the uptake in tumor who reached a CR. Sixteen of 17 patients who achieved
and the retention time in tumor were found to be higher a response of 6 months or more in duration remained
with the 67Cu-Lym-1 than the 131I-Lym-1. Marrow dose alive 6 years after treatment. The whole body clearance
estimates, however, were lower with 67Cu and liver dose was found to be variable between patients, most likely
estimates were higher. Copper-67-Lym-1 was further dependent on the B-cell load and tumor bulk, thus the
administered to 12 patients in a Phase I/II dose escala- tracer study was deemed necessary to administer the
tion trial. Up to four doses of 25 or 50–60 mCi/m2 were MTD to each patient. Hematologic toxicity was dose
administered, the lower dose when marrow involvement limiting at a whole-body radiation absorbed dose of
was present. Dose limiting toxicity was hematologic 75 cGy. These early studies led to development of a
and the ORR was 58% [37]. Although the supply of diagnostic dose to determine pharmacokinetics for
67
Cu is at present uncertain, this remains an isotope worthy selecting a therapy dose that will deliver a whole body
of further study. dose of 75 cGy, the MTD for patients without compro-
The majority of RIT investigators have aimed however mised marrow function (in patients with lower baseline
to deliver the MTD in one injection in order to deliver the counts, 65 cGy is the target whole body dose).
highest dose rate possible to the patient. This has been, at Based on the low toxicity and the clinical results
least in part, due to the concern of immunoglobulin devel- found in the Phase I trials, a trial was designed to evalu-
opment toward the therapeutic antibody, which could ate 131I-tositumomab with the primary clinical endpoint
potentially limit the number of infusions that can be safely being the comparison between the patient’s duration of
administered. Results have been reported by various inves- remission following 131I-tositumomab therapy and the
tigators using single non-myeloablative doses of radiola- duration of remission on the patient’s last chemotherapy
beled antibodies directed against a variety of lymphoid [42]. The study included 60 indolent or transformed
468 Targeted radionuclide therapy of cancer

NHL patients who were refractory prior to chemother- with a median PFS of 6.1 years. Analysis according to
apy. Seventy-four percent of the patients with indolent the Follicular Lymphoma International Prognostic Index
NHL experienced a longer duration of response to risk categories demonstrated that 85% of the patients
131
I-tositumomab compared to 26% who experienced a were intermediate or high risk [46].
longer duration of response to prior chemotherapy (p < Phase II studies of RIT as consolidation following
0.001). The median duration of remission after upfront therapy with a variety of chemotherapy regimens
131
I-labeled antibody was 6.5 months, approximately including CHOP and fludarabine have resulted in high
doubling the 3.4-month median duration of remission overall response rates and excellent PFS [2, 3]. In a phase
patients experienced on their last chemotherapy regi- II trial conducted by the Southwest Oncology Group, the
men. In addition, a response was observed in only 28% complete response/unconfirmed complete response rate
of patients following their last chemotherapy regimen improved from 39% after CHOP to 69% following con-
compared to 65% of patients following a regimen of solidative 131I-tositumomab with an estimated 5-year
131
I-tositumomab (p < 0.001). PFS and overall survival of 67 and 87%, respectively [2].
Updated and long-term data on 53 chemotherapy- Based on these promising results, an ongoing phase III
relapsed/refractory patients, including patients who had trial compares consolidation with 131I-tositumomab fol-
received a prior autologous stem cell transplant (ASCT), lowing CHOP chemotherapy to observation in untreated
that were treated with iodine 131I-tositumomab have been follicular lymphoma patients. The current US Intergroup
provided [43]. Dose-escalations were conducted sepa- phase III trial randomizes untreated follicular lymphoma
rately in patients who had or had not undergone a prior patients to either six cycles of CHOP followed by
131
ASCT until a non-myeloablative maximally tolerated I-tositumomab or to rituximab plus CHOP providing a
whole body dose was established (non-ASCT = 75 cGy, valuable comparison between two contemporary treat-
post-ASCT = 45 cGy) and then 14 additional non-ASCT ment options for the frontline therapy of follicular
patients were treated with 75 cGy. Forty-two of 59 lymphoma. Whether upfront cytoreduction with chemo-
patients (71%) responded and 20 patients (34%) had a therapy benefits previously untreated patients receiving
131
CR. Thirty-five of 42 patients (83%) with low-grade or I-tositumomab is unknown. Clinical outcomes follow-
transformed NHL responded versus 7 of 17 (41%) with ing RIT alone were similar to those achieved with che-
de novo intermediate-grade NHL (p = 0.005). For all 42 motherapy followed by RIT and a randomized trial will
responders, the median progression-free survival was 12 be required to address this issue.
months and 20.3 for those with CR. Seven patients
90
remained in CR 3 to 5.7 years. Sixteen patients were Y-ibritumomab tiuxetan
retreated after progression; nine responded and five had Ibritumomab (IDEC-Y2B8, Zevalin®) is a murine IgG1
a CR. Reversible hematologic toxicity was dose limiting. kappa monoclonal antibody that covalently binds
Only ten patients (17%) had human anti-mouse antibod- MX-DTPA (tiuxetan), which chelates therapeutic 90Y.
ies detected. Long-term, five patients developed elevated A multi-institution Phase I/II study evaluated the safety
thyroid-stimulating hormone levels, five were diagnosed and efficacy of treatment with 90Y-ibritumomab tiuxetan
with myelodysplasia and three with solid tumors. An in 58 patients with low or intermediate grade and mantle-
expanded access study of 359 patients with relapsed or cell NHL [47]. The amount of 90Y-labeled antibody was
refractory low grade or transformed low grade NHL was dose-escalated from 0.2 mCi/kg to 0.4 mCi/kg. Prior to
90
carried out. In 273 patients evaluable for response, the Y treatment, biodistribution studies were conducted
ORR was 58% and the CRR was 27%. Follow up at 17 using a surrogate 111In-labeled antibody conjugate to
months showed that the median duration of response had allow for gamma imaging. Rituximab was pre-adminis-
not yet been reached [44]. A single, well-tolerated treat- tered as the unlabeled blocking antibody (250 mg/m2) to
ment with iodine 131I-tositumomab can, therefore, pro- allow for known sites of disease to be more easily visu-
duce frequent and durable responses in NHL, especially alized and this also decreased the projected dose of radi-
low-grade or transformed NHL. ation to this spleen and marrow. The MTD was 0.4 mCi/
As a first line therapy in follicular lymphoma, kg (0.3 mCi/kg for patients with baseline platelet counts
131
I-tositumomab is currently showing promising results. 100 to 149,000/ml). The only significant toxicity was
Kaminski et al. reported a 95% overall response rate myelosuppression. The ORR for the intent-to-treat pop-
(75% complete response) in 76 previously untreated fol- ulation (n = 51) was 67% (26% CR; 41% PR); for low-
licular lymphoma patients. Molecular remissions were grade disease (n = 34), 82% (26% CR; 56% PR); for
demonstrated in 80% of patients who achieved a com- intermediate-grade disease (n = 14), 43%. Responses
plete response [45]. Actuarial 5-year survival was 59% occurred in patients with bulky disease (≥7 cm; 41%)
John M. Pagel et al. 469

and splenomegaly (50%). The median time to progres- percent of patients screened were eligible for thera-
sion for patients who responded was 12.7 months. The peutic doses of radiolabeled antibody. Patients received
phase I/II trials were followed by a phase III trial that 58–1,168 mg antibody labeled with 234–777 mCi of 131I.
randomized 143 eligible patients to either rituximab or At absorbed doses above 27.25 Gy to lungs, two of four
90
Y-ibritumomab tiuxetan radioimmunoconjugate to patients experienced cardiopulmonary toxicity. Sixteen
demonstrate that the combination of the 90Y radioiso- of 19 patients achieved a CR and 2 of 19 patients
tope to the murine anti-CD20 antibody provided addi- achieved a PR. Mean response duration was in excess of
tional efficacy over the unconjugated (“cold”) rituximab 19 months. A phase II study repeated these results and
alone [48]. A planned interim analysis of the first 90 obtained responses in 15 of 19 patients. At 6 years, the
patients demonstrated an ORR of 80% with projected overall survival was 78% for patients with
90Y-ibritumomab tiuxetan versus 44% for rituximab (P indolent lymphoma and 43% for patients with aggressive
< 0.05). To provide additional evidence of the benefit of histologies [51].
90
Y radioimmunotherapy over rituximab immunother- Based on the results above, a study to evaluate treat-
apy, patients who were nonresponsive or refractory to ment with the MTD of radioactivity was conducted [52].
rituximab were enrolled in an additional trial and treated Twenty-two of 25 patients evaluated with trace labeled
with 90Y-ibritumomab tiuxetan 0.4 mCi/kg. An ORR of doses achieved biodistributions considered adequate to
46% was achieved in these rituximab-refractory patients. receive a therapeutic infusion. Twenty-one patients were
These results provide further evidence of the added treated with therapeutic infusions of 131I-tositumomab
value of 90Y [49]. antibody calculated to deliver not more than 27 Gy to
More recently a phase II trial combining six cycles of normal organs followed by autologous hematopoietic
chemotherapy with cyclophosphamide, doxorubicin, vin- stem cell reinfusion. Seventeen (81%) achieved CR with
cristine, and prednisone (CHOP) followed 6–10 weeks a median duration of response of 38 months.
later by 90Y-ibritumomab tiuxetan was conducted to eval- The relative long-term efficacy of this high-dose RIT
uate the efficacy and safety in untreated elderly diffuse strategy was evaluated via a multivariable cohort analy-
large B-cell lymphoma (DLBCL) patients. In 20 eligible sis of 125 patients with relapsed or refractory follicular
elderly (age ≥ 60 years) patients with previously untreated lymphoma treated with either myeloablative I-131 tosi-
DLBCL using this novel regimen the ORR to the entire tumomab followed by autologous HCT or conventional
treatment regimen was 100%, including 95% CRs and high dose therapy followed by autologous HCT. In this
5% PRs. Four (80%) of the five patients who achieved study, the estimated 5-year OS for high-dose RIT was
less than a CR with CHOP improved their remission sta- 67% and for conventional high-dose therapy was 53%
tus after 90Y-ibritumomab tiuxetan RIT. With a median (p = 0.004) [53]. Likewise, 5-year PFS was 48% for the
follow-up of 15 months, the 2-year PFS was estimated to high-dose RIT group and 29% for the conventional
be 75%, with a 2-year OS of 95%. This study therefore transplant group (p = 0.03). Furthermore, 100-day treat-
has demonstrated the feasibility and tolerability of this ment-related mortality was lower in the high-dose RIT
regimen for elderly patients with DLBCL [4]. group than conventional HCT group (3.7% versus
11.2%) with no evidence of increased MDS/AML at 8
High-dose Radioimmunotherapy with Stem Cell years of follow-up. Based on the exceedingly low non-
Support for Lymphoma hematopoietic toxicity of single-agent HD-RIT followed
Data from prior studies showing an inverse relationship by autologous HCT, this strategy was evaluated in older
of recurrence rates to radiation dosage led investigators adults with relapsed B-NHL. Older patients with refrac-
to hypothesize that if the radiation dose could be further tory or relapsed NHL typically have limited therapeutic
safely escalated to tumor sites, relapse rates would be options largely due to the excessive toxicity associated
reduced without incurring additional toxicity. Press et with potentially curative dose intense regimens and
al. explored the use of myeloablative doses of 131I-labeled co-morbidities [53]. In this study, 24 patients ≥60 years
monoclonal antibodies with ASCT support in 43 patients of age with relapsed or refractory B-NHL received high-
with B cell lymphomas who had failed conventional dose RIT targeted to ≤25–27 Gy to normal organs fol-
chemotherapy [50]. Two anti-CD20 antibodies (tositu- lowed by ASCT. Notable findings from this series
momab and 1F5) and one anti-CD37 antibody (MB-1) included low rates of non-hematopoietic toxicity (<10%
were evaluated. Patients were selected for treatment grade 4), no treatment-related deaths, and an estimated
after tracer studies with increasing mass doses of anti- 51% 3 year PFS.
body that determined that the absorbed dose to tumor These results, although encouraging, led to relapse in the
would be greater than that to normal organs. Sixty-four majority of patients. Therefore, Press et al. combined
470 Targeted radionuclide therapy of cancer

the MTD of 131I-tositumomab with high-dose etoposide the CD33 antigen could be used to treat patients with
and cyclophosphamide prior to autologous HCT [54]. acute myeloid leukemia (AML) [60, 61]. In about half
While the toxicities were significantly greater for the of the patients radiotherapy was delivered with relative
combination regimen than for single-agent therapy, the specificity to the marrow. The liver was the only other
estimated overall survival at 2 years was 83% and the organ showing localization of the radioimmunoconju-
progression free survival is 68%. Non-randomized con- gate and was considered to be the dose-limiting normal
trols treated with identical doses of chemotherapy but organ. Success appeared limited by low CD33 expres-
with total body irradiation had inferior PFS (p < 0.002) sion on target cells and by internalization of the anti-
and OS (p < 0.004) compared to those who received body–antigen complex, leading to deiodination and
radiolabeled antibody based regimen. More recently, a release of 131I. Jurcic et al. studied a humanized anti-CD33
subset of 16 patients with refractory mantle cell lym- antibody (HuM195) radiolabeled with 90Y. Anti-leukemic
phoma who had been treated with this approach were activity was seen, but with prolonged myelosuppression,
reported and showed 3-year PFS and OS from trans- suggesting the utility of this approach as a pretransplant
plantation estimated at 61% and 93%, respectively [55]. preparative agent [62].
Extended phase II studies using this combined high- Matthews et al. treated patients with leukemia using
dose chemoimmunotherapy regimen in various B-NHL BC8, a murine anti-CD45 IgG1 antibody that does not
histologies are ongoing to better define the safety and internalize but is reactive with 85–90% of cases of acute
efficacy of this regimen. leukemia, and the majority of nucleated cells in normal
marrow, but not with cells outside the lymphoid or
hematopoietic lineages. A phase I trial in 44 patients
Leukemia
with AML, or acute lymphoblastic leukemia (ALL)
Abs directed against the CD33, CD45, and CD66 beyond first remission or advanced myelodysplastic
hematopoietic differentiation antigens have been most syndrome (MDS) was carried out in conjunction with
extensively explored for RIT of leukemia. CD33 is an cyclophosphamide, 12 Gy TBI, and stem cell rescue. In
attractive target for RIT of myeloid leukemias, since this dose escalation trial patients were selected for ther-
CD33 is expressed on most myeloid leukemic blasts, but apy only when a diagnostic dose indicated that the bone
not on hematopoietic stem cells [56]. Anti-CD33 Abs marrow and spleen would receive more radiation than
have been shown to rapidly saturate antigenic targets and the other normal organs. Eighty-four percent of patients
the Ab-antigen complexes are rapidly internalized into had favorable biodistributions. This improved biodistri-
the cell. Other alternative targets for RIT that do not bution rate was likely due to the characteristics of the
modulate upon Ab interaction have also been explored. anti-CD45 antibody, with longer retention of the radia-
CD45 is stably expressed at a high density on the surface tion at the tumor. Liver dose from a tracer study was
of virtually all hematopoietic cells and does not undergo used to limit the dose escalation. The MTD was 10.5 Gy
significant modulation upon Ab binding. Most hemato- to the liver. This trial demonstrated that 131I-labeled anti-
logic malignancies, including 85–90% of acute lymphoid CD45 antibody could deliver two to five times more
and myeloid leukemias express CD45 [57, 58]. The radiation to the marrow and spleen compared to normal
CD66 antigen is another target that also does not inter- organs. This approach at the MTD resulted in 24 Gy
nalize or shed into the circulation. CD66 is expressed at more radiation delivered to the marrow and approxi-
high density on maturing hematopoietic cells but is not mately 50 Gy to the spleen, without excessive toxicity in
found on AML blasts [59]. Therefore, cytotoxic radia- a setting of conventional cyclophosphamide/TBI [63].
tion is delivered to bystander leukemic cells by targeting Seven of 25 patients with AML/MDS were disease free
normal CD66 expressing myeloid cells. for a median of 26–100 months at the time of the report,
and three of nine patients with ALL were disease free
Beta emitting Radionuclides for RIT of for 34–82 months post transplant.
The same preparative regimen was used in a later
Leukemia study for patients with AML beyond first remission with
A relatively small number of beta emitters have been relapsed/primary refractory AML, or advanced MDS.
studied in clinical trials for RIT of leukemia. Radioiodine Results suggested that a greater radiation absorbed dose
was first used for RIT of AML because of its well-estab- delivered to marrow may lead to lower post-transplant
lished experience as a medical radiotherapeutic. Two relapse rates for patients with high-risk AML/MDS [64].
groups of investigators demonstrated that 131I-labeled Half of the patients treated received a dose to marrow
monoclonal antibodies (p67 and M195) reactive with <7 cGy/mCi while the remaining half of patients received
John M. Pagel et al. 471

>7 cGy/mCi to marrow. Although the number of patients allogeneic transplantation after 188Re-anti-CD66 condi-
was limited, only one of nine patients relapsed in the tioning, 45% were disease free; in addition, significantly
high marrow-absorbed dose group while six of nine more patients remaining disease free if they were in
patients relapsed in the lower dose cohort. The hazard of remission at the time of transplant, compared to those
relapse in the group that received <7 cGy/mCi to patients who were not in remission at the time of trans-
marrow was more than six-times higher than that seen plant (67% versus 31%). For older high-risk AML or
in the group that received the higher marrow dose. Since MDS patients, RIT using an anti-CD66 antibody labeled
greater estimated radiation doses could be delivered with either 188Re or 90Y was also combined with a
safely by an 131I-anti-CD45 antibody to marrow and reduced intensity conditioning regimen followed by
spleen compared to non-target organs, a trial for patients transplant using T-cell-depleted allografts [69]. Despite
with AML in first remission was conducted [65]. Forty- the low level of toxicities seen in older patients using
nine patients received high dose targeted irradiation this approach, the cumulative incidence of relapse
delivered by 131I-anti-CD45 antibody combined with remained high (55% at 30 months post-transplant). The
busulfan and cyclophosphamide. The radiolabeled anti- risk of relapse was almost 20% higher for patients not in
body delivered an average of 11 Gy to marrow and remission compared to those in either first or second
almost 30 Gy to spleen. These first remission patients remission. These studies highlight the difficulty of tar-
had a low relapse rate (20%) and a 62% estimate of geting multiple low-energy beta emissions to neighboring
3-year disease-free survival after transplantation. These leukemic blasts by relying on crossfire from normal
data appear promising since at least 30% of patients targeted CD66 positive granulopoietic cells.
with AML in first remission would be expected to
develop leukemic relapse after being treated with stan-
Alpha Emitting Radionuclides for RIT of
dard busulfan and cyclophosphamide conditioning and
stem cell transplant [66]. Nevertheless, carefully con- Leukemia
trolled randomized trials will be necessary to defini- The feasibility of using alpha emitting radionuclides
tively assess the contribution of the 131I-anti-CD45 was established in an initial study that employed
antibody to standard transplant regimens. 213
Bi-HuM195 for patients with refractory or relapsed
These encouraging results suggested that this AML or MDS [70]. In 18 patients with refractory or
approach might be applied to older patients. Given the relapsed AML or MDS, 13 patients had decreases in the
reduced toxicity of a low-intensity conditioning regi- percentage of marrow blasts and 10 of 12 evaluable
men in high-risk older patients, the anti-leukemic effect patients had reduced peripheral blood leukemia cells
of a non-ablative approach might also be improved by [62]. 213Bi-HuM195 was given in three to six fractions
the addition of targeted hematopoietic irradiation deliv- over 2–4 days. Uptake of antibody in marrow, spleen,
ered by a radiolabeled antibody. An allogeneic trans- and liver was seen within 10 min. There were no signifi-
plant trial using targeted hematopoietic irradiation cant acute toxicities, however, transient elevated liver
delivered by an 131I-anti-CD45 antibody (BC8) com- function abnormalities occurred. Myelosuppression
bined with fludarabine and 200 cGy TBI has been initi- lasted 34–50 days. Since alpha-particle RIT also has
ated for a particularly poor risk population of advanced promise to be effective in the treatment of minimal leu-
older AML and high-risk MDS patients. This study has kemia states, a follow-up study has been initiated using
demonstrated that at least an average of 27 Gy of tar- chemotherapy to first achieve a significant degree of
geted radiotherapy can be delivered to bone marrow and cytoreduction prior to the delivery of 213Bi-HuM195.
an average of 81 Gy to the spleen, in addition to a stan- Clinical use of 213Bi to date has been otherwise limited,
dard reduced intensity transplant regimen, without a however, since this radionuclide is relatively short lived,
marked increase in day 100 mortality [67]. Whether use which may result in a potentially short marrow resi-
of a radiolabeled antibody combined with a non- dence time of the radionuclide. Recognizing this limita-
myeloablative transplant will reduce post-transplant tion and that 225Ac has a 10 day half-life, a phase I trial
relapse rates for these older patients with high-risk investigating the use of 225Ac-HuM195 for advanced
AML/MDS, however, remains to be determined. myeloid leukemias has been initiated at Memorial Sloan
Rhenium radioisotopes have also been investigated as Kettering Cancer Center [71]. This study holds consid-
part of conditioning regimens prior to HCT. In particu- erable promise for advancing the field of alpha-particle
lar, 188Re labeled anti-CD66 antibody has been used in RIT because of the longer half-life and the generation of
patients with high-risk AML [68]. At a follow-up of 18 short-lived daughter therapeutic alpha-particles (221Fr,
months in 36 relatively younger patients who underwent 217
At, 213Bi) from 225Ac inside a targeted leukemia cell,
472 Targeted radionuclide therapy of cancer

which largely accounts for the increased potency of For example, in both NP-4 and COL-1 studies, complexes
225
Ac-atomic nanogenerators over 213Bi constructs [72]. were detected within 5 min of injection, indicating that
CD25 and CD30 have recently been explored as antibodies directed against circulating antigen are sub-
rational targets for RIT of leukemia as these antigens are ject to variable pharmacokinetics related to the level of
expressed by a high proportion of disease specific neo- circulating antigen [77, 90]. In other anti-CEA antibody
plastic cells and not by normal resting cells. Recently, a studies (e.g., c84.66 antibody), however, tumor burden
pre-clinical model of leukemia labeled with the alpha was noted to alter the pharmacokinetics probably even
emitter 211At has been studied as a potential radioimmu- more so than in patients when antibodies targeting
notherapeutic for patients with CD25 and CD30 posi- non-shedding antigens were used [83].
tive leukemias [73, 74]. These findings have set the
foundation for progression to clinical studies using radi-
olabeled Abs for the treatment of patients with either
Other CEA-Expressing Cancers
CD25 or CD30 expressing leukemias. Phase I studies with anti-CEA antibodies have also been
carried out in patients with ovarian cancer and unre-
spectable or metastatic medullary thyroid cancer (MTC).
RIT of Non-Hematologic Tumors Juweid et al. reported on a dose escalation trial in which
131
A variety of antibody based proteins have been studied I-MN-14 was administered to 11 patients with
for RIT of solid tumors including whole antibodies and advanced ovarian cancer, but a clinical response was
various fragments derived from them, as well as chime- seen in only one patient [77] Juweid reported a study
ric and humanized modifications to minimize immuno- with non-myeloablative doses of 131I-labeled NP-4 and
genicity. Fractionated as well as single doses have been MN-14 intact antibodies and their bivalent fragments in
administered, marrow rescue has been used for high- 17 patients with MTC [91]. Dosages of 131I ranged from
dose studies, and the targeted radionuclide has been 46 to 268 mCi, depending on whether a fragment was
selected from 131I, 90Y, 186Re, or the Auger emitting iso- administered. Minor responses were observed in 5 of 11
tope, 125I. Internalizing and non-internalizing antibodies evaluable patients, mostly with the F(ab’)2 fragments of
have also been evaluated. Most of the antibodies react both antibodies. Juweid also reported on the toxicity
with several tumor types, but we have chosen to catego- and therapeutic potential of high-dose 131I-MN-14
rize these studies according to site of primary disease. F(ab’)2 combined with ASCT in patients with rapidly
progressing metastatic MTC [92]. Twelve patients were
entered into the study and dose escalation was based on
Gastrointestinal Carcinomas prescribed radiation doses to critical normal organs. The
Patients with gastrointestinal malignancies have been highest dose level reported was 12 Gy to critical organs
widely studied in Phase I trials. In general these patients and was not dose limiting. One patient had a partial
have not been exposed to intensive chemotherapy from remission for 1 year, another had a minor response for 3
myelosuppressive drugs and do not have rapidly progres- months, and 10 patients had stabilization of disease last-
sive disease, and thus they are relatively ideal patients for ing between 1 and 16 months. The authors of this study
proof of concept and Phase I studies. A review of several suggested that the anti-tumor responses seen in these
RIT trials for treating gastrointestinal cancer revealed an patients with aggressive, rapidly progressing disease
extremely low response rate [41, 75–89]. was encouraging and warranted further study.
Of almost 300 patients studied in these phase I trials,
tumor responses included only one CR, 4 PR and
although stable disease or minimal responses were not
Prostate Cancer
always reported, this may have been present in 10% of CYT-356, Prostascint™, uses an antibody directed against
patients or more. Marrow toxicity invariably limited a prostate specific antigen and has been studied exten-
dose escalation, even in these patients with often rela- sively as an imaging agent [93]. This antibody has also
tively little prior myelosuppressive chemotherapy. been radiolabeled with 90Y for a phase I therapy study in
Additional strategies will have to be implemented for prostate cancer patients. Twelve patients were studied,
RIT in solid tumor patients to achieve higher response the MTD was below 12 mCi/m2, limited by marrow toxic-
rates. A potential complicating factor in several Phase I ity. The authors believed marrow toxicity to be related to
trials for colon cancer has been the use of anti-CEA extensive bony involvement and prior marrow irradiation
antibodies that interact with circulating CEA antigen to to more than half the marrow. No objective responses
alter the pharmacokinetics of the therapeutic moiety. were seen, but improvement in symptoms were reported.
John M. Pagel et al. 473

KC4, is a pancarcinoma antibody directed against a Three to 15 mg L-6 was administered with 10 mCi 131I per
high molecular weight membrane and cytoplasmic gly- mg antibody and the MTD was 60 mCi/m2. One patient
coprotein. In a dose escalation study, calcium disodium achieved a CR, but 40% of patients also developed a
EDTA was administered for 72 h in conjunction with HAMA response [98]. In a multiple-dose protocol, ten
escalating doses of 90Y-KC4 to patients with refractory heavily pretreated patients were treated with monthly
prostate cancer [94]. Again here, symptomatic improve- injections of 131I-chimeric L-6. The MTD was 60 mCi/m2
ment was reported, but at the low dose of 9 mCi/m2 twice limited by myelotoxicity. In five of the ten patients,
severe marrow toxicity was observed. clinically measurable tumor responses were seen [99].
Meredith et al. has reported on several phase I and II In this study, serum levels of IL-2 receptors were mea-
trials using 131I-CC49 in prostate cancer patients [95, 96]. sured and the increase in serum IL-2 was greatest in the
A phase II study with 75 mCi/m2 131I-CC49 showed patients who responded. Immunoglobulin developed in
symptomatic improvement in two thirds of patients but eight of ten patients at varying times, and did prevent
all patients developed human anti-mouse antibody further therapy doses in four patients. The authors specu-
(HAMA) within 4 weeks of therapy [95, 96]. A trial with lated that the reason for success with the L-6 antibody is
a low-dose of cyclosporin and the dose of 131I-CC49 frac- a combination of the increased vascular permeability
tionated to 38 mCi/m2 at 15-day intervals was unsuccess- from the unlabeled biologically active antibody with
ful because HAMA still developed in the majority of these relatively low doses of radiation, and the activated
patients. The single-dose approach was adapted, with the effector cell mechanism. On other multiple dose proto-
addition of alpha interferon and the higher 75 mCi/m2 131I cols, patients received G-CSF 7 to 20 days following
dose. Again, no objective anti-tumor responses were infusion, or patients underwent immunopheresis using a
seen, but there was substantial bone pain relief seen in goat anti-mouse antibody to reduce the non-bound circu-
several patients. Marrow toxicity was not related to the lating antibody [100]. A high dose study using 150 mCi/
extent of bony involvement. In a high-dose therapy study, m2 given at eight weekly intervals was initiated. Patients
three patients were treated at the initial dose level of had PBSC reinfused when circulating radioactivity reached
100 mCi/m2 131I-CC49, followed by 13.2 Gy TBI given an acceptable level. Thrombocytopenia was successfully
over 4 days, beginning 8 days following RIT. All three managed with this regimen. Multiple doses were limited
patients treated at the first dose level achieved objective by HAMA for the first two patients. The third patient was
tumor response [96]. given cyclosporin which prevented HAMA formation
even after three doses, and she showed evidence of tumor
response [101]. A study now underway with 90Y-DOTA-
Breast Cancer peptide-ChL6 appears to provide enhanced therapeutic
The radiosensitivity of breast cancer, and the results index [102].
reported from the breast cancer trials so far suggest that BrE-3 is an IgG1 antibody directed against the peptide
RIT may play a clinical role, particularly with some of the epitope of the MUC-1 antigen, present on 95% of breast
additional strategies to improve response rates that are cancers that have been evaluated for RIT labeled with 90Y.
still under investigation. Although non-myeloablative DeNardo reported objective responses in three of six
doses have produced disappointing results, ASCT studies patients treated at single low doses. High tumor doses
have shown that 100–300 Gy can be delivered to meta- were estimated, however HAMA developed in five of six
static sites with non-toxic doses to normal organs [97]. patients [103]. Schrier treated nine heavily treated patients
L-6 is an antibody directed toward an antigen expressed with stage IV breast cancer with 90Y-labeled BrE-3 anti-
on ∼50% breast adenocarcinomas, with biological activ- body [104]. Harvested autologous marrow or PBSC were
ity that has been evaluated using 131I and [90]Y. Preliminary reinfused at 15 days in conjunction with G-CSF. Fifteen
work by DeNardo et al. demonstrated the importance of and 20 mCi/m2 90Y was administered. Pharmacokinetic
administering unlabeled antibody prior to radiolabeled data indicated that the 90Y biodistribution was not identi-
antibody to saturate the antigenic sites on normal tissues, cal to the 111In biodistribution. Six patients developed
in this case the vascular endothelium [98]. In a phase I transient grade 4 marrow toxicity but all recovered and no
study, 200 mg cold blocker was administered with 131I other toxicities occurred. The 90Y conjugated with
murine L-6. The cold blocker reduced lung activity from MX-DTPA dissociated from the antibody and deposited
19% to 4%, allowing the radiolabeled antibody to be in the bone, causing the high incidence of marrow toxic-
visualized at the tumor sites. It appeared that a transient ity, bone biopsies confirmed this localization of 90Y in the
inflammatory reaction increased the delivery of subse- bone. Based on 111In imaging, tumor doses 35–56 cGy/
quently administered radiolabeled antibody to tumor. mCi were reported. These values in terms of cGy per mCi
474 Targeted radionuclide therapy of cancer

are higher than with other radioimmunoconjugates, but marrow toxicity was observed at 22 mCi/m2, and a study
the total dose that could be administered was limited by with stem cell support is ongoing. Results are prelimi-
the marrow toxicity. Four of eight evaluable patients nary but appear encouraging in the six first patients
achieved a PR, noted in lymph nodes, skin and marrow studied [109].
lesions, and one achieved a clinical CR. Because of
immunogenicity, a human BrE was developed, and nine
patients received increasing doses of 90Y-hBRe with
Glioma
peripheral blood stem cell support. Although the initial The high-grade malignant gliomas (anaplastic astrocy-
response rates are less encouraging, this study may define tomas and glioblastoma) have a very poor prognosis
the second organ of toxicity [105]. with current surgery, radiotherapy and chemotherapy
CC49 is an antibody that targets the TAG-72 antigen, approaches. The use of specific monoclonal antibodies
which is expressed in 90% of breast carcinomas. labeled with a suitable isotope (131I or 90Y) represents an
Mulligan studied nine patients with 177Lu-DOTA-CC49 effective approach to reduce tumor regrowth. Because
(five of whom were breast cancer patients). Luticium-177 of the difficulty in interpreting diagnostic imaging scans,
was considered an attractive alternative radionuclide and the short survival time in these patients (median 10
with a lower energy beta emission and longer half life months) survival has been used as an endpoint.
than 90Y [106]. However, 177Lu accumulated in the cells Radiation, following surgical resection, offers a several
of the reticuloendothelial system and was retained there month advantage in survival time, but has been limited
for a prolonged period. Images demonstrated the tumor by normal brain tolerance. Targeted radiation is there-
uptake as well as activity in the bone marrow. This fore an attractive approach to increase radiation dose to
resulted in myelosuppression at low doses and the MTD tumor and potentially contribute to increased survival.
was 15 mCi/m2. Reduction in the RES uptake would be Some investigators have injected the antibodies intrave-
required for this to become a useful radioimmunoconju- nously, or have tried to increase the tumor/background
gate. Nine breast cancer patients were also treated with ratio with the avidin/biotin system. In several studies the
131
I-CC49 in conjunction with Thiotepa and total body labeled monoclonal antibodies were injected directly
radiation, followed by stem cell rescue. No life threaten- into the tumoral bed after surgery.
ing toxicities were noted and two patients had a partial
response by imaging criteria [107].
The biological response modifier alpha interferon is
Systemic Injection
able to increase the antigenic expression of TAG-72 and Kalofonos reported a study with 131I-anti-epidermal
increase antibody targeting in tumors [81]. Murray growth factor receptor antibody (EGFr) [110]. Ten
tested this in 15 patients with breast cancer, assessing patients with recurrent gliomas received intravenous or
TAG-72 expression and 131I-CC49 uptake [108]. After 3 intra-arterial administration of the radioimmunoconju-
days of alpha interferon, 3 million units daily, 10 or gate and six showed clinical improvement. Brady treated
20 mCi 131I-CC49 was administered and 48 h later a 101 patients with high grade gliomas by intravenous or
biopsy was taken and compared with a pre-study biopsy. intra-arterial injections of 125I-labeled anti-epidermal
The TAG-72 expression was increased by 45% follow- growth factor receptor antibody, EGF-425 [111].
ing the alpha interferon (p < 0.05), and there was an Patients with both primary and recurrent astrocytomas
apparent increased 131I-CC49 localization in tumor. and glioblastoma multiforme were treated with an aver-
However, because of intra- and inter-patient variability age total dose of 139 mCi 125I, administered divided into
in percentage of tumor cells in the biopsy specimens as three weekly infusions. Brady reported 1 CR and 2 PR
well as heterogeneity of TAG-72 expression, this was of short duration following 125I EGF-425 intravenously.
difficult to evaluate with certainty. Pharmacokinetics The median survival in both groups was improved with
and biodistribution were changed with administration of these relatively radioresistent tumors. Fifteen of these
the alpha interferon. RIT at the MTD with concurrent patients had recurrent malignant astrocytomas and were
alpha interferon administration is under investigation treated with 25 to 130 mCi intra-arterially. The median
and in a preliminary report, 1 of 15 patients had achieved survival was 8 ± 7 months [111].
a PR at the expense of increased marrow toxicity, from Twenty-five patients with malignant astrocytomas were
a more prolonged serum clearance at the higher doses. then treated in a Phase II adjunctive therapy trial [112].
Wong et al. evaluated the high affinity, anti-CEA chi- Four to 6 weeks following surgical debulking (2) or biopsy
meric antibody, cT84.66 in 22 breast cancer patients, in (13), and external beam radiation, one to three doses
a dose escalation trial with 90Y-DTPA. Dose limiting of 125I-labeled antibody 425 was administered intravenously
John M. Pagel et al. 475

or intra-arterially at 7–14 day intervals. Cumulative doses created resection cavity from primary or metastatic
ranged from 40 to 224 mCi. HAMA did not develop in any brain tumor [115] High retention in the cysts with little
of these patients. The 1 year survival was 60% with a systemic dissemination resulted in absorbed dose esti-
projected median survival of 15.6 months. mates of 127–703 Gy. No hematological or neurotoxic-
ity was observed. In preliminary results, all five patients
with recurrent cystic glioma appeared to benefit from
Compartmental Injection the treatment with an increased survival. In the majority
Malignant gliomas tend to spread by local invasion of patients with administration into the surgically cre-
rather than metastases. Thus intralesional, intra cavitary ated cavity, stabilization of disease was observed [116]
or intrathecal administration of the therapeutic radiola- Chimeric 81C6 carrying the alpha emitter 211At is also
beled antibody may be useful to deliver the therapeutic under study for injection into the surgical cavity of
radiation directly to these solid tumors. recurrent gliomas. Dose limiting toxicity has not yet
been established [117].
ERIC-1 antibody was radiolabeled with up to 60 mCi
Intralesional 131
I and administered to nine patients with relapsed
One approach is to inject the radioimmunoconjugate glioma who had a cyst or cavity following prior resec-
through an Ommaya reservoir into the surgical cavity tion [118]. In two patients with cystic lesions, the need
following surgical resection, or directly into a cystic for aspiration was markedly reduced. The short range of
tumor when the lesion has a predominantly cystic com- the 131I beta particles delivers a high dose to only a rim
ponent. Here the antibody binding to antigen serves to of tissue approximately 1 mm thick, around the surgical
prolong retention of the radiation at the target, while cavity. In a study with 90Y-labeled ERIC-1, 15 patients
sparing normal brain tissue. Initial studies with 131I anti- were treated with up to 18 mCi and the cerebral edema
bodies into the surgical cavity demonstrated the feasi- that developed was managed successfully with dexam-
bility of an intralesional or intracavity approach ethasone [119]. Dose estimates from this treatment
following surgical resection. Riva et al. [113] reported ranged from 55 to 351 Gy, depending on whether com-
their experience with intralesional administration of plete binding on antibody occurred or not. Again in this
131
I-labeled antibodies for treatment of both newly diag- study, the two patients with cystic tumors required less
nosed and recurrent glioma. Anti-tenascin antibodies, frequent aspirations. While the overall benefit of the
BC-2 and BC-4 were radiolabeled with up to 65 mCi of RIT could not be easily determined from this group of
131
I, and injected into the resection cavity through a patients, median survival was 6 months from treatment.
catheter. The radioactivity remained at the target site for
an effective half time of 60 h and thus could deliver high
doses to the tumor, on average 42,000 rad/cycle. Up to
Intrathecal
four cycles were given to 50 patients. Although HAMA In neoplastic meningitis, external beam therapy is lim-
developed in some patients, it did not interfere with ited by dose to the normal nervous system. The average
tumor targeting in subsequent cycles. In all there were 3 survival of patients with neoplastic meningitis is 3
CRs, 6 PRs, 11 tumor stabilizations and 19 tumor pro- months from diagnosis. Tumor cells are found floating
gressions recorded. Eleven patients with no radiological freely in the CSF or in sheets lining the meninges. Thus
evidence of disease at the time of treatment remained an intrathecal route of administration bypasses the prob-
disease free. In 26 patients the median time to progres- lems of access to tumor antigen that are present when
sion was 3 months, and in the group of newly diagnosed radioimmunoconjugates are administered systemically.
patients who were treated, time to progression was 7 Papanastassiou treated patients with diffuse leptome-
months. The median survival was 20 months; 17 months ningeal deposits with intrathecal radiolabeled antibod-
in patients with bulky disease, and 23 months in patients ies administered through an Ommaya reservoir. Patients
with minimal or microscopic disease. The median sur- included those with carcinomatous meningitis (n = 7),
vival of these patients is usually 10 months; thus this primitive neuroectodermal tumors (n = 18), and CNS
study suggests an improved outcome for these patients. leukemia (n = 22), with the antibody depending on the
In addition, Riva has reported an improved survival in disease [120]. The radioimmunoconjugate leaves the
the patients with bulky glioblastoma, 30 months [114]. intrathecal space relatively quickly, peak blood flow lev-
Another anti-tenascin antibody, 81C6, radiolabeled els of 6–48% of the injected dose reached the vascular
with up to 100 mCi 131I has been studied in patients with system at 24–56 h following injection. Dosage of 131I
recurrent cystic gliomas and in patients with a surgically ranged from 17 to 90 mCi on 1.7 to 9 mg antibody.
476 Targeted radionuclide therapy of cancer

Transient aseptic mengitis was common following Peptides used for tumor targeting show several advan-
intrathecal RIT, occurring in approximately 60% of tages over antibodies: peptides are small and show rapid
patients. Occasional patients developed seizures. No diffusion into (target) tissues resulting in rapid pharma-
long term sequelae have been observed. Significant cokinetics. Their fast blood clearance could lead to high
myelosuppression occurred in several patients who tumor-to-background ratios shortly after administration
received more than 54 mCi. Response was difficult to of the radiopeptide [122–126]. As exemplified by soma-
assess and was assessed by clearance of cells from the tostatin, analogs of the peptide that are stable in blood
CNS and clinical improvement. Papanastassiou reported show a better tumor uptake than the natural peptides
a 33% overall response rate. This was poorest in the car- although both are degraded after internalization [124].
cinoma patients. The mean time to relapse in 37% of To prevent internalized radionuclides from externaliza-
PNET patients was 10 months, In patients with leuke- tion, a so-called residualizing label consisting of a che-
mia, marked responses of clearance of leukemic cells lator linked to the peptide and a radiometal such as 111In,
177
from the CSF were observed, but for only 4–8 weeks. Lu or 90Y is used. These radiometal/chelator com-
Brown et al. reported on patients with leptomeningeal plexes, are retained in the lysosomes and thus they are
disease and brain tumor resection cavities with subarach- trapped inside the cell [126].
noid communication. Patients with anaplastic gliomas, To date, the 111In-labeled somatostatin analog oct-
ependyomas, medullablastoma and anaplastic astrocy- reotide (OctreoScan®) is the most successful radiopep-
toma received a single dose of up to 100 mCi of 131I 81C6 tide for tumor imaging and has been the first to be
antibody administered intrathecally through an Omaya approved for diagnostic use. It is considered the diag-
reservoir [121] No nonhematological toxicity occurred nostic gold-standard in several types of gastrointestinal
and the MTD was 80 mCi, limited by hematological tox- and other neuroendocrine tumors (NET) and plays a
icity. Ten of 24 evaluable patient developed HAMA, but crucial role in the diagnosis and follow-up of patients
in two patients who were retreated, there was no altera- with these tumors [127–129]. Furthermore, somatosta-
tion in biodistribution of radiolabeled antibody. In 31 tin receptor scintigraphy (SRS) also contributes signifi-
patients with malignant gliomas, one patient showed a cantly to decision making in the management of patients
radiographic PR and disease stabilization occurred in with NET [130–133]. Currently, octreotide-based soma-
42% of patients, representing an apparent increased sur- tostatin analogs labeled with the ß-emitters 90Y or 177Lu
vival rate although this was difficult to evaluate. are undergoing clinical trials for peptide receptor radio-
Bigner used the (Fab’)2 fragment of Mel-14, anti- nuclide therapy (PRRT) of NET.
melanoma antibody directed against anti-proteoglycan Other peptides for tumor targeting in development or
chondroitin sulfate associated protein, to treat a patient undergoing clinical trials are CCK2 receptor-binding
with a brain metastasis from melanoma [116]. The peptides, bombesin, substance P, neurotensin, glucagon-
patient received Mel-14 F(ab’)2 labeled with 37 mCi 131I like peptide-1, and RGD-peptides for imaging of ανβ3
injected into the surgical resection cavity, and achieved expression in tumors [126, 133–136].
a complete local response. Eleven patients were treated
with up to 80 mCi 131I-Mel-14 (Fab’)2 and 81C6 intrath-
ecally via lumbar puncture. They had melanoma (n = 8),
Regulatory Peptides and their Receptors
melanosis (n = 1), oligodendroglioma (n = 1) and glio- Regulatory peptides are potent small messenger mole-
blastoma (n = 1). One patient at 80 mCi had hematologi- cules binding to specific receptors. Usually, they are not
cal toxicity. After treatment, three patients had complete larger than 30–40 amino acids and are able to rapidly
CSF responses, two had partial responses radiologically extravasate and penetrate tissues. Regulatory peptides
and survival ranged from 1 to 11 months, an apparent are synthesized mainly in the brain and gastrointestinal
improvement on the 3 month average survival. tract. They may also play a role in the peripheral ner-
vous system or in the immune system [125, 126, 137].
Due to their hydrophilicity, they do not cross the blood–
brain-barrier in either direction. Therefore, the central
Peptide-Based nervous system and the periphery (e.g., the gastrointes-
Radiopharmaceuticals tinal tract) form two independent regulatory systems
which thus use the same messenger molecules without
After the detection of peptide receptor expression by any danger of confusing interaction of both. All regula-
tumors, somatostatin analogues became the first radiola- tory peptides bind to and act through transmembrane G
beled regulatory peptides used for specific tumor targeting. protein-coupled receptors. The signal transduction of
John M. Pagel et al. 477

these receptors is triggered by the binding of the respec- and thus are retained inside the cells. The most widely
tive peptide to the extracellular domain of its receptor used radiometal chelators are DTPA (diethylene-triamin
which will, in turn, activate the intracellular guanine e-pentaacetate) and DOTA (tetra-azacyclododecane-
nucleotide binding proteins (G proteins). An important N,N’,N”,N” ’-tetra-acetate). For diagnostic purposes,
inactivation pathway is the internalization of the ligand– the DTPA can stably chelate 111In. This chelator is used
receptor complex, physiologically leading to either deg- in the commercially available somatostatin analog
radation or recycling of the internalized receptor to the OctreoScan®. However, chelation of other radiometals
cell surface [125]. This inactivation pathway plays a by DTPA used for therapeutic purposes is not suffi-
crucial role for the use of radiopeptides in scintigraphy ciently stable in vivo. In contrast to DTPA, the cyclic
and PRRT. chelator DOTA demonstrates sufficient stability for a
Regulatory peptides influence many aspects of mam- broad range of other metallic ions, such as 90Y, 177Lu,
153
malian physiology. Their role as flexible messenger Sm, 212/213Bi, or 212Pb [139, 140]. Therefore, it is the most
molecules requires rapid degradation in the blood to frequently used chelator in PRRT.
prevent prolonged action of secreted peptides. Radiometal chelators such as DTPA or DOTA may be
Ubiquitously occurring peptidases are responsible for covalently linked to the N-terminus of a given peptide or
rapid deactivation of regulatory peptides. For radiopep- to the ε-amino group of a lysine moiety. If the N-terminus
tides, rapid degradation in the circulation or in other tis- is essential for receptor-binding or internalization, a
sues would be a major obstacle for reaching the target. lysine moiety within the primary structure has to be used
Thus, peptides used for tumor targeting should be meta- for linking the chelator. Even if lysine is already part of
bolically stable. the peptide, it may be crucial for binding or internaliza-
tion. Thus, introduction of an additional lysine may be
applied to conjugate the chelator to the peptide. Thus, the
Radiolabeling of Peptides problem of development of stable analogs of (regulatory)
The somatostatin analog octreotide was the first peptide peptides not only consists of the labeling alone, but also
used for the scintigraphic detection of NET. Somatostatin of changing the amino acid sequence to obtain a site for
itself has a very short half-life in serum. Thus, octreotide linking the chelator without decreasing the receptor
with its higher stability was used for scintigraphic pur- binding affinity. Furthermore, after modification some
poses. As the easiest method of radiolabeling peptides is peptides will still bind, but will no longer internalize any
radioiodination using 125I, 123I, or 131I [138] a tyrosyl moi- more. Internalization of the peptide/receptor complex is
ety was introduced into the stabilized peptide in position 3, considered a crucial step in the process of in vivo recep-
replacing phenylalanine in the natural sequence. tor targeting with peptides [141], although it has recently
Iodination of the tyrosine residue is relatively easy. Thus, been shown that noninternalizing antagonists could also
123
I-Tyr3-octreotide was the first radiopeptide used for lead to efficient receptor targeting [142].
imaging of NET in a patient [123]. If an amino-acid is to As stated above, peptides for receptor targeting should
be replaced by tyrosine for radioiodination, it should not be metabolically stable. In contrast to other drugs, stabi-
be involved in receptor-binding or activation [126]. lization by binding to serum proteins is not helpful in
However, after internalization, peptides will undergo this context – due to higher blood retention and liver
proteolytic lysosomal degradation. Mono- or di-iodi- clearance. Although high serum stability is warranted,
nated tyrosine will be formed and these metabolites will high tumor-to-background ratios will only be obtained
rapidly be excreted from the cells. Mono- or di-iodinated if there is rapid clearance from the blood. Thus, amino
tyrosine may then be deiodinated, preferably in the liver. acids may be replaced by D-isomers so that serum pep-
Excretion of the radioiodine label from the targeted cells tidases will not affect these modified peptides. Other
is the major drawback of the use of this so-called “non- typical modifications for stabilization are the use of
internalizing” radiolabel in peptide receptor targeting. unnatural amino acids, N-terminal acetylation, or
Thus, the tumor-to-background ratios that can be C-terminal amidation [143]. Only after internalization,
obtained using this approach are suboptimal and the sen- lysosomal degradation will occur. For rapid renal excre-
sitivity of 131I-octreotide for detecting somatostatin tion, hydrophilicity is required. Lipophilic peptides will
receptor expressing tumors was inferior to that of pente- show a higher background activity possibly masking
treotide labeled with the radiometal 111In [124]. tumors in highly perfused organs such as the lungs or
In contrast to radioiodinated amino acids, following the liver in early images. Furthermore, higher liver
internationalization by the target cells, radiometal chela- extraction will even increase liver activity which is not
tor conjugated amino acids are trapped in the lysosomes warranted in abdominal tumors.
478 Targeted radionuclide therapy of cancer

Clinical Experience with Somatostatin tive tumors, but not sst1 and sst4 receptor-positive tumors.
A broad variety of tumors express the somatostatin receptor
Analogs
subtypes 2 and 5, which includes most neuroendocrine
As the physical half-life of 111In is 2.8 days, whole body tumors, lung cancer, breast cancer, differentiated thyroid
images should be acquired not only 4h but also 24-h cancers, but also meningiomas, well-differentiated astro-
after injection of 200 MBq of the 111In-radiopeptide. cytomas, pituitary tumors, or malignant lymphomas and
Due to continuous clearance from non-target tissues, the several others [124, 151–154]. SRS plays a major role in
tumor-to-background ratio will increase over time so the diagnosis of neuroendocrine tumors and their metasta-
that the 24-h images show a higher diagnostic yield than ses. In some gastrointestinal NET, it is considered the
the 4-h images. Twenty-four-hour scans show a mark- diagnostic gold-standard [127–129]. In endocrine tumors
edly higher sensitivity [144]. However, image acquisi- of the pancreas, SRS is positive in 50–90% of the cases,
tion at both time points might help to differentiate dependent on the tumor type. As many insulinomas do not
specific from non-specific uptake. Although 99mTc is the express the subtypes 2 and 5 of the somatostatin receptor,
preferred radionuclide in diagnostic nuclear medicine the sensitivity of SRS is only 40–60% [127]. On the other
(ideal γ-energy of 140 keV, high photon flux, relatively hand, virtually all gastrinomas express somatostatin recep-
low radiation burden, unlimited availability), 111In is tors in a high density and indeed, SRS has a sensitivity of
also quite suitable. Obviously, gamma cameras should up to 90% in these tumors. Carcinoid tumors of the intes-
be equipped with medium energy collimators, because tine may also be detected using SRS with sensitivities
of the higher γ-energy of 111In in comparison to 99mTc. >80% in comparison to only ∼50% for morphologic imag-
The scanning speed should not exceed 5–6 cm per min- ing modalities. Even if CT, MRI, and ultrasonography are
ute to obtain high quality images with high count rates. combined, they will not reach the sensitivity of SRS in
Additional three-dimensional SPECT images (single patients with gastrointestinal NET [127, 145]. Only endo-
photon emission computed tomography) may further scopic ultrasonography will perform better in the detec-
increase the sensitivity and should thus be acquired rou- tion of pancreatic NET. Figure 1 shows an example of a
tinely [144, 145]. If bowel activity obscures abdominal SRS in a patient with an ileal carcinoid.
tumor manifestations, additional 48-h scanning can be The sensitivity of SRS is mainly dependent on the
performed, possibly combined with the use of laxatives expression of the sstr2 and 5 subtypes of the somatosta-
to accelerate the passage of intestinal radioactivity. tin receptor. Thus, insulinomas will be visualized in
Organs with physiologic octreotide-uptake are kidney, only a limited number of patients with a sensitivity
liver (later gall bladder and bowel because of biliary around 50%. Gastrinomas will be visualized with higher
excretion), spleen, thyroid, and pituitary [126]. sensitivities of at least 60–90%, dependent on their size
Lymphocytes (and activated leukocytes) and epithel- [155–157]. It has been shown that the scanning tech-
oid cells in granulomas express somatostatin receptors nique has a strong influence on the results of SRS in
so that scar tissue and granulomas could show up in pancreatic tumors as they may be small and undetect-
SRS [146, 147]. Lymphocytes are also responsible for able on planar images [145]. Thus, using SPECT, the
the splenic uptake. After irradiation of the mediastinum, sensitivity can be expected to be closer to 90% than to
“non-specific” uptake may also be observed. However, 60%. Endoscopic ultrasonography is the only imaging
this uptake is caused by receptors on accumulated technique that shows results equivalent to SRS or even
leukocytes and epitheloid cells [148]. better [127].
Cold somatostatin may also decrease tumor uptake Active and inactive carcinoid tumors of the gastroin-
thus possibly leading to false-negative results. If a patient testinal tract may be detected by SRS with high sensi-
is on medication with somatostatin for inhibition of tivities exceeding 80% [124, 126, 127, 145] Especially
tumor growth [149], receptors may be blocked by cold metastatic disease may even be detected by SRS when
somatostatin so that the splenic uptake drops in compari- morphologic imaging modalities fail to show tumors in
son to the liver uptake. This is an important finding in the unexpected locations or outside their anatomical range.
evaluation of somatostatin scans [132, 145]. Furthermore, SRS can provide information about the
Octreotide, the metabolically stable somatostatin ana- somatostatin receptor status of tumors in vivo. This is
log, has high affinity for the sst2 and sst5 receptor sub- relevant for the decision whether treatment with cold
types, while it has no affinity for subtype 1 and 4 and somatostatin is potentially useful or not. In summary, in
affinity to subtype 3 is low [125, 150]. Thus, clinical patients with carcinoid tumors of the intestine, SRS
somatostatin receptor scintigraphy with 111In-DTPA- should be used first in the diagnostic process together
octreotide mainly visualizes sst2 and sst5 receptor-posi- with abdominal ultrasound [128, 129].
John M. Pagel et al. 479

Figure 1. SRS (24-h scans) of a patient with a carcinoid of the ileum with metastases mainly to liver and bone. Anterior
view on the left, posterior view on the right. Uptake in the liver metastases is high, uptake in the tumor in the left shoulder is
comparable to that in normal liver tissue

Other Somatostatin Analogs available OctreoScan® (111In-DTPA-D-Phe1-octreotide).


Further clinical studies are necessary to finally deter-
Besides octreotide, other somatostatin analogs have mine whether this promising compound may be able to
been approved for clinical use or are under clinical evalu- replace 111In-labeled somatostatin analogs in clinical
ation. Depreotide, a 99mTc-labeled somatostatin analog routine [162, 163].
(Neotect®) has been registered in the US for the charac- Lanreotide is an 111In-labeled somatostatin analog that
terization of solitary pulmonary nodules. Depreotide is a may also be used for cold somatostatin therapy as an
synthetic peptide with a cyclic, 6 amino-acid sequence alternative to cold octreotide (Sandostatin®) [149].
[158, 159]. It is a cost-effective alternative to FDG-PET. Although it was reported that lanreotide has a higher
In comparison to 111In-labeled somatostatin analogs, the affinity for sstr3 as compared octreotide [164], these data
shorter physical half-life of 99mTc allows a simplification could not be confirmed by others [165]. As lanreotide is
of the imaging protocol. A higher activity is injected more lipophilic than octroetide, tumor-to-background
(700–800 MBq) and diagnostic images can already be ratios may be lower than with octreotide [166] Lanreotide
obtained 90–240 min post injection [158, 160] including does not show clear advantages over octreotide.
SPECT. In comparison to CT-guided biopsy, 99mTc-dep-
reotide shows a comparable sensitivity while the specific-
ity is lower [159], due to uptake into inflammatory tissue, Other Peptides for Tumor Targeting
such as granulomas. The specificity of 99mTc-depreotide
scintigraphy is clearly higher than that of CT [161].
Gastrin
Another interesting 99mTc-labeled somatostatin ana- Although SRS is a very potent tool in the diagnosis of
log is 99mTc-HYNIC-D-Phe1-Tyr3-octreotide. It shows NET, it is of only limited value in patients with medul-
favorable results as compared to the commercially lary thyroid carcinoma (MTC) due to limited sstr2
480 Targeted radionuclide therapy of cancer

expression of these tumors. Given the outstanding sensi- demonstrate metastatic MTC with the highest sensitiv-
tivity of the pentagastrin test in the detection of persist- ity in comparison to PET, CT, and SRS [172]. Gastrin
ing or recurrent MTC, the expression of the corresponding receptors are also expressed in many gastrointestinal
receptor type in human MTC was postulated [167–169]. NET [173]. A study comparing SRS with gastrin recep-
Indeed, more then 90% of medullary thyroid carcino- tor scintigaphy (GRS) showed that GRS was positive in
mas (as well as other tumors) were found to express about half of the patients in whom octreotide uptake was
CCK2 receptors [170, 171]. After initial evaluation of missing [172]. Furthermore, more than 20% of all
several radioiodinated gastrin-derived compounds, a patients had higher uptake of gastrin than of octreotide
DTPA-derivatized compound with the residualizing into the tumor lesions. Figure 2 shows a gastrin scan in
111
In label was developed [126]. First clinical studies comparison to a somatostatin scan in a patient with a
show that 111In-DTPA-D-Glu1-Minigastrin is able to bronchial carcinoid.

Figure 2. Patient with a bronchial carcinoid with metastases to bone, liver, and mediastinum. Gastrin scan on the left, somatostatin
scan on the right. Both anterior view, obtained 24 h after injection
John M. Pagel et al. 481

Vasoactive Intestinal Peptide (VIP) from pancreatic ß-cells), but also by other NET like gas-
trointestinal carcinoids [173]. For scintigraphic imaging,
VIP receptors are expressed by most of human epithe- GLP-1 and its analogs exendin 3 and exendin 4 seem to
lial cancers lacking expression of somatostatin recep- have potential [135]. Especially for diagnosis and treat-
tors or of the subtypes SSTR 2 or 5 detectable by SRS ment of insulinoma, GLP-1 might offer new possibilities.
[125]. Thus, VIP receptor scintigraphy was thought to Exendin-4 was modified C-terminally and conjugated
have great potential for diagnosis and possibly even with DTPA. This Exendin-4 analog labeled with
treatment of a wide range of tumors with high incidence, 111
In-had extremely high uptake in insulinoma lesions in
especially colorectal cancer, gastric cancer, and exo- mice and even showed therapeutic efficacy when higher
crine pancreatic cancers. In comparison to somatostatin activity doses were administered [182, 183].
analogs, biodistribution of VIP shows high pulmonary RGD peptides: RGD peptides are a particularly inter-
uptake due to high levels of physiologic VIP receptor esting entity of tumor-targeting agents. RGD-peptides
expression in the lungs. Abdominal uptake is lower in contain the amino acid sequence Arg-Gly-Asp and bind
comparison to octreotide [174]. In clinical studies, to ανß3 integrin. ανß3 integrin is a heterodimeric trans-
Virgolini and co-workers showed that primary tumors membrane proteins involved in cell-cell interaction and
and metastases of intestinal adenocarcinomas as well as mediating cellular adhesion to extracellular matrix pro-
NET could be detected with 123I-labeled VIP [175, 176]. teins such as vitronectin, fibronectin, a.o. [184, 185]
However, in a study of Hessenius et al. no adequate 123I- RGD peptides inhibit neoangiogenesis in growing
VIP uptake into the tumors was observed [177]. These tumors [186]. Specific tumor targeting of radiolabeled
findings were confirmed by autoradiographic studies of RGD peptides could be demonstrated in several animal
resected tumors demonstrating lower VIP receptor models for melanoma, breast cancer, osteosarcoma,
expression in the tumors in comparison to physiologic pancreatic tumors, and ovarian cancer [136, 187–190].
VIP receptor expression in the surrounding tissues. RGD peptides clearing via kidney instead of hepatobil-
iary secretion have been developed. These peptides were
Neurotensin labeled with 99mTc-HYNIC and 111In-DOTA for tumor
imaging and evaluation of biokinetics in a mouse model
Pre-clinical data suggest that 111In-DTPA and 111In-DOTA of a human ovarian cancer xenograft. Target-to-
labeled neurotensin analogs may be of high value in the background ratios as high as 92:1 and 26:1 were
management of patients with exocrine pancreatic cancer achieved using this model [136]. Furthermore, the
[134]. Buchegger et al. have studied the use of Tc-99 m 90
Y-labeled compound showed inhibition of tumor pro-
labeled neurotensin analog NT-XI in patients with ductal gression in an in vivo mouse model. It has been pointed
pancreatic adenocarcinoma. Tumor was only visualized out that RGD peptides seem to be an interesting more
in one out of four patients [178]. general approach to the diagnosis and treatment of a
variety of tumors as neoangiogenesis is not limited to
one specific tumor entity only. However, one has to keep
Bombesin
in mind that the most effective targeting with radiola-
Receptors for bombesin are expressed on colon beled RGD peptides has been demonstrated in mouse/
cancer, glioblastoma, prostate cancer, and SCLC [126]. tumor models in which the tumor cells themselves
Radiolabeled bombesin potentially could be used to express the ανß3 receptor on their cell surface.
visualize prostate cancer. After the development of radio-
metal labeled metabolically stable bombesin analogs Peptide Receptor Radionuclide Therapy
with high receptor affinity, clinical studies are currently
undertaken [179, 180]. A new 99mTc labeled bombesin (PRRT)
analog (demobesin 1) showed very favorable results in a For PRRT, radionuclides with high cytotoxic potential
human prostate cancer mouse model with high prolonged that can be stably chelated are used. The ß-emitter 90Y
tumor uptake [181]. Clinical evaluation of this com- has been the first radionuclide to be applied for PRRT in
pound will have to shows its value. humans. Alternative radionuclides are the ß-emitter 177Lu
and the auger-electron emitter 111In (which is usually
used at lower activities for diagnostic purposes because
Glucagon-like Peptide-1 (GLP-1) of its γ-emission). Other interesting candidates are
GLP-1 is an incretin hormone that induces postprandial α-emitters such as 213/212Bi, or 211At. However, no stable
insulin secretion from pancreatic ß-cells. Thus, receptors and secure labels with α-emitters are available for clinical
for this peptide are expressed by insulinomas (deriving application in PRRT at the moment [126, 191, 192].
482 Targeted radionuclide therapy of cancer

So far, most experience with PRRT is based on the include restricted antigen expression on tumor cells,
use of 90Y and 177Lu-labeled somatostatin analogs. the small fraction of the cardiac output which reaches
Dependent on the inclusion criteria, these studies show the tumor (it is estimated that it takes 400 h for all the
that the rate of minor/partial responses is between blood to pass through a 1 g tumor), and poor vascular
10–35%, while the rate of stabilizations of previously permeability because of vascular spasm and high inter-
progressive disease is higher. Interestingly, some stitial pressure from decreased lymphatic drainage that
patients show a response several months after treatment results in a high pressure gradient compromising transport
[193–202]. There is evidence that application of the towards the center [25]. Therefore several challenges asso-
same cumulative activity results in a better therapeutic ciated with RIT exist that limit the radiation absorbed
response if the number of therapeutic cycles is lower dose, and thus limit the therapeutic ratio. The dose is
[198]. In general, PRRT is very well tolerated and only inadequate because of poor penetration of the antibody
a limited number of patients shows relevant haemato- for reasons just mentioned, and the amount of radioac-
toxicity above grade 3. Nephrotoxicity is one of the tivity that can be injected is limited because of marrow
major problems of this therapeutic concept [203, 204], toxicity. Although dose fractionation is used in chemo-
but may be reduced by co-infusion of basic amino acids therapy and with external beam radiation therapy, the
[205, 206]. In comparison to the toxicity associated with development of HAMA precluded multiple doses on
chemotherapy regimens for NET [207], PRRT is toler- antibody, particularly with murine antibody. RIT may be
ated extremely well by the patients while the overall most effective for small tumors which are more vascular,
response rates seem to be higher. but further work is still required to identify the role of
As the therapeutic success with 111In-labeled oct- RIT in the management of cancer.
reotide is rather low, 90Y was preferred as a radionuclide In order to increase the radiation absorbed dose to the
[208–211]. The data on the use of 177Lu labeled soma- tumor, one can approach the issue of the therapeutic
tostatin analogs are extremely promising as the rate of ratio by either increasing the dose intensity or increas-
complete and partial remissions rises to over 50% in ing the radiation sensitivity of the tumor, or alternately
previously progressive patients. Thus, 177Lu-labeled reducing the toxicity and thereby allowing higher doses
somatostatin analogs seem to be a real progress in the of radioactivity to be administered (Table 4).
treatment of NET [212].

Table 4. Strategies to improve outcome of RIT


Strategies to Improve Outcome Increasing dose Increase radioactivity administered
of RIT intensity Single high dose
Reduce immunogenicity to enable
Targeting radiolabeled Abs and peptides to malignant multiple dosing
cells has led to the establishment of noteworthy princi- Improve the effectiveness of the
ples and encouraging results, yet obstacles still remain administered dose
Increase tumor localization
that must be overcome to achieve optimal targeting and Compartmental administration
elimination of cancer cells by radioimmunoconjugates. Intraperitoneal
For example, the above review indicates clearly that a Intra-lesional
single dose of conventionally radiolabeled murine Intrathecal
Increasing vascular permeability
monoclonal antibody administered at the MTD will not
Hyperthermia
provide sufficient radiation to tumor to be effective as External beam radiation
therapy for patients with solid tumors. In Phase I studies Interleukin-2
with radiolabeled monoclonal antibodies, bone marrow Integrin antagonists
toxicity has been dose limiting and few complete Biological response modifiers
Increasing the dose rate
responses have been seen, even at the MTD. An impor- Smaller molecules
tant reason for the poor results with systemic therapy Pretargeting
using radiolabeled antibodies has been the low accretion Increasing radiation Chemotherapy agents
of antibody in tumors and the fact that antibodies are not sensitivity Halogenated pyrimidines
ideal carriers of radiation to a tumor target. In general, Reducing the exposure Dose fractionation
less than 0.01% of the injected dose localizes to each to normal tissues Immunoadsorption columns
gram of tumor. Explanations for this low accretion Pretargeting
John M. Pagel et al. 483

Increasing Dose Intensity in clinical trials and have established that repetitive
doses of human monoclonal antibodies can be adminis-
Increase Radioactivity Administered tered without evidence of alloimmunization [221]. Thus,
it appears that the problem of HAMA may be solved
The initial approach to increase the dose intensity was to
with human antibodies and dose fractionation may be
administer a single dose of more radioactivity. The dose
becoming more feasible.
limiting toxicity is hematological and can be managed
with transfusions, cytokines and autologous marrow or
peripheral blood stem cell rescue, with reinfusion when Improving the Effect of the Administered
the total body radioactivity is at low levels, usually within dose
10 days. Blumenthal showed that IL-1 and GM-CSF can
allow increased doses of radioactivity to be administered Increasing Tumor Localization
in mice [213]. The addition of hematopoietic growth fac- Because of the physical limitation of accessibility of
tors has aided the recovery of patients undergoing these antibodies to tumors, strategies have been developed to
procedures [214]. In one study, 90 mCi/m2 of 186Re was increase tumor localization. Initially these included
the MTD for a pancarcinoma antibody, and with PBSC changing the labeling procedures, changing the radio-
rescue, doses up to 300 mCi/m2, were used, which was isotope, increasing the mass amount of antibody and
not yet the MTD [215]. Two of three patients with ovarian using an intra-arterial administration, none of which had
cancer treated with peripheral blood HCT achieved a PR, a significant impact. More recently other approaches
whereas there were no responses at non-myeloabaltive have been more successful.
doses [216]. Similarly, 300 mCi/m2 of 131I-CC49 anti-
body was the MTD with marrow rescue [82], compared Compartmental Administration
with 75 mCi/m2 of 131I for this antibody without marrow Intraperitoneal administration of the immunoconjugates
rescue. This approach has increased the MTD, however, for RIT was shown to be feasible, relatively safe, and has
but has not resulted in significantly increased response demonstrated anti-tumor effects as direct tumor cell
rates, and thus additional tactics to achieve tumor regres- exposure results in improved antibody–antigen binding
sion remain necessary. [222]. Several trials have been undertaken in patients
The ability to administer more than one dose of murine with ovarian cancer using intraperitoneal administration.
131
monoclonal antibody has been limited by HAMA [96, I-, 90Y-, 186Re- and 177Lu-labeled antibodies have been
217–219]. Immunosuppressive agents have been inves- used for this purpose [223–228]. Encouraging tumor
tigated to reduce HAMA. Lederman et al. [79] and Lane responses were observed in patients with Stage III ovar-
et al. [78] administered Cyclosporin A to suppress the ian carcinoma who had tumor nodules less than 2 cm in
development of HAMA in studies involving radiola- diameter. Stewart initially reported 5/21 PR following
beled anti-CEA murine monoclonal antibodies with delivery of 131I-OC125 antibodies [228]. However,
modest success. While this was successful in suppress- another study with this radioimmunoconjugate showed
ing HAMA to a F(ab’)2 fragment, this approach has not no benefit [229]. Following intraperitoneal 186Re-labeled
been universally successful with an intact antibody NR-LU-10, 5 of 12 patients with minimal ovarian cancer
[220]. Low-dose cyclosporin, as used by Meredith with achieved a PR [226]. The MTD following intraperitoneal
a highly immunogenic antibody, was unable to signifi- infusion of this radioimmunoconjugate, 150 mCi/m2 was
cantly reduce HAMA following murine CC49 delivery higher than following intravenous infusion (90 mCi/m2)
[95]. Thus, cyclosporin may have some efficacy in because of the reduced blood radioactivity accounting
reducing immunogenicity of murine antibodies in for less marrow exposure [76]. Kavanagh reported anti-
patients, but does not appear to be sufficient to permit tumor effects in patients treated with 90Y-B72.3, and
administration of multiple doses in all patients. increased tolerance to 90Y with administration of EDTA
With modern genetic engineering, chimeric antibod- [230]. Iodine-131-DOTA-CC49 has also been successful
ies have been produced to in an attempt to overcome in achieving tumor responses in patients with <1 cm nod-
HAMA [219]. The immunogenicity of the chimeric ules or extended progression-free survival in patients
antibodies in generally less than murine antibodies, but with occult disease or microscopic disease [223]. 131I
has not been sufficiently reduced to uniformly adminis- MOV-18 was similarly shown to improve survival in
ter multiple dose of RIT [41, 80]. Several humanized minimal disease [231].
antibodies, with only the hypervariable region of the Hird reported anti-tumor activity following 90Y-HFMG
antibody molecule being murine, have now been studied but marrow toxicity limited the administered dose to
484 Targeted radionuclide therapy of cancer

<30 mCi, even with the addition of intravenous EDTA to endothelial cells, either directly or via the activated
chelate the unbound 90Y [225], and in patients with sub- lymphocytes [214]. DeNardo et al. reported that modified
clinical disease there was a prolongation of survival fol- IL-2, PEGIL-2, administered before radiolabeled anti-
lowing one IP dose of 90Y-HFMG [232]. Kosmos body enhanced antibody localization by a factor of two in
reported that immunoglobulin against the chelate pre- mice by increasing vascular permeability to the antibody
cluded more than one treatment using a DOTA chelate [240]. DeNardo also showed the rIL-2 increased the
to 90Y [233]. In a group of 21 patients who had achieved response rate in xenografts treated with 67Cu-Lym-1 by
CR following surgery and conventional chemotherapy, about 20% [241]. In a pre-clinical study, when IL-2 was
one administration of intraperitoneal HMFG1 monoclo- conjugated to an anti-CEA antibody, ZCE025, Nakamura
nal antibody labeled with 18 mCi/m2 90Y improved sur- found that the cytokine function of IL-2 was destroyed,
vival. The median survival had not been reached with a but that vascular permeability specific to the tumor was
maximum follow-up of 12 years and survival at greater increased [242]. Epstein used IL-2 conjugated to TV-1,
than 10 years was 78% [224]. an antibody to fibronectin that targets the basement mem-
Riva reported results in patients with peritoneal gas- brane and causes increased vascular permeability [243].
trointestinal carcinomatosis who received 100 mCi Followed by a radioimmunoconjugate directed against a
131
I-FO23C5 via the intraperitoneal and intravenous tumor-associated antigen, this showed three- to fourfold
route [114]. Therapy was administered in conjunction increased tumor targeting. Increasing the vascular perme-
with Cyclosporin A every 3 months with up to four ability of newly formed vessels also appears to increase
injections. Three complete and six partial responses antibody accumulation. DeNardo et al. used RGB penta-
were observed in 34 patients. Seventeen patients peptide, which is a αvβ3 integrin antagonist and showed
received alpha-interferon to increase the expression of a transient increase of 45–50% deposition of 111In-ChL6
CEA, with an increased the response rate to 59% com- after 24 h in a xenograft study [244].
pared with 29% without alpha-interferon.
Biological Response Modifiers
Increasing Vascular Permeability IL-2 and alpha interferon appear to be successful in
RIT may be most effective for small tumors that are more upregulating the antigen expression of CEA and TAG-
vascular, but manipulations to improve tumor blood flow 72. However, definitive results on the work with alpha
to larger tumors may be worthwhile. The influence of interferon and with IL-2 in clinical trials have yet to be
vascular permeability on antibody uptake and distribu- published. In 1990, Murray et al. showed altered biodis-
tion in tumors has been well documented [234]. Studies tribution and an improved relative tumor uptake of
111
to increase vascular flow or permeability at the tumor In-anti-melanoma antibody in patients who had
using hyperthermia have been carried out by Stickney received alpha-interferon [108]. In other clinical studies
et al. [235]. Hyperthermia reduces interstitial fluid pressure evaluating the effects of alpha interferon, Greiner et al.
and may improve tumor-associated antigen expression. showed increased serum levels of TAG-72 and CEA
Local hyperthermia can also exert direct cytotoxic antigen [245]. Greiner et al. also demonstrated increased
effects, particularly on radioresistant and hypoxic cells levels of both antigens in ascites cells following intrap-
which are nutritionally deprived and acutely acidic, and eritoneal injection of gamma interferon to patients with
also cells in the S phase of the cell cycle. The degree of ovarian cancer and increasing targeting in tumors, but
cell killing depends on the degree and duration of hyper- whether this will translate into increased tumor response
thermia. The results of the preclinical studies to assess rate is still under investigation [246]. Increased response
vascular permeability from external beam radiation prior rates have been noted in patients with breast cancer
to administering immunoconjugate have varied with receiving alpha interferon in conjunction with 131I-labeled
tumor types and location [236, 237]. A three-fold CC49 RIT, and IL-2 have been studied further [81].
increase in antibody uptake was reported when antibody
was administered 1 day following low dose external
beam therapy in patients with hepatoma, without affect-
Increasing the Dose Rate
ing normal liver uptake [96]. Combined RIT/external Increasing the dose rate may be able to be achieved by
beam therapy clinical studies are underway for both using radiolabeled antibody fragments or small molecules
hepatic tumors [238] and head and neck cancer [239]. which penetrate the tumor more rapidly. Early preclinical
Interleukin-2 alone, modified with polyethylene glycol studies supported the theory that total dose delivered may
or conjugated with antibody augments vascular permea- be of primary importance. Thus, the belief that greater
bility and antibody uptake in tumor by a reaction with exposure of tumor to radioactivity as determined by area
John M. Pagel et al. 485

under the curve led to the acceptance of intact antibod- survival advantage in patients with anaplastic astrocy-
ies as the vehicle to carry the therapeutic radiation. toma [260] and responses have been reported in patients
Dose rates effects have been examined by assessing with unresectable liver metastases from colon cancer and
whether radioactivity delivered on an antibody frag- FudR with external beam hepatic irradiation [261].
ment that reaches the target earlier than using an intact
antibody. Fragments may provide an advantage compared
with the slower localization of intact radiolabeled anti-
Reducing the Exposure to Normal Tissues
bodies, even though the retention of the intact antibody at Several strategies have been proposed to reduce the
the tumor is longer. Behr et al. compared response rates exposure of the marrow to radiation. Dose fractionation
of patients with CEA expressing tumors who received the appears to have an impact on reducing marrow toxicity,
intact antibodies and the F(ab’)2 fragment of NP-4 and but repeated fractions are limited by the formation of
MN-14 [75]. The data also showed that more frequent HAMA with murine antibodies as discussed above [34,
responses occurred in patients receiving the radiolabeled 35, 99, 104, 262]. An attempt to reduce toxicity was
fragments, although only minor responses were reported. done by fractionating the intraperitoneal 186Re NR-LU-
Recently studies of 90Y on Octreotide peptide have shown 10 dose and administering a second dose of intraperito-
responses in phase I RIT studies for patients with neu- neal 186Re NR-LU-10 7 days after the first dose [227,
roendocrine tumors [105]. 263]. At the highest dose level of 90 mCi/m2 twice (i.e.,
180 mCi/m2) there was severe marrow toxicity in one of
three patients. In contrast, similar toxicity was seen in
Increasing Radiation Sensitivity two of three patients in the single dose study at only
Radiation sensitizing chemotherapeutic agents such as 150 mCi/m2.
5-fluorouracil (5-FU) and cis-platinum have been used Meredith et al. have incorporated this strategy into sev-
in combination with 30 Gy external beam therapy and eral clinical trials with B72.3 in patients with colorectal
have resulted in improved response rates in carcinoma cancer and with CC49 in patients with colon and prostate
of the esophagus, colon, anus, cervix, bladder, and head cancer, and have confirmed a reduction in myelotoxicity
and neck compared with chemotherapy alone [247]. but the number of infusions was still limited by HAMA
Radiation enhancement with these drugs has also been [80, 95] Another approach is to remove the circulating
seen in vitro with low dose-rate radiation. [248, 249] radioactivity that has not localized at the tumor site by
Drugs to increase radiation sensitivity to RIT have been a clearing agent. Such a clearing agent could either be
studied in mouse xenograft models. Paclitaxel and 90Y- administered intravenously or be extracorporeal, i.e., part
chL6 [250], topotecan and 90Y-huBrE [251], cisplatin of an immunoabsorption column [264]. Candidate clear-
with 131I-323/A3 [252] and 131A33 [249], gemcitabine ing agents include anti-antibodies directed at the Fc por-
and B72.3 [253] have been studied. In pre-clinical stud- tion of the immunoconjugate or the avidin/biotin system
ies, combinations of RIT with paclitaxel have induced with one component linked to the immunoconjugate and
cures without increased toxicity [250] and topotecan the other to the clearing agent [265].
with RIT has shown an increased survival [251]. Clinical The extracorporeal approach is being evaluated with
studies with taxol are in progress. The effects of these external immunoadsorption columns [264]. In this pro-
combination approaches are dependent on the timing cedure, the patient’s blood is separated into cells and
and dosing of the chemotherapeutic agents, and will plasma by a cell separator, phereis machine, and the
require considerable study in clinical trials. plasma is then circulated through a column which spe-
Another approach to increase radiation sensitization is cifically removes the radiolabeled antibody, either by
to use halogenated pyrimidines as radiation sensitizers another antibody, e.g., goat anti-mouse antibody or by
[254]; 5-iododeoxyuridine [255, 256], fluorodeoxyuri- avidin removing a biotinylated radiolabeled antibody.
dine (FUdR) and bromodeoxyuridine (BudR) [257–259] DeNardo et al. have employed some of these approaches
have been most widely studied [255–257, 260]. These and have reported responses in patients with advanced
act as a thymidine analogue that is incorporated into breast cancer with multiple doses of 131I-labeled chime-
DNA via the thymidine salvage pathway. This appears to ric antibody, L6 [264]. Some patients received G-CSF
increase DNA susceptibility to radiation and inhibits 7–20 days following infusion while others underwent
DNA repair. In vitro studies and xenograft studies of immunophoresis using a goat anti-mouse antibody to
these pyrimidines in conjunction with RIT have shown reduce the non-bound circulating antibody. Four of nine
increased tumor growth delay [256]. Clinical studies patients who were able to receive more than one dose
using BudR and external beam therapy have shown a achieved a PR [101].
486 Targeted radionuclide therapy of cancer

Pretargeted RIT Table 5. Potential advantages and disadvantages of


pretargeted RIT systems
Conventionally radiolabeled antibodies or other large Potential advantages Potential disadvantages
molecules remain in the circulation for long periods of
• Favorable biodistribution • Ab constructs may be
time exposing the normal organs, especially the radio- and blood clearance difficult to manufacture
sensitive bone marrow, to radiation. The prolonged cir- • Achieves high • Monovalent (to tumor antigen)
culating radiation limits the dose that can be safely target-to-non-target Ab fragment constructs may
administered and ultimately limits the efficacy of the organ ratios decrease tumor residence time
approach. An alternative approach, pretargeted RIT, has • Flexibility with respect • Tetravalent constructs (e.g.,
to isotope and antigen SA) may cross-link
been used to administer the antibody and the therapeutic target Ab-antigens and internalize
radioisotope separately with some means of bringing • Immunogenicity (e.g., SA)
them together at the tumor. This has been an attractive • Hapten release from
approach since the targeting specificity of the antibody processed Ab may poison
is retained, but the whole body radiation dose is sub- tumor-associated conjugate
• Perceived as complex with
stantially decreased because the radioisotope is admin- respect to dosing and timing
istered on a small molecule, which is rapidly is removed
from the body through the kidneys if it does not bind
(Fig. 3). The potential advantages and disadvantages of followed by the bivalent hapten, labeled with 40 to
pretargeted RIT systems are summarized in Table 5. 200 mCi 131I. The MTD without stem cell rescue was
Goodwin et al. pioneered the use of pretargeting, ini- 100 mCi. For patients receiving >100 mCi, 4 of the 15
tially studying bispecific antibodies. The first step patients had grade III/IV thrombocytopenia. At 12
involved pre-localizing an unlabeled antibody in tumors months, there were 3 PR, one stable disease and nine
that recognized both the tumor associated antigen and a progressions in 13 patients evaluated. The pretargeted
hapten, and then administering a radiolabeled hapten approach with bifunctional antibodies and AES was
that disappeared from the blood stream rapidly [266] also studied in 26 patients with MTC. Biodistribution
With a moderate affinity between the hapten and the was variable among patients, and dose limiting myelo-
antibody, tumor uptake and radiation were sub-optimal toxicity was reached at 60 mCi/m2 131I. Anti-tumor,
and a strategy with a higher affinity system, the affinity minor responses and stablilization of disease were also
enhancement system (AES) was developed [267, 268]. observed [267], but this preliminary study showed no
Here a bivalent hapten is used, that can cross-link the improvement over the conventionally labeled antibody
bispecific antibody bound to the tumor, thus improving as regards tumor response or MTD.
tumor localization of the radioactivity. More recent Another popular approach takes advantage of the
bispecific agents developed include a “universal” mole- ultra-high affinity avidin-biotin system (Kd = 10.15 M).
cule where one arm’s specificity reacts with the chelator Avidin-biotin has been widely used in in vitro applica-
used for various radionuclides [269]. The AES system tions but was first translated into in vivo localizing strat-
has been evaluated for patients with small cell lung can- egies by Goodwin, Meares and McCall [266]. Both
cer. An anti-CEA x anti-DTPA-indium bispecific anti- avidin and its bacterial counterpart streptavidin have
body was injected to assess feasibility, and this was been used in pretargeting applications. Two and three

1st Step
Tumor localization 2nd step
Blood clearance Radio-delivery

mAb
Streptavidin Radiolabeled Biotin

Figure 3. Pretargeted RIT example to remove unbound Ab constructs from the bloodstream prior to the administration of a
therapeutic radionuclide. Divalent Ab-SA conjugate followed by delivery of radiolabeled biotin [278]
John M. Pagel et al. 487

step pretargeting approaches were described with the achieved a higher therapeutic ratio than that achieved
antibody carrying either the avidin/streptavidin or biotin with conventional RIT using the same antibody.
for imaging and therapy [270]. Clinical data with both avidin-biotin approaches indi-
Imaging studies using bifunctional antibodies and the cate a substantial improvement in the tumor-to-marrow
avidin-biotin approach demonstrated the concept and absorbed dose ratios, with acceptable radiation dose to
that improved tumor to background activity ratios could other normal organs. A significant reduction in marrow
be achieved [270–273]. A variety of tumors and anti- toxicity using the pretargeting approach to treat patients
bodies have been studied using this technique. The pro- with Grades III or IV glioma has been noted by Paganelli
cedure has been safe and well tolerated, with improved and coworkers. They used doses of 150 mCi 90Y-DOTA-
sensitivity due to the higher tumor-to-background biotin, about six to ten times the MTD by the conven-
activity ratios in comparison to conventional radioim- tional approach, with negligible hematologic toxicity
munoscintigraphy. The multistep approach, often requiring and no acute or delayed side effects [270, 276]. At 2
72–96 h for completion has not been a practical months, in 45 patients there was a tumor effect in 20%,
approach for imaging purposes. Rather, the potential of 55% of patients had disease stabilization and 25%
this approach may be greater for therapy applications. progressed. The tumor responses were maintained in
A number of approaches have been under study for ther- 11% of patients at 12 months. Grade III/IV marrow tox-
apeutic applications. These include the reverse avidin- icity occurred in two of five patients treated twice at the
biotin approach for targeted radionuclide therapy where highest dose level. Antibody developed to the streptavidin,
the antibody is conjugated to streptavadin rather than and only after repeated administrations to the biotiny-
biotin, and the biotin is linked to the radionuclide. The lated antibody.
preclinical performance of this system has been detailed
by Axworthy and coworkers, who reported 80–100% Pretargeted RIT for Lymphoma and
cures of subcutaneous small cell, breast and colon carci-
noma xenografts (250 mm3) at 90Y doses of up to
Leukemia
800 mCi/mouse with negligible hematologic toxicity Pretargeted RIT was evaluated for treatment of patients
[274]. With conventionally labeled antibodies, 200 uCi with relapsed or refractory NHL. In the first study, the ten
was dose limiting because of marrow toxicity. An early patients enrolled had received prior therapy, including
clinical study evaluated whether an improved tumor-to- high-dose chemotherapy and peripheral stem cell trans-
red marrow therapeutic ratio could be achieved using plant (n = 3); 131I-tositumomab antibody therapy (n = 1);
pretargeted RIT compared with conventional RIT, and and prior rituximab (n = 6). In this preliminary study, chi-
at the same time preserve the efficiency of tumor target- meric, anti-CD20 antibody (C2B8, rituximab) was chemi-
ing [275]. Forty-three patients with adenocarcinomas cally conjugated to streptavidin. Thirty-four hours after
reactive to an anti-EPCAM adenocarcinoma murine the antibody conjugate was administered, a clearing agent
monoclonal antibody, NR-LU-10 were studied. The (synthetic biotin-N-acetyl-galactosamine) was adminis-
antibody–streptavidin conjugate was injected, followed tered to remove non-localized conjugate from the circula-
by a biotin-galactose-human serum albumin clearing tion. A DOTA-biotin ligand, labeled with 111In for imaging
agent, followed by 90Y-DOTA-biotin as the final step for and/or 90Y for therapy was administered 18 h later [277].
therapy. In some patients, the conjugate was radiola- In three patients, the C2B8/SA conjugate was radiolabeled
beled with 186Re as an imaging tracer to assess biodistri- with a trace amount of 186Re in order to assess pharma-
bution of the conjugate and effectiveness of the clearing cokinetics and biodistribution using gamma camera imag-
agent. Indium-111-DOTA-biotin was co-injected with ing. Rhenium-186 C2B8/SA images confirmed that the
90
Y-DOTA-biotin to assess the biodistribution, pharma- conjugate localized to known tumor sites and that the
cokinetics and dosimetry of the 90Y-DOTA-biotin. No clearing agent removed >95% of the conjugate from the
significant adverse events were initially observed after circulation. The images demonstrated tumor targeting of
administration of any of the components. The non- the DOTA-biotin within 10 min after the injection and
tumor-bound antibody-conjugate was efficiently cleared identified previously unknown disease in several patients.
from the circulation by the clearing agent. The mean Localization of radioactivity in normal organs was low
tumor-to-marrow absorbed dose ratio when using the and unbound radiobiotin was rapidly excreted from the
optimized pretargeting schema was 63:1, compared whole body and normal organs. The median tumor-to-
with a 6:1 ratio reported previously for conventional whole body dose ratio of 35:1 was higher than previously
RIT. This initial study confirmed that the pretargeted reported with conventional radiolabeled antibodies.
RIT approach appeared to be safe and feasible, and Seven patients received 30 or 50 mCi/m2 90Y-DOTA-biotin.
488 Targeted radionuclide therapy of cancer

The regimen was safe and well tolerated; only grade I/II objective clinical remissions. With continued research,
non-hematologic toxicity was observed, the most preva- RIT has the potential to find a place as first-line treatment
lent toxicity was primarily fatigue. Hematologic toxicity in radiosensitive tumors, as treatment for small volume
was also not severe; five of the seven patients who received disease, or as a radiation boost in combination with other
30 or 50 mCi/m2 of 90Y-DOTA-biotin experienced only treatments, such as with stem cell rescue or as adjuvant
transient grade III (but no grade IV) toxicity. Although six therapy. Investigators are optimistic that these approaches
of ten patients developed humoral immune responses to may improve the cure rates of the thousands of patients
the streptavidin, these were delayed and transient. Six of with acute leukemia who are diagnosed each year.
seven patients who received 30 or 50 mCi/m2 90Y (51 to
109 mCi total dose) achieved objective tumor regression,
including 3 CR and 1 PR. Although these results are pre-
liminary, they suggested that even heavily treated patients
Summary
with NHL can tolerate high doses of radioactivity with the
pretargeted RIT approach without requiring stem cell RIT is a complex, multidisciplinary effort that still faces
transplantation. many challenges. We have come to understand the obsta-
Recent murine studies using human leukemia xeno- cles and have realized that a single high-dose of a radio-
grafts have demonstrated that a single treatment of pre- labeled moiety administered systemically is unlikely to
targeted 90Y-DOTA-biotin after delivery of an anti-CD45 be successful in curing most cancers. In most of the RIT
antibody-strepavidin conjugate results in a minimal level trials, radiation absorbed doses higher than 30 Gy have
of toxicity, and tumor-to-blood ratios of absorbed radio- only been infrequently reported, and responses cannot be
activity improved by as much as 30-fold over those seen typically induced in the relatively resistant solid tumors.
with a directly radiolabeled Ab [278]. It is estimated that However, these doses may provide clinical benefit in cer-
at least twice as much absorbed radiation can be deliv- tain circumstances, in small volume or subclinical dis-
ered to the marrow and five-times more to the spleen, ease or with additional manipulations. The tumor
using pretargeted anti-CD45 RIT as compared to doses regressions observed in pre-clinical studies and in patients
estimated to be delivered by a directly labeled antibody. with B-cell lymphoma and leukemia have encouraged
Improved reagents for pretargeting have been devel- investigators to pursue antibodies and peptides as vehicles
oped, including a synthetic clearing agent and fusion for targeted therapy. In the hematological malignancies,
proteins that include single-chain Fv fused to streptavidin response rates are high enough and of long enough duration,
monomers (scFv)4−SA [14, 279]. The synthetic clearing that two radioimmunoconjugates for NHL have been
agent consists of a defined dendrimeric type synthetic approved for use in the United States. For hematological
molecule with multiple galactosamine units and a biotin malignancies, there may well be a place for RIT as first
for binding circulating streptavidin containing protein and line treatment with current technology. Although it seems
transferring it to the liver via the Ashwell receptors [280]. that the field of RIT has been moving very slowly, because
Advantages of the synthetic clearing agent are defined by of the multiple obstacles that had to be overcome, there is
reproducible in vivo behavior, lack of biotin release fol- still much optimism for targeted therapy as the newer
lowing liver uptake and lack of competition for tumor approaches are being investigated.
targeting by radiolabeled DOTA-biotin. The fusion protein The inadequate delivery of radiation to tumor has been
improvement that has been validated for several different the major problem compromising RIT and increasing the
antibodies also results in expression of a defined protein fraction of the radioimmunoconjugate that localizes in
by E. coli, retained binding for biotin, comparable tumor the target tumor tissue has been difficult. Pretargeting the
targeting compared to whole IgG antibodies and economic antibody prior to injection of the radioisotope may be
advantages for protein production. These results have one strategy that offers promise for lowering marrow
prompted large-scale cGMP production of the anti-CD45 exposure to radioactivity and increasing the peak dose
fusion protein for pretargeted RIT trials to be conducted in rate to the tumor site. The dilemma of the proper posi-
patients with advanced myeloid leukemias. tioning of RIT and the design of future clinical trials
A preliminary clinical study using 90Y-DOTA-biotin remains. It appears that RIT may be used to treat small
pretargeted with either an anti-CD20 fusion protein disease or as adjuvant therapy for solid tumors, unless
(B9E9FP) yielded encouraging results in 12 patients the approaches described above are shown to be success-
[281]. The ratio of average tumor to whole-body radiation ful in markedly improving the amount of radiation
dose was 49:1 in this study and no significant hemato- deposited at the tumor. Despite the use of new peptides
logic toxicities related to the pretargeted RIT and with in diagnosis and therapy of tumors and non-malignant
John M. Pagel et al. 489

diseases that have already been described, the most excit- 16. James K. Human monoclonal antibodies and engineered antibodies
ing development in the near future may be the combina- in the management of cancer. Semin Cancer Biol. 1990;1:243–253.
17. Schwarz SW, Connett JM, Anderson CJ, et al. Evaluation of a
tion of different peptides and radionuclides in PRRT. direct method for technetium labeling intact and F(ab’)2 1A3, an
New developments include the development of multiva- anticolorectal monoclonal antibody. Nucl Med Biol. 1994;21:
lent peptides that have a higher uptake in the target cell 619–626.
[173], and the development of new peptide analogs with 18. Yazaki PJ, Wu AM, Tsai SW, et al. Tumor targeting of radiometal
labeled anti-CEA recombinant T84.66 diabody and t84.66 mini-
affinity to more somatostatin receptor subtypes [282].
body: comparison to radioiodinated fragments. Bioconjug Chem.
2001;12:220–228.
19. Buchmann I, Bunjes D, Rattat D. Myeloablative Radioimmunotherapy
References with 188Re Labeled Anti-CD66-Mab Prior to Stem Cell Transplantation
of leukemia patients. J Nucl Med. 2001;42.
1. Kaminski MS TM, Estes J, Kolstad A, Ross CW, Zasadny K, 20. Adelstein SJ, Kassis AI. Radiobiologic implications of the micro-
Regan D, Kison P, Fisher S, Kroll S, Wahl RL. 131I-tositumomab scopic distribution of energy from radionuclides. Int J Rad Appl
therapy as initial treatment for follicular lymphoma. N Engl J Med. Instrum B. 1987;14:165–169.
2005;352:496–498. 21. Howell RW, Rao DV, Sastry KS. Macroscopic dosimetry for radio-
2. Press OW UJ, Braziel RM, Maloney DG, Miller TP, Leblanc M, immunotherapy: nonuniform activity distributions in solid tumors.
Fisher RI, Group. SO. Phase II trial of CHOP chemotherapy fol- Med Phys. 1989;16:66–74.
lowed by tositumomab/iodine I-131 tositumomab for previously 22. Humm JL. Dosimetric aspects of radiolabeled antibodies for tumor
untreated follicular non-Hodgkin’s lymphoma: five-year follow-up therapy. J Nucl Med. 1986;27:1490–1497.
of Southwest Oncology Group Protocol S9911. J Clin Oncol. 23. Wheldon TE, O’Donoghue JA, Barrett A, Michalowski AS. The
2006;24(25):4143–4149. curability of tumours of differing size by targeted radiotherapy
3. Leonard JP, Coleman M, Kostakoglu L, et al. Abbreviated chemo- using 131I or 90Y. Radiother Oncol. 1991;21:91–99.
therapy with fludarabine followed by tositumomab and iodine I 24. Macklis RM, Lin JY, Beresford B, Atcher RW, Hines JJ, Humm
131 tositumomab for untreated follicular lymphoma. J Clin Oncol. JL. Cellular kinetics, dosimetry, and radiobiology of alpha-particle
2005;23:5696–5704. radioimmunotherapy: induction of apoptosis. Radiat Res.
4. Zinzani PL, Tani M, Fanti S, et al. A phase II trial of CHOP che- 1992;130:220–226.
motherapy followed by yttrium 90 ibritumomab tiuxetan (Zevalin) 25. Jain RK. Determinants of tumor blood flow: a review. Cancer Res.
for previously untreated elderly diffuse large B-cell lymphoma 1988;48:2641–2658.
patients. Ann Oncol. 2008;19:769–773. 26. Hassfjeld S BM. The development of the a-particle emitting radio-
5. Wessels BW, Rogus RD. Radionuclide selection and model nuclides 212Bi and 213Bi and their decay chain related radionuclides
absorbed dose calculations for radiolabeled tumor associated anti- for therapeutic applications. Chem Rev. 2001;101:2019–2036.
bodies. Med Phys. 1984;11:638–645. 27. Beierwaltes W. Radioimmunotherapy of cancer: Historical per-
6. Zalutsky M. Radiohalogenation of antibodies: Chemical aspects. spectives and prospects for the future. NATO Conference on
In: Srivastava S, ed. Radiolabeled Monoclonal Antibodies for Radiolabeled Monoclonal Antibody and External Scintigraphy:
Imaging and Therapy. New York: Plenum Press; 1988. Science. 1979.
7. Fowler JF. Radiobiological aspects of low dose rates in radioim- 28. Order S, Klein J, Leichner P, et al. Advances in Iodine-131
munotherapy. Int J Radiat Oncol Biol Phys. 1990;18:1261–1269. Labeled Antiferritin Immunoglobulin Cancer Therapy. Cancer
8. Dillehay LE, Williams JR. Radiobiology of dose-rate patterns Biol. 1982;34.
achievable in radioimmunoglobulin therapy. Front Radiat Ther 29. Order S, Klein J, Leichner P. Radiation Therapy of Hepatoma with
Oncol. 1990;24:96–103; discussion 121–122. I-313 and Y-90 Labeled Antiferritin Antibodies. International
9. Wessels BW. Current status of animal radioimmunotherapy. Conference on Monoclonal Antibody Immunoconjugates for
Cancer Res. 1990;50:970s–973s. Cancer; 1986:27–28.
10. Hernandez MC, Knox SJ. Radiobiology of radioimmunotherapy: 30. Order S, Vriesendorp H, Klein J, et al. A phase I study of 90-yttrium
targeting CD20 B-cell antigen in non-Hodgkin’s lymphoma. Int J antiferritin: Dose escalation and tumor dose. Antibody Immunoconj
Radiat Oncol Biol Phys. 2004;59:1274–1287. Radiopharm. 1988;1:6.
11. Breitz HB, Fisher DR, Weiden PL, et al. Dosimetry of rhenium- 31. Vriesendorp HM, Quadri SM. Radiolabeled immunoglobulin ther-
186-labeled monoclonal antibodies: methods, prediction from apy in patients with Hodgkin’s disease. Cancer Biother Radiopharm.
technetium-99 m-labeled antibodies and results of phase I trials. J 2000;15:431–445.
Nucl Med. 1993;34:908–917. 32. Bierman PJ, Vose JM, Leichner PK, et al. Yttrium 90-labeled anti-
12. Fritzberg A, Beaumier P, Bottino B, Reno J. Approaches to ferritin followed by high-dose chemotherapy and autologous bone
Improved Antibody-and Peptide-mediated Targeting for Imaging marrow transplantation for poor-prognosis Hodgkin’s disease.
and Therapy of Cancer. J Cont Release. 1994;28. J Clin Oncol. 1993;11:698–703.
13. Buchsbaum DJ. Experimental radioimmunotherapy. Semin Radiat 33. Epstein AL, Marder RJ, Winter JN, et al. Two new monoclonal
Oncol. 2000;10:156–167. antibodies, Lym-1 and Lym-2, reactive with human B-lymphocytes
14.Goshorn S, Sanderson J, Axworthy D, Lin Y, Hylarides M, Schultz J. and derived tumors, with immunodiagnostic and immunothera-
Preclinical evaluation of a humanized NR-LU-10 antibody- peutic potential. Cancer Res. 1987;47:830–840.
streptavidin fusion protein for pretargeted cancer therapy. Cancer 34. DeNardo GL, DeNardo SJ, Lamborn KR, et al. Low-dose, frac-
Biother Radiopharm. 2001;16:109–123. tionated radioimmunotherapy for B-cell malignancies using 131I-
15. Huse WD, Sastry L, Iverson SA, et al. Generation of a large com- Lym-1 antibody. Cancer Biother Radiopharm. 1998;13:239–254.
binatorial library of the immunoglobulin repertoire in phage 35. DeNardo GL, DeNardo SJ, Goldstein DS, et al. Maximum-
lambda. Science. 1989;246:1275–1281. tolerated dose, toxicity, and efficacy of (131)I-Lym-1 antibody for
490 Targeted radionuclide therapy of cancer

fractionated radioimmunotherapy of non-Hodgkin’s lymphoma. 53. Gopal AK, Rajendran JG, Petersdorf SH, et al. High dose chemo-
J Clin Oncol. 1998;16:3246–3256. radioimmunotherapy with autologous stem cell support for
36. DeNardo GL, Kukis DL, Shen S, DeNardo DA, Meares CF, relapsed mantle cell lymphoma. Blood. 2002, 99(9): 3158–62.
DeNardo SJ. 67Cu-versus 131I-labeled Lym-1 antibody: comparative 54. Dinndorf PA, Andrews RG, Benjamin D, Ridgway D, Wolff L,
pharmacokinetics and dosimetry in patients with non-Hodgkin’s Bernstein ID. Expression of normal myeloid-associated antigens
lymphoma. Clin Cancer Res. 1999;5:533–541. by acute leukemia cells. Blood. 1986;67:1048–1053.
37. O’Donnell RT, DeNardo GL, Kukis DL, et al. 67Copper-2- 55. Nakano A, Harada T, Morikawa S, Kato Y. Expression of leuko-
iminothiolane-6-[p-(bromoacetamido)benzyl-TETA-Lym-1 for cyte common antigen (CD45) on various human leukemia/lym-
radioimmunotherapy of non-Hodgkin’s lymphoma. Clin Cancer phoma cell lines. Acta Pathol Jpn. 1990;40:107–115.
Res. 1999;5:3330s–3336s. 56. Taetle R, Ostergaard H, Smedsrud M, Trowbridge I. Regulation
38. Kaminski MS, Zasadny KR, Francis IR, et al. Radioimmunotherapy of CD45 expression in human leukemia cells. Leukemia.
of B-cell lymphoma with [131I]anti-B1 (anti-CD20) antibody. N 1991;5:309–314.
Engl J Med. 1993;329:459–465. 57. Noworolska A, Harlozinska A, Richter R, Brodzka W. Non-
39. Kaminski MS ZK, Francis IR, Fenner MC, Ross CW, Milik AW, specific cross-reacting antigen (NCA) in individual maturation
Estes J, Tuck M, Regan D, Fisher S, Glenn SD, Wahl RL. Iodine- stages of myelocytic cell series. Br J Cancer. 1985;51:371–377.
131-anti-B1 radioimmunotherapy for B-cell lymphoma. J Clin 58. Appelbaum F, Matthews D, Eary JF, et al. The use of radiolabeled
Oncol. 1996;14:1974–1981. anti-CD33 antibody to augment marrow irradiation prior to marrow
40. Kaminski MS, Zelenetz AD, Press OW, et al. Pivotal study of transplantation for acute myelogenous leukemia. Transplantation.
iodine I 131 tositumomab for chemotherapy-refractory low-grade 1992;54:829–833.
or transformed low-grade B-cell non-Hodgkin’s lymphomas. J Clin 59. Schwartz MA, Lovett DR, Redner A, et al. Dose-escalation trial of
Oncol. 2001;19:3918–3928. M195 labeled with iodine 131 for cytoreduction and marrow abla-
41. Kaminski MS, Estes J, Zasadny KR, et al. Radioimmunotherapy tion in relapsed or refractory myeloid leukemias. J Clin Oncol.
with iodine (131)I tositumomab for relapsed or refractory B-cell non- 1993;11:294–303.
Hodgkin lymphoma: updated results and long-term follow-up of the 60. Jurcic JG, McDevitt MR, Sgouros G, et al. Phase I trial of targeted
University of Michigan experience. Blood. 2000;96:1259–1266. alpha-particle therapy for myeloid leukemias with Bismuth-213-
42. Koastakolu L, Ali A, Coleman R, et al. Efficacy and safety of bexxar HuM195. Proc Am Soc Clin Oncol. 1999;18:7a.
in a large multicenter expanded access study. J Nucl Med. 2001;42 61. Matthews DC, Appelbaum FR, Eary JF, et al. Phase I study of
(abst). (131)I-anti-CD45 antibody plus cyclophosphamide and total body
43. Kaminski MS, Tuck M, Estes J, et al. 131I-tositumomab therapy as irradiation for advanced acute leukemia and myelodysplastic syn-
initial treatment for follicular lymphoma. N Engl J Med. drome. Blood. 1999;94:1237–1247.
2005;352:441–449. 62. Pagel J, Gooley T, Rajendran J, et al. Targeted radiotherapy using
44. Kaminski M, Hamstra D, Estes J, Wahl R. The impact of FLIPI on 131I-anti-CD45 antibody followed by allogeneic hematopoietic
outcome of frontline treatment with single-agent I-131 tositumomab cell transplantation (HCT): the relationships among dosimetry,
for follicular lymphoma (FL). J Clin Oncol. 2006;24 (abst). bone marrow uptake, and relapse. Eur J Nucl Med Mol Imaging.
45. Witzig TE. The use of ibritumomab tiuxetan radioimmunotherapy 2006;33:S193.
for patients with relapsed B-cell non-Hodgkin’s lymphoma. Semin 63. Pagel JM, Appelbaum FR, Eary JF, et al. 131I-anti-CD45 antibody
Oncol. 2000;27:74–78. plus busulfan and cyclophosphamide before allogeneic hematopoi-
46. Witzig TE, Gordon LI, Cabanillas F, et al. Randomized controlled etic cell transplantation for treatment of acute myeloid leukemia in
trial of yttrium-90-labeled ibritumomab tiuxetan radioimmuno- first remission. Blood. 2006;107:2184–2191.
therapy versus rituximab immunotherapy for patients with relapsed 64. Blaise D, Maraninchi D, Michallet M, et al. Long-term follow-up of a
or refractory low-grade, follicular, or transformed B-cell non- randomized trial comparing the combination of cyclophosphamide
Hodgkin’s lymphoma. J Clin Oncol. 2002;20:2453–2463. with total body irradiation or busulfan as conditioning regimen for
47. Witzig TE. Radioimmunotherapy for patients with relapsed B-cell patients receiving HLA-identical marrow grafts for acute myeloblas-
non-Hodgkin lymphoma. Cancer Chemother Pharmacol. 2001;48 tic leukemia in first complete remission. Blood. 2001;97:3669–3671.
Suppl 1:S91–95. 65. Pagel J, Appelbaum F, Sandmaier B, et al. 131I-anti-CD45 anti-
48. Press OW, Eary JF, Appelbaum FR, et al. Radiolabeled-antibody body plus fludarabine, low-dose total body irradiation and periph-
therapy of B-cell lymphoma with autologous bone marrow support eral blood stem cell infusion for elderly patients with advanced
[see comments]. N Engl J Med. 1993;329:1219–1224. acute myeloid leukemia (AML) or high-risk myelodysplastic
49. Johnson TA, Press OW. Therapy of B-cell lymphomas with mono- syndrome (MDS). Blood. 2005;106.
clonal antibodies and radioimmunoconjugates: the Seattle experi- 66. Bunjes D, Buchmann I, Duncker C, et al. Rhenium 188-labeled
ence. Ann Hematol. 2000;79:175–182. anti-CD66 (a, b, c, e) monoclonal antibody to intensify the condi-
50. Liu SY, Eary JF, Petersdorf SH, et al. Follow-up of relapsed B-cell lym- tioning regimen prior to stem cell transplantation for patients with
phoma patients treated with iodine-131- labeled anti-CD20 antibody high-risk acute myeloid leukemia or myelodysplastic syndrome:
and autologous stem-cell rescue. J Clin Oncol. 1998;16:3270–3278. results of a phase I-II study. Blood. 2001;98:565–572.
51. Gopal AK, Gooley TA, Maloney DG, et al. High-dose radioim- 67. Ringhoffer M, Blumstein N, Neumaier B, et al. 188Re or
munotherapy versus conventional high-dose therapy and autolo- 90Y-labelled anti-CD66 antibody as part of a dose-reduced con-
gous hematopoietic stem cell transplantation for relapsed follicular ditioning regimen for patients with acute leukaemia or myelodys-
non-Hodgkin lymphoma: a multivariable cohort analysis. Blood. plastic syndrome over the age of 55: results of a phase I-II study.
2003;102:2351–2357. Br J Haematol. 2005;130:604–613 (abst).
52. Press OW, Eary JF, Gooley T, et al. A phase I/II trial of iodine-131- 68. McDevitt MR, Ma D, Lai LT, et al. Tumor therapy with targeted
tositumomab (anti-CD20), etoposide, cyclophosphamide, and atomic nanogenerators. Science. 2001;294:1537–1540 (abst).
autologous stem cell transplantation for relapsed B-cell lymphomas. 69. Rosenblat T, McDevitt M, Pandit-Taskar N, et al. Phase I trial of
Blood. 2000;96:2934–2942. the targeted alpha-particle nano-generator actinium-225 (225Ac)-
John M. Pagel et al. 491

HuM195 (Anti-CD33) in acute myeloid leukemia. Blood. 87. Yu B, Carrasquillo J, Milenic D, et al. Phase I trial of iodine
2007;110 (abst). 131-labeled COL-1 in patients with gastrointestinal malignan-
70. Mulford DA, Scheinberg DA, Jurcic JG. The promise of targeted cies: influence of serum carcinoembryonic antigen and tumor
{alpha}-particle therapy. J Nucl Med. 2005;46 Suppl 1:199S-204S. bulk on pharmacokinetics. J Clin Oncol. 1996;14:1798–1809.
71. Zhang M, Yao Z, Zhang Z, et al. The anti-CD25 monoclonal anti- 88. Yorke ED, Beaumier PL, Wessels BW, Fritzberg AR, Morgan
body 7G7/B6, armed with the {alpha}-Emitter 211At, provides AC, Jr. Optimal antibody-radionuclide combinations for clinical
effective radioimmunotherapy for a murine model of leukemia. radioimmunotherapy: a predictive model based on mouse phar-
Cancer Res. 2006;66:8227–8232. macokinetics. Int J Rad Appl Instrum B. 1991;18:827–835.
72. Zhang M, Yao Z, Patel H, et al. Effective therapy of murine models 89. Juweid M, Sharkey RM, Behr T, et al. Targeting and initial radio-
of human leukemia and lymphoma with radiolabeled anti-CD30 immunotherapy of medullary thyroid carcinoma with 131I-labeled
antibody, HeFi-1. Proc Natl Acad Sci U S A. 2007;104:8444–8448. monoclonal antibodies to carcinoembryonic antigen. Cancer Res.
73. Behr TM, Sharkey RM, Juweid ME, et al. Phase I/II clinical 1995;55:5946s–5951s.
radioimmunotherapy with an iodine-131-labeled anti-carcinoem- 90. Juweid ME, Hajjar G, Stein R, et al. Initial experience with high-
bryonic antigen murine monoclonal antibody IgG. J Nucl Med. dose radioimmunotherapy of metastatic medullary thyroid cancer
1997;38:858–870. using 131I-MN-14 F(ab)2 anti-carcinoembryonic antigen MAb
74. Breitz HB, Weiden PL, Vanderheyden JL, et al. Clinical experience and AHSCR. J Nucl Med. 2000;41:93–103.
with rhenium-186-labeled monoclonal antibodies for radioimmuno- 91. Sodee DB, Malguria N, Faulhaber P, Resnick MI, Albert J,
therapy: results of phase I trials. J Nucl Med. 1992;33:1099–1109. Bakale G. Multicenter ProstaScint imaging findings in 2154
75. Juweid M, Sharkey R, Swayne L, et al. Phase I dose-escalation patients with prostate cancer. The ProstaScint Imaging Centers.
trial of I-131-labeled MN-14 anticarcinoembryonic antigen Urology. 2000;56:988–993.
(CEA) monoclonal antibody in patient with epithelial ovarian 92. Abdel-Nabi H, Spaulding M, Farrell E, Derby I, Lamonica D.
cancer. Tumor Target. 1996;2 (abst). Treatment of Refractory Prostate Carcinoma with Y-90 KC4.
76. Lane DM, Eagle KF, Begent RH, et al. Radioimmunotherapy of J Nucl Med. 1995;36.
metastatic colorectal tumours with iodine-131-labelled antibody 93. Meredith RF, Bueschen AJ, Khazaeli MB, et al. Treatment of
to carcinoembryonic antigen: phase I/II study with comparative metastatic prostate carcinoma with radiolabeled antibody CC49.
biodistribution of intact and F(ab’)2 antibodies. Br J Cancer. J Nucl Med. 1994;35:1017–1022.
1994;70:521–525. 94. Meredith R, Khazaeli M, Carabasi M, LoBuglio A.
77. Ledermann JA, Begent RH, Massof C, Kelly AM, Adam T, Radioimmunotherapy of Prostate Cancer. In: Riva P, ed. Therapy
Bagshawe KD. A phase-I study of repeated therapy with radiola- of Malignancies with Radioconjugate Monoclonal Antibodies:
belled antibody to carcinoembryonic antigen using intermittent Present Possibilities and Future Perspectives. Philadelphia, PA:
or continuous administration of cyclosporin A to suppress the Harwood Academic; 1999.
immune response. Int J Cancer. 1991;47:659–664. 95. DeNardo S. Radioimmunotherapy for Breast Cancer: Systemic
78. Meredith RF, Khazaeli MB, Plott WE, et al. Phase I trial of Tumor Target Irradiation. Adv Oncol. 2000;15:5.
iodine-131-chimeric B72.3 (human IgG4) in metastatic colorectal 96. Goodman GE, Hellstrom I, Yelton DE, et al. Phase I trial of chi-
cancer. J Nucl Med. 1992;33:23–29. meric (human-mouse) monoclonal antibody L6 in patients with
79. Murray JL, Macey DJ, Grant EJ, et al. Enhanced TAG-72 expres- non-small-cell lung, colon, and breast cancer. Cancer Immunol
sion and tumor uptake of radiolabeled monoclonal antibody CC49 Immunother. 1993;36:267–273.
in metastatic breast cancer patients following alpha-interferon 97. DeNardo SJ, Mirick GR, Kroger LA, et al. The biologic window for
treatment. Cancer Res. 1995;55:5925s–5928s. chimeric L6 radioimmunotherapy. Cancer. 1994;73:1023–1032.
80. Tempero M, Leichner P, Dalrymple G, et al. High-dose therapy 98. DeNardo SJ, O’Grady LF, Richman CM, et al. Radioimmunotherapy
with iodine-131-labeled monoclonal antibody CC49 in patients for advanced breast cancer using I-131-ChL6 antibody. Anticancer
with gastrointestinal cancers: a phase I trial. J Clin Oncol. Res. 1997;17:1745–1751.
1997;15:1518–1528. 99. Richman CM, DeNardo SJ, O’Donnell RT, et al. Dosimetry-
81. Wong JYC, Chu DZ, Yamauchi DM, et al. A phase I radioimmuno- based therapy in metastatic breast cancer patients using 90Y
therapy trial evaluating 90yttrium-labeled anti-carcinoembryonic monoclonal antibody 170H.82 with autologous stem cell support
antigen (CEA) chimeric T84.66 in patients with metastatic CEA- and cyclosporin A. Clin Cancer Res. 1999;5:3243s-3248s.
producing malignancies. Clin Cancer Res. 2000;6:3855–3863. 100. DeNardo SJ, Richman CM, Goldstein DS, et al. Yttrium-90/
82. Meredith RF, Khazaeli MB, Liu T, et al. Dose fractionation of indium-111-DOTA-peptide-chimeric L6: pharmacokinetics,
radiolabeled antibodies in patients with metastatic colon cancer. dosimetry and initial results in patients with incurable breast can-
J Nucl Med. 1992;33:1648–1653. cer. Anticancer Res. 1997;17:1735–1744.
83. Meredith RF, Khazaeli MB, Plott WE, et al. Initial clinical evalu- 101. DeNardo SJ, Kramer EL, O’Donnell RT, et al. Radioimmunotherapy
ation of iodine-125-labeled chimeric 17–1A for metastatic colon for breast cancer using indium-111/yttrium-90 BrE-3: results of a
cancer. J Nucl Med. 1995;36:2229–2233. phase I clinical trial. J Nucl Med. 1997;38:1180–1185.
84. Rosenblum M, Macey D, Podoloff D, et al. A phase I pharma- 102. Schrier DM, Stemmer SM, Johnson T, et al. High-dose 90Y
cokinetic, toxicity and dosimetry study of 131-I labeled IMMU- Mx-diethylenetriaminepentaacetic acid (DTPA)-BrE-3 and
4F(ab’)2 in patients with advanced colorectal carcinoma. autologous hematopoietic stem cell support (AHSCS) for the
Antibody Immunoconj Radiopharm. 1993;6:239–255. treatment of advanced breast cancer: a phase I trial. Cancer Res.
85. Scott A, Divgi C, Kemeny N, et al. Radioimmunotherapy with 1995;55:5921s–5924s.
I-131 labeled monoclonal antibody CC49 in colorectal cancer. 103. Kramer EL, Liebes L, Wasserheit C, et al. Initial clinical evaluation
Eur J Nucl Med 1992;8. of radiolabeled MX-DTPA humanized BrE-3 antibody in patients
86. Welt S, Divgi CR, Kemeny N, et al. Phase I/II study of iodine with advanced breast cancer. Clin Cancer Res. 1998;4:1679–1688.
131-labeled monoclonal antibody A33 in patients with advanced 104. Mulligan T, Carrasquillo JA, Chung Y, et al. Phase I study of
colon cancer. J Clin Oncol. 1994;12:1561–1571. intravenous 177Lu-labeled CC49 murine monoclonal antibody in
492 Targeted radionuclide therapy of cancer

patients with advanced adenocarcinoma. Clin Cancer Res. of patients with leptomeningeal neoplasms or primary brain
1995;1:1447–1454. tumor resection cavities with subarachnoid communication:
105. Carabasi M, Meredith R, Khazeli M, Tilden A, Brezovich I, phase I trial results. Clin Cancer Res. 1996;2:963–972.
Schlom J. Combined modality radiation using radioimmunother- 120. Reubi J, Landolt A. High density of somatostatin receptors in
apy and external beam radiation for breast and prostate cancer. pituitary tumors from acromegalic patients. J Clin Endocrinol
Cancer Biother Radiopharm. 1999;14. Metab. 1984;59:1148–1151.
106. Murray JL, Zukiwski AA, Mujoo K, Rosenblum MG. 121. Lamberts S, Bakker W, Reubi J, Krenning E. Somatostatin-
Recombinant alpha-interferon enhances tumor targeting of an receptor imaging in the localization of endocrine tumors. N Engl
antimelanoma monoclonal antibody in vivo. J Biol Response J Med 1990;323:1246–1249.
Mod. 1990;9:556–563. 122. Krenning EP, Kwekkeboom DJ, Bakker WH, et al. Somatostatin
107. Wong JY, Somlo G, Odom-Maryon T, et al. Initial clinical experi- receptor scintigraphy with [111In-DTPA-D-Phe1]- and
ence evaluating Yttrium-90-chimeric T84.66 anticarcinoembry- [123I-Tyr3]-octreotide: the Rotterdam experience with more than
onic antigen antibody and autologous hematopoietic stem cell 1000 patients. Eur J Nucl Med. 1993;20:716–731.
support in patients with carcinoembryonic antigen-producing 123. Reubi JC. Regulatory peptide receptors as molecular targets for
metastatic breast cancer. Clin Cancer Res. 1999;5:3224s-3231s. cancer diagnosis and therapy. Q J Nucl Med. 1997;41:63–70.
108. Kalofonos HP, Pawlikowska TR, Hemingway A, et al. Antibody 124. Behr T, Gotthardt M, Barth A, Béhé M. Imaging tumors with
guided diagnosis and therapy of brain gliomas using radiolabeled peptide-based radioligands. Q J Nucl Med. 2001;45:189–200.
monoclonal antibodies against epidermal growth factor receptor and 125. Modlin IM, Tang LH. Approaches to the diagnosis of gut neu-
placental alkaline phosphatase. J Nucl Med. 1989;30:1636–1645. roendocrine tumors: the last word (today). Gastroenterology.
109. Brady L, Woo D, Markoe A, et al. Radioimmunotherapy with 1997;112:583–590.
125I-EGF-425 in patients with brain tumors: Preliminary results 126. Ricke J, Klose K. Imaging procedures in neuroendocrine tumors.
of a Phase II clinical trial. Antibody Immunoconj Radiopharm. Digestion. 2000;62:39–44.
1990;3:11. 127. Ricke J, Klose K, Mignon M, Öberg K, Wiedenmann B.
110. Brady L, Miyamoto C, Woo D, et al. Malignant astrocytomas Standardization of imaing in neuroendocrine tumours: results of
treated with iodine – 125 labeled monoclonal antibody 425 a European Delphi process. Eur J Radiol. 2001;37:8–17.
against epidermal growth factor receptor: a Phase II trial. Int J 128. Termanini B, Gibril F, Reynolds JC, et al. Value of somatostatin
Rad Oncol Biol Phys. 1991;22:6. receptor scintigraphy: a prospective study in gastrinoma of its effect
111. Riva P, Franceschi G, Riva N, Casi M, Santimaria M, Adamo M. on clinical management. Gastroenterology. 1997;112:335–347.
Role of nuclear medicine in the treatment of malignant gliomas: 129. Lebtahi R, Cadiot G, Sarda L, et al. Clinical impact of somatosta-
the locoregional radioimmunotherapy approach. Eur J Nucl Med. tin receptor scintigraphy in the management of patients with neu-
2000;27:601–609. roendocrine gastroenteropancreatic tumors. J Nucl Med.
112. Riva P, Tison G, Franchesci N, Casi M, Moscatelli G. Successful 1997;38:853–858.
treatment of metastatic gastrointestinal cancer by means of radio- 130. Gibril F, Reynolds JC, Chen CC, et al. Specificity of somatostatin
immunotherapy. J Nucl Med. 1992;33 (abst). receptor scintigraphy: a prospective study and effects of false-
113. Zalutsky MR, Moseley RP, Coakham HB, Coleman RE, Bigner positive localizations on management in patients with gastrino-
DD. Pharmacokinetics and tumor localization of 131I-labeled anti- mas. J Nucl Med. 1999;40:539–553.
tenascin monoclonal antibody 81C6 in patients with gliomas and 131. Gotthardt M, Dirkmorfeld LM, Wied MU, et al. Influence of
other intracranial malignancies. Cancer Res. 1989;49:2807–2813. somatostatin receptor scintigraphy and CT/MRI on the clini-
114. Bigner DD, Brown M, Coleman RE, et al. Phase I studies of cal management of patients with gastrointestinal neuroendo-
treatment of malignant gliomas and neoplastic meningitis with crine tumors: an analysis in 188 patients. Digestion. 2003;68:
131I-radiolabeled monoclonal antibodies anti-tenascin 81C6 and 80–85.
anti-chondroitin proteoglycan sulfate Me1-14 F (ab’)2 – a pre- 132. de Visser M, Janssen PJ, Srinivasan A, et al. Stabilized
liminary report. J Neurooncol. 1995;24:109–122. 111In-labelled DTPA- and DOTA-conjugated neurotensin ana-
115. Zalutsky MR, Reardon DA, Akabani G, et al. Clinical experience logues for imaging and therapy of exocrine pancreatic cancer.
with alpha-particle emitting 211At: treatment of recurrent brain Eur J Nucl Med Mol Imaging. 2003;30:1134–1139.
tumor patients with 211At-labeled chimeric antitenascin mono- 133. Gotthardt M, Fischer M, Naeher I, et al. Use of the incretin hor-
clonal antibody 81C6. J Nucl Med. 2008;49:30–38. mone glucagon-like peptide-1 (GLP-1) for the detection of insu-
116. Papanastassiou V, Pizer BL, Coakham HB, Bullimore J, Zananiri linomas: initial experimental results. Eur J Nucl Med Mol
T, Kemshead JT. Treatment of recurrent and cystic malignant Imaging. 2002;29:597–606.
gliomas by a single intracavity injection of 131I monoclonal anti- 134. Janssen ML, Oyen WJ, Dijkgraaf I, et al. Tumor targeting with
body: feasibility, pharmacokinetics and dosimetry. Br J Cancer. radiolabeled alpha(v)beta(3) integrin binding peptides in a nude
1993;67:144–151. mouse model. Cancer Res. 2002;62:6146–6151.
117. Hopkins K, Chandler C, Bullimore J, Sandeman D, Coakham H, 135. Reubi JC. Neuropeptide receptors in health and disease: the
Kemshead JT. A pilot study of the treatment of patients with molecular basis for in vivo imaging. J Nucl Med.
recurrent malignant gliomas with intratumoral yttrium-90 radio- 1995;36:1825–1835.
immunoconjugates. Radiother Oncol. 1995;34:121–131. 136. Behr TM, Gotthardt M, Becker W, Behe M. Radioiodination of
118. Papanastassiou V, Pizer BL, Chandler CL, Zananiri TF, Kemshead monoclonal antibodies, proteins and peptides for diagnosis and
JT, Hopkins KI. Pharmacokinetics and dose estimates following therapy. A review of standardized, reliable and safe procedures
intrathecal administration of 131I-monoclonal antibodies for the for clinical grade levels kBq to GBq in the Gottingen/Marburg
treatment of central nervous system malignancies. Int J Radiat experience. Nuklearmedizin. 2002;41:71–79.
Oncol Biol Phys. 1995;31:541–552. 137. Meares CF, Moi MK, Diril H, et al. Macrocyclic chelates of
119. Brown MT, Coleman RE, Friedman AH, et al. Intrathecal radiometals for diagnosis and therapy. Br J Cancer Suppl.
131I-labeled antitenascin monoclonal antibody 81C6 treatment 1990;10:21–26.
John M. Pagel et al. 493

138. Heppeler A, Froidevaux S, Mäcke H, et al. Radiometal-Labelled 156. Menda Y, Kahn D. Somatostatin receptor imaging of non-small
Macrocyclic Chelator-Derivatised Somatostatin Analogue with cell lung cancer with 99 mTc depreotide. Semin Nucl Med.
Superb Tumour-Targeting Properties and Potential for Receptor- 2002;32:92–96.
Mediated Internal Radiotherapy. Chem Eur J. 1999;5:1974–1981. 157. Goldsmith SJ, Kostakoglu L. Role of nuclear medicine in the
139. Breeman WA, Hofland LJ, de Jong M, et al. Evaluation of radio- evaluation of the solitary pulmonary nodule. Semin Ultrasound
labelled bombesin analogues for receptor-targeted scintigraphy CT MR. 2000;21:129–138.
and radiotherapy. Int J Cancer. 1999;81:658–665. 158. Blum J, Handmaker H, Lister-James J, Rinne N. A multicenter trial
140. Ginj M, Zhang H, Waser B, et al. Radiolabeled somatostatin with a somatostatin analog (99 m)Tc depreotide in the evaluation of
receptor antagonists are preferable to agonists for in vivo peptide solitary pulmonary nodules. Chest. 2000;117:1232–1238.
receptor targeting of tumors. Proc Natl Acad Sci U S A. 159. Grewal RK, Dadparvar S, Yu JQ, et al. Efficacy of Tc-99 m
2006;103:16436–16441. depreotide scintigraphy in the evaluation of solitary pulmonary
141. Jakubke H. Peptide – Chemie und Biologie. Heidelberg: Spektrum nodules. Cancer J. 2002;8:400–404.
Akademischer Verlag. 1996. 160. Bangard M, Behe M, Guhlke S, et al. Detection of somatostatin
142. Jamar F, Fiasse R, Leners N, Pauwels S. Somatostatin receptor receptor-positive tumours using the new 99 mTc-tricine-HYNIC-
imaging with indium-111-pentetreotide in gastroenteropancreatic D-Phe1-Tyr3-octreotide: first results in patients and comparison
neuroendocrine tumors: safety, efficacy and impact on patient with 111In-DTPA-D-Phe1-octreotide. Eur J Nucl Med.
management. J Nucl Med. 1995;36:542–549. 2000;27:628–637.
143. Kwekkeboom DJ, Krenning EP. Somatostatin receptor imaging. 161. Gabriel M, Decristoforo C, Donnemiller E, et al. An intrapatient
Semin Nucl Med. 2002;32:84–91. comparison of 99 mTc-EDDA/HYNIC-TOC with 111In-DTPA-
144. Sreedharan SP, Kodama KT, Peterson KE, Goetzl EJ. Distinct octreotide for diagnosis of somatostatin receptor-expressing
subsets of somatostatin receptors on cultured human lympho- tumors. J Nucl Med. 2003;44:708–716.
cytes. J Biol Chem. 1989;264:949–952. 162. Virgolini I, Britton K, Buscombe J, Moncayo R, Paganelli G,
145. Blum JE, Handmaker H, Rinne NA. The utility of a somatosta- Riva P. In- and Y-DOTA-lanreotide: results and implications of
tin-type receptor binding peptide radiopharmaceutical (P829) in the MAURITIUS trial. Semin Nucl Med. 2002;32:148–155.
the evaluation of solitary pulmonary nodules. Chest. 163. Reubi JC, Schar JC, Waser B, et al. Affinity profiles for human
1999;115:224–232. somatostatin receptor subtypes SST1-SST5 of somatostatin
146. Stoffel M, Jamar F, Donckier J, et al. Increased uptake of radiotracers selected for scintigraphic and radiotherapeutic use.
indium-111 pentetreotide up to 10 years after external thoracic Eur J Nucl Med. 2000;27:273–282.
irradiation: report of two cases. Eur J Nucl Med. 1996;23: 164. Behr T, Kann P, Gotthardt M, Béhé M, Arnold R. Nuclear medi-
723–726. cine diagnosis and therapy of neuroendocrine tumors of the gas-
147. Öberg K. Established clinical use of octreotide and lanreotide in trointestinal tract, including carcinoids. Der Nuklearmediziner.
oncology. Chemotherapy. 2001;47 Suppl 2:40–53. 2003;26:121–133.
148. Krenning E, Kwekkeboom D, Pauwels S, LK LK, Reubi J. 165. Behr T, Jenner M, Béhé M, et al. Radiolabeled peptides for tar-
Somatostatin Receptor Scintigraphy. Vol. 1995. New York: Raven geting of cholecystokinin-B/gastrin receptor expressing tumors:
Press; 1995. from preclinical development to initial clinical results. J Nucl
149. Reubi JC, Lang W, Maurer R, Koper JW, Lamberts SW. Med. 1999;40:1029–1044.
Distribution and biochemical characterization of somatostatin 166. Behr TM, Jenner N, Radetzky S, et al. Targeting of cholecystoki-
receptors in tumors of the human central nervous system. Cancer nin-B/gastrin receptors in vivo: preclinical and initial clinical
Res. 1987;47:5758–5764. evaluation of the diagnostic and therapeutic potential of radiola-
150. Reubi JC, Waser B, Foekens JA, Klijn JG, Lamberts SW, Laissue belled gastrin. Eur J Nucl Med. 1998;25:424–430.
J. Somatostatin receptor incidence and distribution in breast can- 167. Behr TM, Behe M, Angerstein C, et al. Cholecystokinin-B/gas-
cer using receptor autoradiography: relationship to EGF recep- trin receptor binding peptides: preclinical development and eval-
tors. Int J Cancer. 1990;46:416–420. uation of their diagnostic and therapeutic potential. Clin Cancer
151. Reubi JC, Waser B, Sheppard M, Macaulay V. Somatostatin Res. 1999;5:3124s–3138s.
receptors are present in small-cell but not in non-small-cell pri- 168. Reubi JC, Waser B. Unexpected high incidence of cholecystoki-
mary lung carcinomas: relationship to EGF-receptors. Int J nin-B/gastrin receptors in human medullary thyroid carcinomas.
Cancer. 1990;45:269–274. Int J Cancer. 1996;67:644–647.
152. Reubi J, Waser B, Hagen MV, et al. In vitro and in vivo detection 169. Reubi JC, Schaer JC, Waser B. Cholecystokinin(CCK)-A and
of somatostatin receptors in human malignant lymphomas. Int J CCK-B/gastrin receptors in human tumors. Cancer Res.
Cancer. 1992;50:895–900. 1997;57:1377–1386.
153. de Kerviler E, Cadiot G, Lebtahi R, Faraggi M, Le Guludec D, 170. Gotthardt M, Grass J, Béhé M, Schipper M, Schlieck A, Wied M.
Mignon M. Somatostatin receptor scintigraphy in forty-eight In-111-DTPA-DGlu-1-Minigastrin in the diagnosis of gastrointes-
patients with the Zollinger-Ellison syndrome. GRESZE: Groupe tinal neuroendocrine tumors: a comparison to In-111-pentetreotide.
d’Etude du Syndrome de Zollinger-Ellison. Eur J Nucl Med. J Nucl Med. 2003;44:74P.
1994;21:1191–1197. 171. Dijkgraaf I, Kruijtzer JA, Liu S, et al. Improved targeting of the
154. Gibril F, Reynolds JC, Doppman JL, et al. Somatostatin receptor alpha(v)beta (3) integrin by multimerisation of RGD peptides.
scintigraphy: its sensitivity compared with that of other imaging Eur J Nucl Med Mol Imaging. 2007;34:267–273.
methods in detecting primary and metastatic gastrinomas. A pro- 172. Virgolini I, Kurtaran A, Raderer M, et al. Vasoactive intestinal
spective study. Ann Intern Med. 1996;125:26–34. peptide receptor scintigraphy. J Nucl Med. 1995;36:1732–1739.
155. Zimmer T, Stölzel U, Bader M, et al. Endoscopic ultrasonogra- 173. Virgolini I, Raderer M, Kurtaran A, et al. Vasoactive intestinal
phy and somatostatin receptor scintigraphy in the preoperative peptide-receptor imaging for the localization of intestinal ade-
localization of insulinomas and gastrinomas. Gut. 1996;39: nocarcinomas and endocrine tumors. N Engl J Med.
562–568. 1994;331:1116–1121.
494 Targeted radionuclide therapy of cancer

174. Kurtaran A, Leimer M, Kaserer K, et al. Combined use of 193. Paganelli G, Bodei L, Chinol M, et al. Receptor-mediated radio-
111In-DTPA-D-Phe-1-octreotide (OCT) and 123I-vasoactive therapy with 90Y-DOTATOC: results of a phase I study. J Nucl
intestinal peptide (VIP) in the localization diagnosis of medullary Med. 2001;42:36P.
thyroid carcinoma (MTC). Nucl Med Biol. 1996;23:503–507. 194. Waldherr C, Pless M, Maecke H, Haldemann H, Mueller-Brandt
175. Hessenius C, Bader M, Meinhold H, et al. Vasoactive intestinal J. The clinical value of [90Y-DOTA]-d-Phe1-Tyr3-octreotide
peptide receptor scintigraphy in patients with pancreatic adeno- (90Y-DOTATOC) in the treatment of neuroendocrine tumours: a
carcinomas or neuroendocrine tumours. Eur J Nucl Med. clinical phase II study. Ann Oncol. 2001;12:941–945.
2000;27:1684–1693. 195. Waldherr C, Pless M, Maecke HR, et al. Tumor response and
176. Buchegger F, Bonvin F, Kosinski M, et al. Radiolabeled neuro- clinical benefit in neuroendocrine tumors after 7.4 GBq (90)
tensin analog, 99 mTc-NT-XI, evaluated in ductal pancreatic Y-DOTATOC. J Nucl Med. 2002;43:610–616.
adenocarcinoma patients. J Nucl Med. 2003;44:1649–1654. 196. Waldherr C, Schumacher T, Maecke H, et al. Does tumor response
177. Van de Wiele C, Dumont F, Vanden Broecke R, et al. depend on the number of treatment sessions at constant injected
Technetium-99 m RP527, a GRP analogue for visualisation of dose using 90Y-DOTATOC in neuroendocrine tumours? Eur J
GRP receptor-expressing malignancies: a feasibility study. Eur J Nucl Med. 2002;29.
Nucl Med. 2000;27:1694–1699. 197. de Jong M, Valkema R, Jamar F, et al. Somatostatin receptor-
178. Breeman WA, de Jong M, Erion JL, et al. Preclinical comparison targeted radionuclide therapy of tumors: preclinical and clinical
of (111)In-labeled DTPA- or DOTA-bombesin analogs for recep- findings. Semin Nucl Med. 2002;32:133–140.
tor-targeted scintigraphy and radionuclide therapy. J Nucl Med. 198. Smith MC, Liu J, Chen T, et al. OctreoTher: ongoing early clinical
2002;43:1650–1656. development of a somatostatin-receptor-targeted radionuclide
179. Nock B, Nikolopoulou A, Chiotellis E, et al. [99 mTc]Demobesin antineoplastic therapy. Digestion. 2000;62 Suppl 1:69–72.
1, a novel potent bombesin analogue for GRP receptor-targeted 199. Valkema R, Kvols L, Jamar F, et al. Phase I study of therapy with
tumour imaging. Eur J Nucl Med Mol Imaging. 2003;30: 90Y-SMT487 (OctreoTher) in patients with somatostatin receptor-
247–258. positive tumors. J Nucl Med. 2002;43.
180. Wild D, Behe M, Wicki A, et al. [Lys40(Ahx-DTPA-111In)NH2] 200. Jonard P, Jamar F, Walrand S, et al. Tumor dosimetry based on
exendin-4, a very promising ligand for glucagon-like peptide-1 PET86Y-DOTA-Tyr3-octreotide (SMT487) and CT-scan predicts
(GLP-1) receptor targeting. J Nucl Med. 2006;47:2025–2033. tumor response to 90Y-SMT487 (OctreoTher). J Nucl Med.
181. Wicki A, Wild D, Storch D, et al. [Lys40(Ahx-DTPA-111In) 2000;41:111P.
NH2]-Exendin-4 is a highly efficient radiotherapeutic for gluca- 201. Behr TM, Behe M, Kluge G, et al. Nephrotoxicity versus anti-
gon-like peptide-1 receptor-targeted therapy for insulinoma. Clin tumour efficacy in radiopeptide therapy: facts and myths about
Cancer Res. 2007;13:3696–3705. the Scylla and Charybdis. Eur J Nucl Med Mol Imaging.
182. Ruoslahti E, Pierschbacher MD. New perspectives in cell adhe- 2002;29:277–279.
sion: RGD and integrins. Science. 1987;238:491–497. 202. Boerman OC, Oyen WJ, Corstens FH. Between the Scylla and
183. Plow EF, Haas TA, Zhang L, Loftus J, Smith JW. Ligand binding Charybdis of peptide radionuclide therapy: hitting the tumor and
to integrins. J Biol Chem. 2000;275:21785–21788. saving the kidney. Eur J Nucl Med. 2001;28:1447–1449.
184. Brooks PC, Montgomery AM, Rosenfeld M, et al. Integrin alpha 203. Rolleman EJ, Valkema R, de Jong M, Kooij PP, Krenning EP.
v beta 3 antagonists promote tumor regression by inducing apop- Safe and effective inhibition of renal uptake of radiolabelled oct-
tosis of angiogenic blood vessels. Cell. 1994;79:1157–1164. reotide by a combination of lysine and arginine. Eur J Nucl Med
185. Haubner R, Wester HJ, Burkhart F, et al. Glycosylated RGD- Mol Imaging. 2003;30:9–15.
containing peptides: tracer for tumor targeting and angiogenesis 204. Béhé M, Kluge G, Angerstein C, Becker W, Behr T. Kidney pro-
imaging with improved biokinetics. J Nucl Med. 2001;42:326–336. tection agents during radiopeptide therapy in mice. Eur J Nucl
186. van Hagen PM, Breeman WA, Bernard HF, et al. Evaluation of a Med. 2002;29:S301.
radiolabelled cyclic DTPA-RGD analogue for tumour imaging 205. Rougier P, Mitry E. Chemotherapy in the treatment of neuroen-
and radionuclide therapy. Int J Cancer. 2000;90:186–198. docrine malignant tumors. Digestion. 2000;62 Suppl 1:73–78.
187. Haubner R, Wester HJ, Reuning U, et al. Radiolabeled alpha(v) 206. Valkema R, De Jong M, Bakker WH, et al. Phase I study of
beta3 integrin antagonists: a new class of tracers for tumor target- peptide receptor radionuclide therapy with [In-DTPA]oct-
ing. J Nucl Med. 1999;40:1061–1071. reotide: the Rotterdam experience. Semin Nucl Med.
188. Haubner R, Wester HJ, Weber WA, et al. Noninvasive imaging of 2002;32:110–122.
alpha(v)beta3 integrin expression using 18F-labeled RGD- 207. McCarthy KE, Woltering EA, Anthony LB. In situ radiotherapy
containing glycopeptide and positron emission tomography. with 111In-pentetreotide. State of the art and perspectives. Q J
Cancer Res. 2001;61:1781–1785. Nucl Med. 2000;44:88–95.
189. Imam SK. Advancements in cancer therapy with alpha-emitters: 208. Anthony LB, Woltering EA, Espenan GD, Cronin MD, Maloney
a review. Int J Radiat Oncol Biol Phys. 2001;51:271–278. TJ, McCarthy KE. Indium-111-pentetreotide prolongs survival in
190. de Jong M, Kwekkeboom D, Valkema R, Krenning EP. gastroenteropancreatic malignancies. Semin Nucl Med.
Radiolabelled peptides for tumour therapy: current status and 2002;32:123–132.
future directions. Plenary lecture at the EANM 2002. Eur J Nucl 209. Schipper M, Gotthardt M, Béhé M, Arnold R, Behr T. Therapeutic
Med Mol Imaging. 2003;30:463–469. efficacy and toxicity of In-111-DTPA-D-Phe-octreotide in patients
191. Chinol M, Bodei L, Cremonesi M, Paganelli G. Receptor- with advanced neuroendocrine tumors. J Nucl Med. 2003;44:137P.
mediated radiotherapy with Y-DOTA-DPhe-Tyr-octreotide: the 210. Kwekkeboom DJ, Bakker WH, Kam BL, et al. Treatment of
experience of the European Institute of Oncology Group. Semin patients with gastro-entero-pancreatic (GEP) tumours with the
Nucl Med. 2002;32:141–147. novel radiolabelled somatostatin analogue [177Lu-DOTA(0),Tyr3]
192. Paganelli G, Zoboli S, Cremonesi M, et al. Receptor-mediated octreotate. Eur J Nucl Med Mol Imaging. 2003;30:417–422.
radiotherapy with 90Y-DOTA-D-Phe1-Tyr3-octreotide. Eur J 211. Blumenthal RD, Sharkey RM, Forman D, Wong G, Goldenberg
Nucl Med. 2001;28:426–434. DM. Cytokine intervention permits dose escalation of radioanti-
John M. Pagel et al. 495

body. An analysis of myelostimulation by bolus versus continu- MOv18: clinical results in patients with minimal residual disease
ous infusion of IL-1/GM-CSF. Cancer. 1994;73:1083–1092. of ovarian cancer. Eur J Cancer. 1995;31A:686–690.
212. DeNardo G, DeNardo S, Kukis D, Diril H, Suey C, Meares C. 230. Hird V, Maraveyas A, Snook D, et al. Adjuvant therapy of ovar-
Strategies for enhancement of radioimmunotherapy. Int J Rad ian cancer with radioactive monoclonal antibody. Br J Cancer.
Appl Instrum B. 1991;18:633–640. 1993;68:403–406.
213. Su F-M, Lyen L, Breitz H, Weiden P, Fritzberg A. 186-Rhenium 231. Kosmas C, Maraveyas A, Gooden CS, Snook D, Epenetos AA.
MAG2GABA antibody radiolabeling for high dose radioimmu- Anti-chelate antibodies after intraperitoneal yttrium-90-labeled
notherapy studies. In: Nicolini M, Bandoli G, Mazzi U eds. monoclonal antibody immunoconjugates for ovarian cancer ther-
Fourth International Symposium on Technetium in Chemistry apy. J Nucl Med. 1995;36:746–753.
and Nuclear Medicine. Vol. Technetium and Rhenium in 232. Sands H, Jones PL, Shah SA, Palme D, Vessella RL, Gallagher
Chemistry and Nuclear Medicine. Seminario Maggiore, BM. Correlation of vascular permeability and blood flow with
Bressnanone (Italy): SG Editorial; 1995. monoclonal antibody uptake by human Clouser and renal cell
214. Surwit EA, Childers JM, Krag DN, et al. Clinical assessment of xenografts. Cancer Res. 1988;48:188–193 (abst).
111In-CYT-103 immunoscintigraphy in ovarian cancer. Gynecol 233. Stickney D, Gridley D, Kirk G, Slater J. Enhancement of mono-
Oncol. 1993;48:285–292. clonal antibody binding to melanoma with single dose radiation
215. Dillman R. Human antimouse and antiglobulin responses to mono- or hyperthermia. Nat Cancer Inst Monger. 1985:6 (abst).
clonal antibodies. Antibody Immunoconj Radiopharm. 1990;3:15. 234. Kalofonos H, Rowlinson G, Epenetos AA. Enhancement of
216. Klein J. Effects of anti-antibodies on radiolabeled antibody therapy. monoclonal antibody uptake in human colon tumor xenografts
Antibody Immunoconj Radiopharm. 1988;1:10. following irradiation. Cancer Res. 1990;50:159–163.
217. LoBuglio AF, Wheeler RH, Trang J, et al. Mouse/human chimeric 235. Warhoe K, DeNardo S, Wolkov H, et al. Evidence for external beam
monoclonal antibody in man: kinetics and immune response. irradiation enhancement of radiolabeled monoclonal antibody uptake
Proc Natl Acad Sci U S A. 1989;86:4220–4224. in breast cancer. Antibody Immunoconj Radiopharm. 1992;5:9.
218. Weiden PL, Wolf SB, Breitz HB, et al. Human anti-mouse antibody 236. Buchegger F, Allal AS, Roth A, et al. Combined radioimmuno-
suppression with cyclosporin A. Cancer. 1994;73:1093–1097. therapy and radiotherapy of liver metastases from colorectal
219. Breitz H, Seiler C, Weiden P, al. e. Re-186 16.88 l gM and cancer: a feasibility study. Anticancer Res. 2000;20:1889–1896.
88BV69 l gM human antibody studies to assess potential for 237. Epenetos A. Combined radiolabeled antibodies and radiotherapy for
radioimmunotherapy. j Nucl Med. 1994;34. the treatment of head and neck cancer. J Immunother. 1994;16:1.
220. Colcher D, Esteban J, Carrasquillo JA, et al. Complementation of 238. DeNardo G, DeNardo S, Lamborn K, al. e. Enhancement of
intracavitary and intravenous administration of a monoclonal tumor uptake of monoclonal antibody in nude mice with PEG
antibody (B72.3) in patients with carcinoma. Cancer Res. IL-2. Antibody Immunoconj Radiopharm. 1991;4:11.
1987;47:4218–4224. 239. DeNardo G, Kukis D, DeNardo S, et al. Enhancement of Cu-67–
221. Alvarez RD, Partridge EE, Khazaeli MB, et al. Intraperitoneal 2IT-BAT-LYM-1 therapy in mice with human Burkitt’s lymphoma
radioimmunotherapy of ovarian cancer with 177Lu-CC49: a (Raji) using interleukin-2(rIL-2). Tumor Target. 1996;2:1.
phase I/II study. Gynecol Oncol. 1997;65:94–101. 240. Nakamura K, Kubo A. Biodistribution of iodine-126-labeled
222. Epenetos AA, Hird V, Lambert H, Mason P, Coulter C. Long term monoclonal antibody/IL-2 immunoconjugate in athymic mice
survival of patients with advanced ovarian cancer treated with bearing human tumor xenograft. Tumor Target. 1996;2:1.
intraperitoneal radioimmunotherapy. Int J Gynecol Cancer. 241. Epstein AL, Khawli LA, Hornick JL, Taylor CR. Identification of
2000;10:44–46. a monoclonal antibody, TV-1, directed against the basement
223. Hird V, Stewart JS, Snook D, et al. Intraperitoneally administered membrane of tumor vessels, and its use to enhance the delivery of
90Y-labelled monoclonal antibodies as a third line of treatment in macromolecules to tumors after conjugation with interleukin 2.
ovarian cancer. A phase 1-2 trial: problems encountered and pos- Cancer Res. 1995;55:2673–2680.
sible solutions. Br J Cancer Suppl. 1990;10:48–51. 242. DeNardo SJ, Burke PA, Leigh BR, et al. Neovascular targeting with
224. Jacobs AJ, Fer M, Su FM, et al. A phase I trial of a rhenium cyclic RGD peptide (cRGDf-ACHA) to enhance delivery of radio-
186-labeled monoclonal antibody administered intraperitoneally immunotherapy. Cancer Biother Radiopharm. 2000;15:71–79.
in ovarian carcinoma: toxicity and clinical response. Obstet 243. Greiner JW, Guadagni F, Goldstein D, et al. Evidence for the
Gynecol. 1993;82:586–593. elevation of serum carcinoembryonic antigen and tumor-associated
225. Salk D, Lesley T, Wiseman G, al. e. A phase I trial of a fraction- glycoprotein-72 levels in patients administered interferons.
ated dose intraperitoneal administration of Rhenium-186 mono- Cancer Res. 1991;51:4155–4163.
clonal antibodies in ovarian cancer. Antibody Immunoconj 244. Greiner JW, Ullmann CD, Nieroda C, et al. Improved radioimmu-
Radiopharm. 1992;4 (abst). notherapeutic efficacy of an anticarcinoma monoclonal antibody
226. Stewart JS, Hird V, Snook D, et al. Intraperitoneal radioimmuno- (131I-CC49) when given in combination with gamma-interferon.
therapy for ovarian cancer: pharmacokinetics, toxicity, and Cancer Res. 1993;53:600–608.
efficacy of I-131 labeled monoclonal antibodies. Int J Radiat 245. Rotman M, Kuruvilla AM, Choi K, et al. Response of colo-rectal
Oncol Biol Phys. 1989;16:405–413. hepatic metastases to concomitant radiotherapy and intravenous
227. Mahe MA, Fumoleau P, Fabbro M, et al. A phase II study of infusion 5 fluorouracil. Int J Radiat Oncol Biol Phys.
intraperitoneal radioimmunotherapy with iodine-131-labeled 1986;12:2179–2187.
monoclonal antibody OC-125 in patients with residual ovarian 246. Remmenga SW, Colcher D, Gansow O, Pippen CG, Raubitschek A.
carcinoma. Clin Cancer Res. 1999;5:3249s–3253s. Continuous infusion chemotherapy as a radiation-enhancing
228. Kavanagh J, Rosenblum M, Kudelka A. Pharmacokinetics, side agent for yttrium-90-radiolabeled monoclonal antibody therapy
effects and tissue distribution of intraperitoneal B72.3-GYK- of a human tumor xenograft. Gynecol Oncol. 1994;55:115–122.
DTPA 90Y with and without EDTA in ovarian cancer. Can Invest. 247. Tschmelitsch J, Barendswaard E, Williams C, Jr., et al. Enhanced
1992;10:4. antitumor activity of combination radioimmunotherapy
229. Crippa F, Bolis G, Seregni E, et al. Single-dose intraperitoneal (131I-labeled monoclonal antibody A33) with chemotherapy
radioimmunotherapy with the murine monoclonal antibody I-131 (fluorouracil). Cancer Res. 1997;57:2181–2186.
496 Targeted radionuclide therapy of cancer

248. DeNardo SJ, Kukis DL, Kroger LA, et al. Synergy of Taxol and 265. Barbet J, Kraeber-Bodere F, Vuillez JP, Gautherot E, Rouvier E,
radioimmunotherapy with yttrium-90-labeled chimeric L6 anti- Chatal JF. Pretargeting with the affinity enhancement system
body: efficacy and toxicity in breast cancer xenografts. Proc Natl for radioimmunotherapy. Cancer Biother Radiopharm.
Acad Sci U S A. 1997;94:4000–4004. 1999;14:153–166.
249. Ng B, Liebes L, Kramer E, et al. Synergistic activity of radioim- 266. Le Doussal JM, Martin M, Gautherot E, Delaage M, Barbet J.
munotherapy with Y-90 MX-DTPA HuBre-3 and prolinged In vitro and in vivo targeting of radiolabeled monovalent and
Topotecan infusion in human breast cancer xenografts. J Nucl divalent haptens with dual specificity monoclonal antibody
Med. 1997;38. conjugates: enhanced divalent hapten affinity for cell-bound anti-
250. Kievit E, Pinedo HM, Schluper HM, Haisma HJ, Boven E. body conjugate. J Nucl Med. 1989;30:1358–1366.
Comparison of monoclonal antibodies 17–1A and 323/A3: the 267. Sharkey RM, McBride WJ, Karacay H, et al. A universal pretar-
influence of the affinity on tumour uptake and efficacy of radio- geting system for cancer detection and therapy using bispecific
immunotherapy in human ovarian cancer xenografts. Br J Cancer. antibody. Cancer Res. 2003;63:354–363.
1996;73:457–464. 268. Paganelli G, Malcovati M, Fazio F. Monoclonal antibody pretar-
251. Okazaki S, Tempero M, Colcher D. Combinatin radioimmuno- getting techniques for tumour localization: the avidin-biotin sys-
therapy and chemotherapy with 131I B72.3 and gemcitabine. tem. International Workshop on Techniques for Amplification of
Cancer Res. 1998;39. Tumour Targetting. Nucl Med Commun. 1991;12:211–234.
252. Prasad B, Lee MS, Hendrickson FR. Irradiation of hepatic metas- 269. Kalofonos HP, Rusckowski M, Siebecker DA, et al. Imaging of
tases. Int J Radiat Oncol Biol Phys. 1977;2:129–132. tumor in patients with indium-111-labeled biotin and streptavi-
253. Clifton KH, Szybalski W, Heidelberger C, Gollin FF, Ansfield FJ, din-conjugated antibodies: preliminary communication [see
Vermund H. Incorporation of I-125 labeled iododeoxyuridine comments]. J Nucl Med. 1990;31:1791–1796.
into the deoxyribonucleic acid of murine and human tissues fol- 270. Paganelli G, Belloni C, Magnani P, et al. Two-step tumour target-
lowing therapeutic doses. Cancer Res. 1963;23:1715–1723. ting in ovarian cancer patients using biotinylated monoclonal
254. Santos O, Pant KD, Blank EW, Ceriani RL. 5-Iododeoxyuridine antibodies and radioactive streptavidin. Eur J Nucl Med.
increases the efficacy of the radioimmunotherapy of human 1992;19:322–329.
tumors growing in nude mice. J Nucl Med. 1992;33:1530–1534. 271. Stickney D, Slater J, Kirk G, al. e. Bifunctional antibydy: ZCE/
255. Buchsbaum DJ, Khazaeli MB, Davis MA, Lawrence TS. CHA 111-Indium BLEDTA-IV clinical imaging in colorectal
Sensitization of radiolabeled monoclonal antibody therapy using carcinoma. Antibody Immunoconj Radiopharm. 1989;2:1–13.
bromodeoxyuridine. Cancer. 1994;73:999–1005. 272. Axworthy DB, Reno JM, Hylarides MD, et al. Cure of human
256. Kinsella TJ, Mitchell JB, Russo A, et al. Continuous intravenous carcinoma xenografts by a single dose of pretargeted yttrium-90
infusions of bromodeoxyuridine as a clinical radiosensitizer. J with negligible toxicity. Proc Natl Acad Sci U S A. 2000;97:
Clin Oncol. 1984;2:1144–1150. 1802–1807.
257. Morstyn G, Miller R, Russo A, Mitchell J. 131-iodine conjugated 273. Breitz HB, Weiden PL, Beaumier PL, et al. Clinical optimization
antibody cell kill enhanced by bromodeoxyuridine. Int J Radiat of pretargeted radioimmunotherapy with antibody-streptavidin
Oncol Biol Phys. 1984;10:1437–1440. conjugate and 90Y-DOTA-biotin. J Nucl Med. 2000;41:131–140.
258. Phillips T, Prados M, Bodell W, Levin V, V U, Gutin P. Rationale 274. Paganelli G, Grana C, Chinol M, et al. Antibody-guided three-
for and experience with clinical trials of halogenated pyrimidines step therapy for high grade glioma with yttrium- 90 biotin. Eur J
in malignant gliomas: the UCSF/NCOG experience. In: Dewey Nucl Med. 1999;26:348–357.
W, Edington M, Fry R, al. e, eds. Radiation Research: A Twentieth 275. Weiden P, Breitz H, Appelbaum J, et al. Radioimmunotherapy in
Century Perspective. Vol. 191. San Diego, CA: Academic; the treatment of Non-Hodgkin’s lymphoma (NHL.): Advantage
1992:601–606. of pretarget RIT (PRIT). Cancer Biother Radiopharm. 1999;14.
259. Ajlouni MI, Merrick HW, Skeel RT, Dobelbower RR, Jr. Concomitant 276. Pagel JM, Hedin N, Drouet L, et al. Conventional and Pretargeted
radiation therapy and constant infusion FUdR for unresectable Radioimmunotherapy Using an Anti-Murine CD45 Monoclonal
hepatic metastases. Am J Clin Oncol. 1990;13:532–535. Antibody in a Syngeneic, Disseminated Murine Leukemia Model.
260. Richman CM, DeNardo SJ, O’Grady LF, DeNardo GL. Blood. 2006;108.
Radioimmunotherapy for breast cancer using escalating fractionated 277. Schultz J, Lin Y, Sanderson J, et al. A tetravalent single-chain
doses of 131I-labeled chimeric L6 antibody with peripheral blood antibody-streptavidin fusion protein for pretargeted lymphoma
progenitor cell transfusions. Cancer Res. 1995;55:5916s–5920s. therapy. Cancer Res. 2000;60:6663–6669.
261. Breitz HB, Durham JS, Fisher DR, et al. Pharmacokinetics and 278. Theodore L, Fritzberg A, Schultz J, Axworthy D. Evolution of a
normal organ dosimetry following intraperitoneal rhenium-186- pretarget radioimmunotherapeutic regimen. In: Abrams P,
labeled monoclonal antibody. J Nucl Med. 1995;36:754–761. Fritzberg A, eds. Radioimmunotherapy of Cancer. New York:
262. DeNardo GL, Maddock SW, Sgouros G, Scheibe PO, DeNardo Marcel Dekker; 2000:195–221
SJ. Immunoadsorption: an enhancement strategy for radioimmu- 279. Forero A, Weiden PL, Vose JM, et al. Phase 1 trial of a novel anti-
notherapy. J Nucl Med. 1993;34:1020–1027. CD20 fusion protein in pretargeted radioimmunotherapy for
263. Goodwin DA, Meares CF. Pretargeting: general principles; B-cell non-Hodgkin lymphoma. Blood. 2004;104:227–236. Epub
October 10–12, 1996. Cancer. 1997;80:2675–2680. 2004 Mar 2002.
264. Goodwin DA, Meares CF, McCall MJ, McTigue M, Chaovapong W. 280. Reubi JC, Eisenwiener KP, Rink H, Waser B, Macke HR. A new
Pre-targeted immunoscintigraphy of murine tumors with indium-111- peptidic somatostatin agonist with high affinity to all five soma-
labeled bifunctional haptens. J Nucl Med. 1988;29:226–234. tostatin receptors. Eur J Pharmacol. 2002;456:45–49.
14 Stem cell/bone marrow transplantation
as biotherapy
ROBERT K. OLDHAM

stable chimeric state can be demonstrated [84], but


Autologous Bone Marrow GVHD (acute and chronic) results in the recognition of
Transplantation recipient tissues by transplanted donor T lymphocytes
and occurs in more than 50% of ALBMT patients. More
It is clear that bone marrow transplantation has become than half of these individuals will have a fatal outcome
a major technique in the treatment of metastatic cancer. [35, 47, 84]. A variety of techniques have been used to
The use of autologous bone marrow transplantation has attempt to eliminate T cells from these marrow grafts.
allowed for much higher doses of chemotherapy to be These have included monoclonal antibodies [63, 72],
given in an attempt to eliminate all of the cancer cells monoclonal antibody with complement [8, 33, 40, 51,
from the patient with the sacrifice of normal bone mar- 56, 64, 74, 80, 95], immunotoxins [23, 52, 65, 90], and
row function in the process. Autologous, cryopreserved physical methods [6, 36, 62] to eliminate T cells from the
bone marrow or peripheral blood stem cells (PBSC) can donor graft. Allogeneic BMT is covered later in this
be reinfused to reconstitute stem cells and bone marrow chapter, but various techniques to eliminate T cells have
function. This process is still dose limited by damage to been reviewed [37]. Many of the techniques to deplete
the gastrointestinal tract, liver, lung, heart and other the T cells for ALBMT are similar to techniques being
critical organs [25, 42]. Some progress has been made used to eliminate residual tumor cells in ABMT.
using this technique to allow dose escalation with che- Therefore, a review of this literature is critical to the
motherapeutic agents. Escalation of doses to the level reader who wants to understand all the current techniques
causing damage to secondary target organs is the current available for elimination of subsets of cells from bone
limitation of this technique. marrow prior to transplantation.
The problems of graft versus host disease (GVHD) ABMT is the most popular current method of bone
limits the use of allogeneic bone marrow transplantation marrow reconstitution and offers the advantage of avoid-
(ALBMT), although matching techniques and immuno- ing GVHD. Bone marrow cells in numbers (1 − 5 × 1010)
suppression have improved the results for allogeneic sufficient to reconstitute the individual’s marrow function,
grafts [87, 92]. By contrast, autologous bone marrow through the stem-cell transplant, can easily be obtained
transplantation (ABMT) is a technique that does not by multiple punctures of the bone with marrow aspira-
involve GVHD and allows for significant drug dose esca- tion and then storage of the separated cells in liquid
lation. Unfortunately, residual tumor cells can exist in nitrogen. These cells can be thawed and reinfused after
autologous bone marrow. Techniques must be developed high-dose chemotherapy with consistent reconstitution
to effectively eliminate all replicating tumor cells from of the bone marrow. In fact, bone marrows can now be
these specimens such that the cancer is never reinitiated separated and divided in such a way as to prepare one to
in the patient after cure by high dose therapy [68]. three grafts from a single bone marrow donor.
Considerable progress has been made in leukemias More recent application of this reconstituting tech-
and lymphomas, and there is an ongoing and increasing nique has involved the use of PBSC harvests through
effort with BMT in solid tumors [59, 72, 82, 86]. leukapheresis. This technique allows for stem cells to be
Histocompatible donors can be selected leading to harvested by high volume leukapheresis and represents
successful allogeneic transplants [24, 59, 84]. The per- a selection method for stem cells that avoids many of the
fect transplant only occurs with identical twins (available problems of tumor cell infiltration in the bone marrow.
in less than 1 in 300 transplants), but matching of donors Although peripheral blood may contain circulating
at the HLA-A, -B, -C, and -D loci and selecting for those tumor cells, the evidence thus far indicates that consid-
individuals with negative mixed lymphocyte cultures erable positive selection of stem cells can be effected by
have improved the results of ALBMT. Engraftment of a a peripheral blood harvest. This may obviate purging of

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 497
© Springer Science + Business Media B.V. 2009
498 Stem cell/bone marrow transplantation as biotherapy

bone marrow or at least make the purging process easier Ficoll-Hypaque gradients, isopyknic sedimentation on
by virtue of having fewer tumor cells in the stem-cell discontinuous Percoll gradients, and the use of centrifu-
preparations. The techniques for removing tumor cells gation in blood-cell separators without a gradient are all
from autologous marrow are numerous and have been techniques that have been used to separate mononuclear
extensively discussed elsewhere [48, 61, 65, 89, 94]. cells containing the stem-cell fraction [9, 21, 29, 43].
Two comprehensive reviews can be found in the work of These techniques have eliminated the problems of clump-
McIntyre [53] and Gross and Gee [37]. ing and cell lysis, yield high numbers of progenitor stem
cells, and routinely give marrow engraftment in patients.
The technique of removal, manipulation, and preparation
for ABMT and the technique of peripheral stem-cell iso-
The Preparation of Bone Marrow lation by leukapheresis in an outpatient setting have
for Transplantation in ABMT gained wide clinical acceptance. These techniques are
now available and being utilized broadly in the clinical
Marrow manipulation must be carried out to preserve
practice of oncology, such that ABMT has become a
the pluripotent hematopoietic stem cell in sufficient
major treatment technique in patients with previously
numbers to insure engraftment. All of the procedures to
refractory solid tumors [4, 10, 30, 32, 46, 76, 78, 84].
eliminate tumor cells discussed in this chapter must be
done in such a way to preserve adequate stem-cell activ-
ity. Clonogenic assays can be used to determine the
number and function of stem cells by assaying these Techniques to Eliminate Specific
preparations for granulocyte/macrophage colony form-
ing cells (GM-CFC), burst forming unit-erythroid
Subpopulations of Cells (Tumor
(BFU-E), and granulocyte, erythroid, macrophage, and Cells and Lymphocytes) from
megakaryocyte colony-forming cells (GEMM-CFC).
Long-term bone marrow cultures may also be used with Marrow Specimens
the Dexter culture system to measure cell renewal in There has been a huge number of specific techniques
these systems [2, 22, 28, 32, 54]. Recently, simple flow utilized to attempt to eliminate either tumor cells and/or
cytometry assessment of CD-34 lymphocyte numbers T cells from bone marrow. Elimination of the former is
has been used to predict stem cell numbers. While these essential in ABMT, and elimination of the latter is desir-
assays are useful to measure progenitor proliferation able in allogeneic BMT. Some of these techniques
and differentiation, the critical test is marrow engraft- depend on a biological agent and thus represent a form
ment in the patient treated by the ABMT technique. of ex vivo biotherapy, and others utilize chemotherapy,
ABMT hematopoietic recovery is usually seen in the radiotherapy, or physical techniques to effect marrow
20- to 40-day period after bone marrow infusion. White purging. Each of these techniques will be reviewed with
blood cell recovery generally precedes platelet recov- an emphasis on biotherapeutic techniques for bone
ery; and delayed engraftment can lead to fatal complica- marrow purging.
tions. These patients are often isolated from potential Although the techniques of purging tumor cells in
surrounding infectious risk and must be carefully moni- ABMT studies will be reviewed, it has been reported
tored for viral, bacterial, and fungal infections during that the purging of autologous marrow has no impact on
the course of ABMT. Once the white count recovers to survival in patients with lymphoma. Data from the
the level of 500 granulocytes/mm3 and the platelet count European Blood and Marrow Transplant Lymphoma
exceeds 30,000/mm3, the risk of infection and bleeding Registry (EBMT) demonstrated that the purging of bone
markedly decreases. ABMT impairs immune function marrow for tumor cells did not affect the rate of hemato-
but much less so than ALBMT. Immune recovery may logic engraftment or the risk of procedure-related death.
take months, and in some patients immune competence There was no significant difference in survival for
is never really fully reconstituted [27, 31, 49, 66, 99]. patients whose bone marrow was purged vs. case con-
To prepare marrow aspirates for transplantation, some trols left unpurged [83, 96].
positive selection of the appropriate reconstituting cells
by physical means is commonly done. This may include
the simple task of cryopreservation, which eliminates a Ex vivo Purging with Chemotherapy
large number of red blood cells, but may also include Various agents have been utilized to exploit the differen-
separation procedures to obtain mononuclear cells from tial toxicity for a specific chemotherapy drug on tumor
the marrow [9, 21, 43]. Isopyknic centrifugation of cells in contrast to the activity on the marrow stem
Robert K. Oldham 499

cells. Preclinical studies have demonstrated the effect tainly demonstrate the usefulness of this approach in
of corticosteroids [44], VP-16 and Verapamil [11], eliminating T cells. The application of this technique to
4-hydroperoxycyclophosphamide (4-HC) and platinum ABMT for purging malignant lymphocytes carrying the
[60], 1-β-d-arabinofuranosylcytosine (Ara-C) and deoxycy- antigens recognized by the Campath antibodies is the next
tidine [34], VP-16 and corticosteroids [79], and alkyl logical step. In multiple myeloma, antibody plus com-
lysophospholipids (ALP) [94]. These studies indicate that plement [88] can eliminate residual myeloma cells from
each of these approaches can eliminate tumor cells and/or the marrow. Antibody plus complement has been used in
T cells (glucocorticoids) from marrows to be reinfused in preclinical and clinical studies [55] to purge myelocytic
experimental systems and some in clinical trials [41]. leukemia cells from marrow prior to ABMT. An inter-
Mafosfamide [69] and 4-HC, both relatives of cyclo- esting study of an antibody conjugated to Adriamycin,
phosphamide, have been utilized in clinical studies to where the antibody also fixed complement, has been
eliminate residual tumor cells. These cyclophosphamide reported, but the preclinical activities demonstrated
derivatives, as well as other chemotherapy agents, insufficient selectivity for clinical application [98].
appear to be promising in the elimination of residual In addition to antibody alone and antibody-fixing
tumor cells from these marrow grafts [37]. complement, there has been a series of studies using
antibody conjugated to toxins, usually ricin, to prepare
immunotoxins that can be used in bone marrow purging
Biophysical and Physical Approaches [67, 93]. These immunotoxins have been used in ABMT
A variety of biophysical methods, including photoradia- for T-cell acute lymphoblastic leukemia [91] and to
tion [38], laser photoradiation [39], and isotope-medi- eliminate T cells to decrease GVHD in ALBMT. These
ated purging [50] have been used to purge marrow of ricin immunotoxins have been utilized in clinical stud-
unwanted tumor cells. These studies, sometimes with ies in marrow purging of solid tumors such as neuro-
chemotherapy purging, have primarily been used in pre- blastoma [13] and breast cancer [77].
clinical studies.
Physical separation methods such as elutriation [58]
can be used to positively select bone marrow stem cells
Combination Techniques
for ABMT, and this technique can be used as a negative Investigators are pursuing combination techniques with
selection technique against tumor cells in ABMT and T antibody tied to magnetic spheres (biotherapy plus
cells in ALBMT. Clinical studies of this technique are physical separation) to eliminate residual marrow tumor
underway and appear promising. Cell separators can be cells. These studies are very well reviewed by Gross and
utilized for the separation of mononuclear cells for mar- Gee [37] and will not be covered in detail here. Suffice
row [1] or peripheral blood stem cells [97] for transplan- it to say that various physical techniques are being
tation. Various other physical approaches have been developed where bone marrow is being purged by the
used, but the most promising and straightforward dispersion of magnetic beads coated with antibody.
approaches involve machines that can carry out such These antibody-coated beads attach to tumor cells and
processing in an automated format. can be extracted from bone marrow by passing the prep-
aration over a magnet. By pulling out the residual tumor
cells, one can prepare a bone marrow free of residual
Biotherapeutic Approaches tumor cells for reinfusion. These studies certainly appear
A variety of biotherapeutic approaches have been used to interesting, and further preclinical studies and early
treat bone marrow prior to reinfusion. Lymphokine acti- clinical applications are being pursued [37].
vated killer (LAK) cells induced by interleukin 2 [18]
have been used to purge bone marrow of residual tumor
cells with interesting preclinical results. Antibody alone
Stem-cell Selection
and antibody plus complement have been used by a vari- There is now increasing evidence that biological tech-
ety of investigators for the elimination of residual tumor niques to select stem cells by positive methods will result
cells [70]. The Campath series of antibodies may have in enhanced marrow preparations for ABMT. Antibody to
the dual use of purging lymphoma or leukemia cells CD-34 can be used to select stem cells from bone marrow
from the marrow and may also be useful in the in vivo or peripheral blood [5, 15]. This technique has mainly
elimination of residual tumor cells after ABMT. More been applied to bone marrow, but with the development
than 520 patients have been treated in various European of PBSC collection techniques [97], the same positive
transplant centers in an approach to deplete T cells and stem-cell selection process could be applied to peripheral
lessen GVHD in allogeneic BMT [16]. These results cer- blood. High speed clinical cell sorters and antibody-based
500 Stem cell/bone marrow transplantation as biotherapy

positive selection systems are both used in clinical trials as GVHD as has been well understood in allogeneic bone
methods to select and purify peripheral blood stem cells marrow transplantation since the early days of this tech-
for ABMT. nique [71]. GVHD has been well characterized for many
A technique with far-reaching implications is to cul- years. It is known to have an acute and more chronic vari-
ture stem cells continuously in vitro [17, 57]. This ety, primarily manifest by a clinical syndrome of skin
approach may allow for the repeated infusion of autolo- rash, liver and gastrointestinal dysfunction as well as a
gous bone marrow stem cells free of any residual tumor variety of autoimmune type effects [71]. A variety of
cells and without the presence of any immunologically strategies, primarily total body irradiation and/or sys-
active T cells [3]. The potential for the broad application temic chemotherapy has been used to reduce the number
of culture stem cells is obvious, even to the point of cryo- of T cells from the graft responsible for GVHD. These
preserving such stem cells for each interested individual, strategies have been more or less effective, usually requir-
while living and healthy, in anticipation of their use later ing lifelong treatment of patients who have experienced
in life in the presence of disease. Preserving cord blood or allogeneic GVHD.
fetal tissues for use later in life as stem cells is also being Of particular interest has been the concept of autolo-
explored. Cord blood is now routinely used for reconsti- gous GVHD which was initially quite a controversial
tution of marrow function after marrow ablative therapy theory [73]. The autoreactive cells found in autologous
[20]. Such techniques, while interesting, bring up the GVHD can cause damage to a variety of organs, most of
whole question of how far technology should take us in which are less severe than that seen in allogeneic BMT.
anticipating disease and dealing with the potential for Histocompatibility differences cannot explain this phe-
developing specific diseases. There are various social, nomenon since these autologous transplants cannot have
political, and economic consequences of techniques that histocompatibility differences. Therefore, immune dis-
may predict or be useful in the treatment of future dis- regulation is felt to play the major role in autologous
eases. In fact, the whole area of genetic therapy is now GVHD. Treatment is usually not necessary since the
coming forth as a potential major form of biotherapy. Not syndrome is normally benign and symptomatic care is
only can stem cells be grown in vitro for eventual use in usually sufficient for patients with manifestations of
reconstituting bone marrow, gene therapy with electropo- autologous GVHD [19].
ration and other techniques can be used to insert specific
genes in normal or defective human cells [45]. Very prim-
itive stem cells may carry few transplantation antigens
and after long-term culture may be useful broadly in
Graft-versus-tumor Effects
reconstituting stem cells. It is clear that biotherapy and Early evidence that bone marrow cells of allogeneic
gene therapy will lead to tremendous advances and oppor- BMT producing a graft-versus-leukemia effect has
tunities but also bring forth major considerations of “who prompted the more general idea of graft-versus-tumor
pays for clinical research” and “who should have access (GVT) effect of both ALBMT and ABMT. Most stud-
to these new techniques.” (See Chapter 3) ies found a correlation between GVH and reduced
relapse rate for patients with leukemia treated by
ALBMT. Similar observations have been seen, though
less striking, in ABMT [73]. The presence of autoreac-
Allogeneic Bone Marrow tive T cells related to Ia antigens in many of these syn-
dromes and also noting that Ia antigens are expressed
Transplantation on leukemias and lymphomas with perhaps similar
Allogeneic BMT has been successful in treating certain antigens being present on certain solid tumors. The
lymphoid and hemopoietic neoplasms. Initially, the total potential for identifying these cells and using them after
body irradiation plus high dose chemotherapy followed ex vivo application continues to intrigue investigators in
by ALBMT was felt to work primarily through the cytore- this area. Whether these cells may be purely T cells or
ductive effects of the chemoradiotherapy. Innovative sug- be some form of LAK or NK cell is currently under
gestions by Professor Georges Mathé as early as 1965 investigation. The ability to grow large numbers of acti-
and follow-up evidence reviewed by George Santos in vated T cells opens the possibility of utilizing cells
1972 suggested that there was an anti-tumor effect identified by transplantation techniques as useful for
independent of the chemoradiotherapy from ALBMT. their GVT activities [19].
This has more recently been clarified as a form of graft- The idea of using ABMT as primary therapy to reduce
versus-tumor (GVT) effect similar to the concept of tumor load and then to exploit the immunosuppression
Robert K. Oldham 501

that follows to administer partially or unmatched donor earlier, more intuitive observations of Mathé and Santos
lymphocyte infusions(DLI) for their further biothera- [71]. As summarized by Bishop [7], the non-ablative
peutic effect until the host eliminates them is being (mini) allogenic bone marrow transplant is being inves-
tested in several European centers and less frequently in tigated as a potentially less toxic transplant where the
the USA. Complete responses of persistent disease post donor cells are functioning as a form of adoptive cellu-
ABMT has caused considerable enthusiasm for this lar therapy using the GVT effect as biotherapy.
approach in some circles [19]. Studies of ALBMT comparing myeloablative condi-
Recent evidence has provided further credence to tioning regimens with non-myeloablative treatment in
the concept that the immune system is active in treating lymphoma and chronic lymphocytic leukemia has shown
leukemia post allogeneic BMT. This information has that the outcomes were similar and that the nonablative
come from studies in patients with chronic myelogenous regimens could be applied to patients with more comor-
leukemia and includes the following factors. bidities and in older age groups [81], however, the greater
power of myeloablative ALBMT has also been demon-
1. Marrow grafts that have been T-cell depleted have a
strated when used after failure of ABMT in lymphoma.
higher rate of leukemic relapse and less GVHD.
Studies from the International Bone Marrow Transplant
2. An inverse relationship has been seen in HLA identical
Registry have demonstrated complete remission in
BMT between the occurrence of GVHD and leukemia
selected patients who have failed ABMT and then treated
relapse.
with myeloablative ALBMT [26].
3. The GVT effect arises from a reduction in leukemic
Biotherapeutic approaches toward the elimination of
recurrences post allogeneic BMT compared to
residual tumor cells appear to be the most promising
homozygous twin transplants.
with regard to ABMT. Similar approaches to eliminate T
4. Relapsed allogeneic BMT CML patients can be rein-
cells from allogenic bone marrow may broaden its appli-
duced into complete remission simply by infusing
cation. The two most promising techniques appear to
donor leukocytes [85].
rest in the use of antibody to negatively or positively
select appropriate cell populations in marrow transplan-
tation and the use of physical separation methods, such
Conclusion as leukapheresis, to positively select for replicating stem
cells. Most specifically, the technique of peripheral blood
ABMT and ALBMT offer major opportunities in cancer
leukapheresis to select for stem cells, perhaps with their
biotherapy [19]. With ABMT the major question relates
eventual long-term culture for repetitive use, does appear
to the sensitivity of cancer to a dose escalation strategy of
to be a highly promising technique to provide stem-cell
chemotherapy and radiotherapy. If these modalities can
reconstitution in patients lethally damaged by high-dose
eliminate cancer from the patient at doses between the
chemotherapy and radiotherapy. These studies are at an
marrow lethal dose and the maximum tolerated dose to
early stage but they are rapidly evolving, with the tech-
the next organ system (gut, liver, lung, heart, etc.), then
nique of ABMT being already widespread in the clinical
the technique of ABMT with high-dose therapy may be
practice of oncology [10, 75]. The availability of colony-
broadly applicable and curative in certain solid tumors.
stimulating factors (see Chapters 17 and 18) provides a
This technique will always have major, inherent toxicity
further mechanism for the manipulation of stem-cell
because of the toxicities of the therapeutic agents.
numbers and activity. The outpatient use of autologous
However, short-term toxicity and even some risk of long-
bone marrow transplantation through leukapheresis to
term effects would be tolerated by patients with lethal
select the stem cells and the use of colony-stimulating
diseases if complete remissions and prolonged remission
factors to support stem cell growth in vitro and in vivo
duration can be engendered. On the other hand, if the
appear to be promising approaches in the treatment of
dose-response curve for human solid tumors is insuffi-
advanced solid tumors [12, 14, 76].
ciently steep to allow for the induction of complete
response and improved survival with these techniques,
then the broader application of this approach may not be
warranted, given the significant toxicities involved. References
Recent results, summarized above, using allogenic 1. Areman EM, Cullis H, Sacher RA, et al. Automated isolation of
peripheral blood mononuclear cells are particularly mononuclear cells using the Fenwall CS3000 blood cell separator.
encouraging, with clear GVT effects now being seen in In: Bone marrow purging and processing. New York: Wiley-Liss,
CML and other leukemias/lymphomas, confirming the 1990:379–385.
502 Stem cell/bone marrow transplantation as biotherapy

2. Ash RC, Detrick RA, Zanjani ED. Studies of human pluripotential 21. Ellis WM, Georgiou GM, Roberton DM, et al. The use of discon-
hemopoietic stem cells (CFU-GEMM) in vitro. Blood 1981; tinuous Percoll gradients to separate populations of cells from
58:309–316. human bone marrow and peripheral blood. J Immunol Methods
3. Balducci E, Azzarello G, Valenti MT, et al. The impact of progeni- 1984; 66:9–16.
tor enrichment, serum, and cytokines on the ex vivo expansion of 22. Fauser AD, Messner HA. Identification of megakaryocytes, mac-
mobilized peripheral blood stem cells: a controlled trial. Stem rophages, and eosinophils in colonies of human bone marrow con-
Cells 2003; 21:33–40. taining neutrophilic granulocytes and erythroblasts. Blood 1979;
4. Beelen DW, Ouabeck K, Graeven U, et al. Acute toxicity and first 53:1023–1027.
clinical results of intensive postinduction therapy using a modified 23. Filipovich AH, Vallera DA, Youle RJ, et al. Ex-vivo treatment of
busulfan and cyclophosphamide regimen with autologous bone donor bone marrow with anti-T-cell immunotoxins for prevention
marrow rescue in first remission of acute myeloid leukemia. Blood of graft-versus-host disease. Lancet 1984; 1:469–472.
1989; 74:1507–1516. 24. Fox NS, Chervenak FA, McCullough LB. Ethical considerations
5. Berensen RJ, Bensinger WI, Hill R,et al. Stem cell selection: clini- in umbilical cord blood banking. Obstet Gynecol January 1, 2008;
cal experience. In: Bone marrow purging and processing. New 111(1):178–182.
York: Wiley-Liss, 1990:403–413. 25. Frei E III, Canellos GP. Dose: a critical factor in cancer chemo-
6. Berensen RJ, Levitt LJ, Levy R, et al. Cellular immunoabsorption therapy. Am J Med 1980; 69:585–594.
using monoclonal antibodies: Selective removal of T cells from 26. Freytes CO, Loberiza FR, Rizzo JD, et al. Myeloablative alloge-
peripheral blood and bone marrow. Transplantation 1984; neic hematopoietic stem cell transplantation in patients who expe-
38:136–142. rience after autologous stem cell transplantation for lymphoma: a
7. Bishop MR. Non-myeloablative allogenic hematopoetic stem cell report of the International Bone Marrow Transplant Registry.
transplantation as adoptive cellular therapy. Principles Pract Biol Blood December 1, 2004; 104(12):3797–3803.
Ther Cancer Updates 2001; 2(1):1–9. 27. Friedrich W, Goldmann SF, Vetter U, et al. Immunoreconstitution
8. Blazar BR, Quinones RR, Heinitz KJ, et al. Comparison of three in severe combined immunodeficiency after transplantation of
techniques for the ex vivo elimination of T cells from human bone HAL-hapolidentical, T-cell-depleted bone marrow. Lancet 1984;
marrow. Exp Hematol 1985; 13:123–128. 1:761–764.
9. Bodger MP, Hann IM, Maclean RF, et al. Enrichment of pluripo- 28. Gartner S, Kaplan H. Long-term culture of human bone marrow
tent hemopoietic progenitor cells from human bone marrow. Blood cells. Proc Natl Acad Sci USA 1980; 77:4756–4759.
1984; 64:774–779. 29. Georgiou GM, Roberton DM, Ellis WM, et al. Enrichment from
10. Brandwine JM, Callum J, Rubinger M, et al. An evaluation of out- human bone marrow using a discontinuous Percoll gradient and
patient bone marrow harvesting. J Clin Oncol 1989; 7:648–650. soybean agglutinin in comparison with Ficoll-paque. Clin Exp
11. Cairo MS, Toy C, Sender L, et al. Combination chemotherapy and Immunol 1983; 53:491–496.
Verapamil to purge drug resistant leukemia cells from human bone 30. Gianni AM, Siena S, Bregni M, et al. Granulocyte-macrophage
marrow. In: Bone marrow purging and processing. New York: colony-stimulating factor to harvest circulating haemopoietic stem
Wiley-Liss, 1990:47–55. cells for autotransplantation. Lancet 1989; 2:580–585.
12. Caron PC, Scheinberg DA. Therapy of acute chronic leukemias using 31. Goodnough LT, Rudnick S, Price TH, et al. Increased preoperative
immunologic agents. Cancer Investigation 1996; 154(4):342–352. collection of autologous blood with recombinant human erythro-
13. Cassano WF, Zaytoun AM. Specific killing of neuroblastoma cells poietin therapy. N Engl J Med 1989; 321:1163–1168.
in vitro by immunotoxins. In: Bone marrow purging and process- 32. Gordon MY, Goldman JM, Gordon-Smith EC. 4-Hydroperoxy
ing. New York: Wiley-Liss, 1990:217–223. cyclophosphamide inhibits proliferation by human granulocyte-
14. Chao NJ, Schriber JR, Grimes K, et al. Granulocyte colony-stimu- macrophage colony-forming cells. (GM-CFC) but spares more
lating factor “mobilized” peripheral blood progenitor cells acceler- primitive progenitor cells. Leukemia Res 1985; 9:1017–1021.
ate granulocyte and platelet recovery after high dose chemotherapy. 33. Granger S, Janossy G, Francis G, et al. Elimination of T-lymphocytes
Blood 1993; 81:2031–2035. from human bone marrow with monoclonal T-antibodies and
15. Civin CI, Strauss LC, Fackler MJ, et al. Positive stem cell selec- cytolytic complement. Br J Haematol 1982; 50:367–374.
tion: basic science. In: Bone marrow purging and processing. New 34. Grant S, Howe C, Kuczynski T. Selective eradication of leukemic
York: Wiley-Liss, 1990:387–402. (L-CFU) versus normal (CFU-GM) myeloid progenitors in sus-
16. Cobbold SP, Hale G, Clark MR, et al. Purging in auto- and allografts: pension culture utilizing a prolonged exposure in 1-β-d-
CAMPATH monoclonal antibodies which use human complement arabinofuranosylcytosine (Ara-C) and deoxycytidine (dCyd). In:
and other natural effector mechanisms. In: Bone marrow purging Bone marrow purging and processing. New York: Wiley-Liss,
and processing. New York: Wiley-Liss, 1990:139–154. 1990:69–77.
17. Coutinho LH, Testa NG, Chang J, et al. The use of cultured bone 35. Grebe SC, Streilein JW. Graft-versus-host reactions: a review. Adv
marrow cells in autologous transplantation. In: Bone marrow purg- Immunol 1976; 22:119–221.
ing and processing. New York: Wiley-Liss, 1990:415–433. 36. Greenberg PL, Baker S, Link M, et al. Immunologic selection of
18. Cramer DV, Long GS. Lymphokine-activated killer (LAK) cell hemopoietic precursor cells utilizing antibody-mediated plate
purging of bone marrow. In: Bone marrow purging and processing. binding (“panning”). Blood 1985; 65:190–197.
New York: Wiley-Liss, 1990:125–137. 37. Gross S, Gee AP, Worthington-White DA. Transporting bone mar-
19. Devetten M, Armitage JO. Hematopoietic cell transplantation: row for in vitro purging before autologous reinfusion. In: Bone
progress and obstacles. Ann Oncol 2007; 18(9):1450–1456. marrow purging and processing. New York: Wiley-Liss,
20. Eapen M, Rubinstein P, Zhang MJ, et al. Comparable long-term 1990:541–549.
survival after unrelated and HLA-matched sibling donor hematopoi- 38. Gulati S, Atzpodien J, Lemoli RM, et al. Photoradiation methods
etic stem cell transplantations for acute leukemia in children for purging autologous bone marrow grafts. In: Bone marrow
younger than 18 months. J Clin Oncol 2006; 24(1):145–1515. purging and processing. New York: Wiley-Liss, 1990:87–102.
Robert K. Oldham 503

39. Gulliya KS, Batagllino M, Matthews JL. Breast cancer and laser 58. Noga SJ, Wagner JE, Rowley SD, et al. Using elutriation to engi-
photoradiation therapy: an in vitro model for autologous bone mar- neer bone marrow allografts. In: Bone marrow purging and pro-
row purging. In: Bone marrow purging and processing. New York: cessing. New York: Wiley-Liss, 1990:345–361.
Wiley-Liss, 1990:103–107. 59. O’Reilly RJ. Allogeneic bone marrow transplantation: current sta-
40. Hale G, Bright S, Chumbley G, et al. Removal of T cells from bone tus and future directions. Blood 1983; 62:941–964.
marrow for transplantation: a monoclonal antilymphocyte antibody 60. Peters RH, Brandon CS, Lobelia AA, et al. Combinations of
that fixes human complement. Blood 1983; 62:873–882. 4-dydroperoxycyclophosphamide (4-HC) and cisplatin for bone
41. Herve P, Racadot E, Flesch M, et al. Prevention of graft-versus-host marrow purging in autologous marrow transplantation: an update.
disease: Elimination of T-lymphocytes from bone marrow cells by In: Bone marrow purging and processing. New York: Wiley-Liss,
complement-dependent cytolysis with a combination of pan-T 1990:57–68.
monoclonal antibodies (letter). Presse Med 1984; 13(14):886–887. 61. Poynton CH, Reading CL. Monoclonal antibodies: the possibili-
42. Herzig GP. Autologous marrow transplantation for cancer therapy. ties for cancer therapy. Exp Biol 1984; 43:13–33.
In: McCullough J, Sandler SG, eds. Advances in immunobiology, 62. Poynton CH, Reading CL, Dicke KA. In: Dicke KA, Spitzer G,
blood cell antigens, and bone marrow transplantation. New York: Zander AR, eds. Autologous bone marrow transplantation. edited
Alan R. Liss, 1984:319–335. by K.A. Houston: University of Texas M.D. Anderson Hospital
43. Ho WG, Champlin RE, Feig SA, et al. Transplantation of ABH and Tumor Institute, 1985:433–437.
incompatible bone marrow: gravity sedimentation of donor mar- 63. Prentice HG, Blacklock HA, Janossy G, et al. Use of anti-T-cell
row. Br J Haematol 1984; 57:155–162. monoclonal antibody OKT3 to prevent acute graft-versus-host dis-
44. Kapoor N, Tutschka PJ, Copelan EA. Bone marrow purging with ease in allogeneic bone marrow transplantation for acute leukae-
glucocorticoids. In: Bone marrow purging and processing. New mia. Lancet 1982; 1:700–703.
York: Wiley-Liss, 1990:39–46. 64. Prentice HG, Blacklock HA, Janossy G, et al. Depletion of T lym-
45. Keating A, Toneguzzo F. Gene transfer by electroporation: a model phocytes in donor marrow prevents significant graft-versus-host
for gene therapy. In: Bone marrow purging and processing. New disease in matched allogeneic leukaemic marrow transplant recipi-
York: Wiley-Liss, 1990:491–498. ents. Lancet 1984; 1:472–476.
46. Kessinger A, Armitage JO, Smith DM, et al. High-dose therapy 65. Quinones RR, Youle RJ, Kersey JH. Anti-T cell monoclonal anti-
and autologous peripheral blood stem cell transplantation for bodies conjugated in ricin as potential reagents for human GVHD
patients with lymphoma. Blood 1989; 74:1260–1265. prophylaxis: effect on the generation of cytotoxic T cells in both
47. Korngold R, Sprent J. Lethal GVHD across minor histocompati- peripheral blood and bone marrow. J Immunol 1984;
bility barriers: nature of the effector cells and role of the H-2 com- 132:678–683.
plex. Immunol Rev 1983; 71:5–29. 66. Rappeport JM, Dunn MG, Parkman R. T lymphocytes in the
48. Krolick KA, Uhr JW, Vitetta ES. Selective killing of leukaemia peripheral blood of bone marrow transplant recipients.
cells by antibody-toxin conjugates: implications for autologous Transplantation 1983; 36:674–680.
bone marrow transplantation. Nature 1982; 295:604–605. 67. Raso V, Ritz J, Busala M, et al. Monoclonal antibody-ricin A chain
49. Lum LC, Seugneuret MC, Storb RF, et al. In vitro regulation of conjugate selectively cytotoxic for cells bearing the common acute
immunoglobulin synthesis after marrow transplantation. I: T-cell lymphoblastic leukemia antigen. Cancer Res. 1982; 42:457–464.
and B-cell deficiencies in patients with and without chronic graft- 68. Rill DR, Santana VM, Roberts WM, et al. Direct demonstration
versus-host disease. Blood 1981; 58:431–439. that autologous bone marrow transplantation for solid tumors can
50. Macklis RM. Radioisotope-mediated purging in bone marrow return a multiplicity of tumorigenic cells. Blood 1994;
transplantation. In: Bone marrow purging and processing. New 84(2):380–383.
York: Wiley-Liss, 1990:109–123. 69. Rizzoli V, Mangoni L. Pharmaceutical-mediated purging with
51. Martin PJ, Hansen JA. Quantitative assays for detection of residual mafosfamide in acute and chronic myeloid leukemias. In: Bone
T cells of T-depleted human marrow. Blood 1985; 65:1134–1140. marrow purging and processing. New York: Wiley-Liss,
52. Martin PJ, Hansen JA, Vitetta ES. A ricin A chain-containing 1990:21–38.
immunotoxin that kills human T lymphocytes in vitro. Blood 1985; 70. Rodt H, Kolb JH, Netzel B, et al. Effect of anti-T-cell globulin on
66:908–912. GVHD in leukemic patients treated with BMT. Transplant Proc
53. McIntyre EA. The use of monoclonal antibodies for purging autol- 1981; 13:257–261.
ogous bone marrow in the lymphoid malignancies. Clin Haematol 71. Santos GW. History of bone marrow transplantation. Clin Haematol
1986; 15:249–267. 1983; 12:611–639.
54. Metcalf D. Detection and analysis of human granulocyte-mono- 72. Santos GW. Bone marrow transplantation in leukemia: Current
cyte precursors using semi-solid cultures. Clin Haemotol 1979; status. Cancer 1984; 54:2732–2740.
8:263–285. 73. Santos GW. Syngeneic or autologous graft-versus-host disease. Int
55. Mills LE, Ball ED, Howell AL, et al. Efficacy of bone marrow J Cell Cloning 1989; 7:92–99.
purging in AML using monoclonal antibodies and complement. 74. Sharp TG, Sachs DH, Fauci AS, et al. T cell depletion of human
In: Bone marrow purging and processing. New York: Wiley-Liss, bone marrow using monoclonal antibody and complement-medi-
1990:165–170. ated lysis. Transplantation 1983; 35:112–120.
56. Mitsuyasu RT, Chaplin RE, Ho WG, et al. Prospective randomized 75. Sheridan WP, Wolf M, Lusk J, et al. Granulocyte colony-stimulating
controlled trial of ex vivo treatment of donor bone marrow with factor and neutrophil recovery after high-dose chemotherapy and
monoclonal anti-T cell antibody and complement for prevention of autologous bone marrow transplantation. Lancet 1989; 2:891–895.
graft-versus-host disease: a preliminary report. Transplant Proc 76. Sheridan WP, Begley GC, Juttner CA, et al. Effect of peripheral-
1985; 17:482–485. blood progenitor cells mobilized by filgrastim (G-CSF) on platelet
57. Naughton BA, Jacob L, Naughton GK. A three dimensional culture recovery after high dose chemotherapy. Lancet 1992; 339:640–644.
system for the growth of hematopoietic cells. In: Bone marrow 77. Shpall EJ, Anderson IC, Bast RC Jr, et al. Immunopharmacologic
purging and processing. New York: Wiley-Liss, 1990:435–445. purging of breast cancer from bone marrow for autologous bone
504 Stem cell/bone marrow transplantation as biotherapy

marrow transplantation. In: Bone marrow purging and processing. 89. Treleaven JG, Kemshead JT. Removal of tumour cells from bone
New York: Wiley-Liss, 1990:321–336. marrow: an evaluation of the available techniques. Hematol Oncol
78. Siena S, Bregni M, Brando B, et al. Circulation of CD34+ 1985; 3:65–75.
hematopoietic stem cells in the peripheral blood of high-dose 90. Vallera DA, Quinones RR, Azemove SM, et al. Monoclonal anti-
cyclophosphamide-treated patients: enhancement by intravenous body-toxin conjugates reactive against human T lymphocytes:
recombinant human granulocyte-macrophage colony-stimulating A comparison of antibody linked to intact ricin toxin with antibody
factor. Blood 1989; 74:1905–1914. linked to ricin A chain. Transplantation 1984; 37:387–392.
79. Skala JP, Rogers PCJ, Chan K-W, et al. Effect of methylpredniso- 91. Vallera DA, Uckun FM. Bone marrow purging with immunotoxins
lone and VP-16 on acute lymphocytic leukemia cells. In: Bone for treatment of T-cell acute lymphoblastic leukemia (T-ALL). In:
marrow purging and processing. New York: Wiley-Liss, Bone marrow purging and processing. New York: Wiley-Liss,
1990:79–86. 1990:191–205.
80. Slavin S, Waldmann H, Or R, et al. Prevention of graft-versus-host 92. van Rhee F, Lyn F, Cullous JO, et al. Relapse of chronic myeloid
disease in allogeneic bone marrow transplantation for leukemia by leukemia after allogeneic bone marrow transplant: the case for giving
T cell depletion in vitro prior to transplantation. Transplant Proc donor leukocyte transfusions before the onset of hematological
1985; 17:465–467. relapse. Blood 1994; 83:3377–3383.
81. Sorror ML, Storer BE, Maloney DG, et al. Outcomes after alloge- 93. Vitetta ES, Uhr JW. The potential use of immunotoxins in trans-
neic hematopoietic cell transplantation with nonmyeloablative or plantation, cancer therapy, and immunoregulation. Transplantation
myeloablative conditioning regimens for treatment of lymphoma 1984; 37:535–538.
and chronic lymphocytic leukemia. Blood January 1, 2008; 94. Vogler WR, Olson AC, Berdel WE, et al. Purging leukemia remis-
111(1):446–452. sion marrows with alkyl-lysophospholipids, preclinical and clinical
82. Sparkes MC, Crist ML, Sparkes RS, et al. Gene markers in human results. In: Bone marrow purging and processing. New York:
bone marrow transplantation. Vox Sang 1977; 33:202–205. Wiley-Liss, 1990:1–20.
83. Stewart AK, Vescio R, Schiller G, et al. Purging of autologous 95. Waldmann H, Polliak A, Hale G, et al. Elimination of graft-versus-
peripheral-blood stem cells using CD34 selection does not improve host disease by in vitro depletion of alloreactive lymphocytes with
overall or progression-free survival after high-dose chemotherapy a monoclonal rat anti-human lymphocyte antibody (CAMPATH-1).
for multiple myeloma: results of a multicenter randomized con- Lancet 1984; 2:483–486.
trolled trial. J Clin Oncol 2001; 19(17):3771–3779. 96. Williams CD, Goldstone AH, Pearce RM, et al. Pursing of bone
84. Sullivan KM, Shulman HM, Storb R, et al. Chronic graft-versus- marrow in autologous bone marrow transplantation for non-Hodg-
host disease in 52 patients: adverse natural course and successful kin’s lymphoma: A case-matched comparison with unpurged cases
treatment with combination immunosuppression. Blood 1981; by the European Blood and Marrow Transplant Lymphoma
57:267–276. Registry. J Clin Oncol 1996; 14:2454–2464.
85. Sullivan KM, Storb R. Allogeneic marrow transplantation. Cancer 97. Williams SF, Bitran JD, Richards JM, et al. Peripheral blood-derived
Invest 1984; 2:27–38. stem cell collections for use in autologous transplantation after high
86. Takaue Y, Watanabe T, Kawano Y, et al. Isolation and storage of dose chemotherapy: an alternative approach. In: Bone marrow purg-
peripheral blood hematopoietic stem cells for autotransplantation ing and processing. New York: Wiley-Liss, 1990:461–469.
into children with cancer. Blood 1989; 74:1245–1251. 98. Winter JN, Bass B, Bringman T, Nedwin G. Preclinical evaluation
87. Thomas ED. Karnofsky Memorial Lecture: Marrow transplanta- of immunoconjugates consisting of doxorubicin linked to comple-
tion for malignant diseases. J Clin Oncol 1983; 1:517–531. ment-fixing monoclonal antibody DLC-48 for bone marrow purg-
88. Tong AW, Dalton WS, Tsuruo T, et al. Elimination of chemoresis- ing of B-cell lymphomas. In: Bone marrow purging and processing.
tant myeloma clonogenic cells from human bone marrow by New York: Wiley-Liss, 1990:171–183.
monoclonal antibody and complement. In: Bone marrow purging 99. Witherspoon R, Lum LG, Storb R. Immunologic reconstitution
and processing. New York: Wiley-Liss, 1990:15–164. after human marrow grafting. Semin Hematol 1984; 21:2–10.
15 Cellular immunotherapy (CI), where have
we been and where are we going?
JOHN R. YANNELLI

Abbreviations used CI, Cellular Immunotherapy; NSCLC, subject; he says if the field were successful it would
Non-small cell lung cancer; DC, dendritic cell; TIL, bankrupt America. While being accurate to some degree,
tumor infiltrating lymphocyte; LAK, lymphokine activated this concept has forced a re-evaluation and a forced
killer cell; MHC, major histocompatibility complex; reduction in the in vitro approaches. A new focus has
IL-2, Interleukin-2; GM-CSF, granulocyte macrophage arisen which is on generating the effector cells in the
colony stimulating factor; TCR, T cell receptor; LDTA, patients, using vaccine therapies as stimulus. The majority
lymphocyte defined tumor antigens; MLTC, Mixed of newer CI protocols to date reflect this approach.
lymphocyte tumor cell culture Vaccines coupled with pre-treatment conditioning using
chemotherapeutic agents is the way of the future, at
least for the next few years.
Overview The current chapter will serve as a historical review
and present some of the new and exciting approaches
Previous reviews have detailed the development of being utilized in CI which is nearing the 25th anniver-
cellular therapies using immunologically nonspecific sary of Steve Rosenberg’s report on LAK cells in the
lymphokine activated killer cells (LAK), activated killer New England Journal of Medicine [1]. The Author’s
monocytes (AKM), and eventually tumor specific tumor experience since this time should serve to enlighten and
infiltrating lymphocytes (TIL). The innovations in large hopefully be comprehensive enough to allow the Reader
scale cell culture methodologies and the developments to consider CI and realize its value to cancer treatment.
in immune assessment approaches have made the thera-
pies more widespread as well as more effective against
a variety of tumor histologies.
While surgery, radiation therapy, and chemotherapy A Recap of where the Field
remain the accepted conventional treatments for cancer;
immunotherapy continues to make a case as a therapy to
has been
be used in the adjuvant setting. Surgery reduces tumor In the early 1980s, focus of CI was on the use of the
burden, radiation therapy kills residual tumor cells in innate arm (Natural killer cells, NK cells; Lymphokine
the surgical field, and chemotherapy kills residual and activated killer cells, LAK cells; and macrophages) [2]
micrometastatic disease both systemically and also in of the cellular immune response to treat primary and
the surgical field. However, neither radiation nor che- metastatic cancer. The idea that immune effector cells
motherapy provide tumor specificity and both can inca- such as LAK could be activated to recognize and kill
pacitate patients due to their effects on normal cells, tumor cells across a range of tumor histologies with little
particularly the bone marrow. Cellular immunotherapy consideration for immune specificity drove the laboratory
(CI), a potential fourth modality of cancer treatment, and clinical studies [1, 3, 4]. Interestingly, pre-clinical
can target tumor cells sparing normal cells and tissues. and murine studies being conducted at this time at the
The difficulty that is now faced, in 2008, focuses on NCI ([5–7], began to lay the framework for the future
the overall effectiveness of CI and justifying its use direction that CI would take. Their studies on lymphocytes
based on the cost which includes: cell culture reagents, that infiltrate tumors (tumor infiltrating lymphocytes, TIL)
technician time and recombinant cytokines. In the were fundamental to current CI approaches which now
current funding environment, these costs have mounted focus on antigen targeted therapies.
to levels which make it difficult for researchers at aca- In some cases, especially in melanoma, TIL were
demic NIH supported centers to seriously utilize CI, at shown to have functional reactivity against tumor cells
least in the way it has been practiced in the past. I have and were shown to traffic to tumor sites [8–11]. This
a colleague who scoffs at me when I talk about this ability to home to tumor is considered critical for

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 505
© Springer Science + Business Media B.V. 2009
506 Cellular immunotherapy (CI), where have we been and where are we going?

clinical effectiveness [8]. The discovery of the T cell alter tolerigenic mechanisms to allow the destruction of
receptor (TCR) and details regarding the three dimensional tumor but spare normal tissue. But, considering expres-
structure formed by the interaction of antigenic peptides sion patterns of some of the LDTAs, the overall benefit
and self-MHC (either class I or class II; major histo- of immediate tumor cell destruction and the potential
compatibility complex, MHC) furthered our under- for memory against tumor recurrence probably outweigh
standing and provided more rationale for their clinical some of these concerns.
use [12–15]. On a practical level, improved and stream- Our expertise and initial successes in immunotherapy
lined cell culture techniques, the bulk production of were gained in melanoma patients [47]. While the
newly identified growth and/or differentiation inducing majority of LDTAs have been described in melanoma, a
cytokines such as IL-2 and the identification of lympho- highly immunogenic tumor, LDTAs have also been
cyte defined tumor antigens (LDTAs) facilitated large identified in other solid (breast, colon and lung for
scale growth of these tumor reactive T cells (TIL) for example) and liquid tumors (lymphomas and leukemias)
clinical trials. (See Reviews [48–51]). This provided the rationale
TIL populations are comprised of varying numbers of to try CI in these tumors, once considered non-
CD4 helper and/or CD8 cytotoxic T cells (CTL). CD4 immunogenic and not candidates for experimental
helper cells recognize self MHC class II and 18–23 therapies. The goal is to improve CI in melanoma and
length amino acid peptides derived from tumor associ- apply the concepts and improvements to existing thera-
ated antigens. Specific CD4 cells help to orchestrate pies for other tumor histologies. All this considered,
immune responses through cytokine production. In some with the extensive knowledge and experiences gained
cases, certain populations of CD4+ cells, co-expressing thus far, investigators are now able to do what was origi-
CD25, have been shown to suppress immune responses nally intended in the early 1980s. That is to use defined
to tumor [16, 17]. This population will be further tumor antigens in vitro to generate more homogeneous
discussed below. CD8+ CTL recognize MHC class I anti-tumor T cell populations. The relative monetary
antigens and eight to ten amino acid length peptides. savings over earlier bulk T cell approaches using these
Following recognition, CTL are capable of cytokine selected T cell populations made the therapy attractive
production and killing of tumor cells by direct granule for clinical investigators, laboratory personnel, and
mediated cytotoxicity as well as delivering death sig- regulatory agencies. Extending these advances to the
nals mediated by FAS (See Reviews) [18–20]. These current use of vaccines to educate the immune system
two mechanisms may actually be preferentially used in with these antigen preparations in vivo against specific
different anatomic sites. Thus, a means to tailor CTL tumor antigen targets has become a more logical recent
therapies is suggested [21]. Much of what was learned focus. It may be more advantageous to generate anti-tumor
concerning lymphocyte recognition of tumors was immunity in vivo, an active form of immunotherapy,
gained from studies using TIL derived from melanoma and spare the uncertainties associated with large scale
patients. In addition to tumor biopsies, melanoma spe- cell culture. Using sound immunologic based principals
cific T cells have also been derived from both patient learned in viral and bacterial systems, these vaccines
and normal donor peripheral blood [5, 6, 22–42]. Both should be capable of boosting existing anti-tumor
melanoma specific TIL and specific T cells derived from immune responses and help to generate new responses
peripheral blood have been utilized as tools to identify against tumor antigens that have escaped recognition.
LDTAs using a variety of cellular and molecular tech- A point to consider, however, is whether immuniza-
niques (see reviews: [37], [43–45]. tion strategies will be effective against bulky metastatic
As mentioned above, in some cases, CI can target disease. It is possible that vaccines may only play a role
tumor cells and spare normal cells. With our greater in the adjuvant setting. Currently, the therapies are
understanding of the nature of LDTAs, however, it is structured in the following way. Surgery remains the
now clear that a number of tumor antigens are normal first line of therapy as a de-bulking procedure. Radiation
cell proteins with altered expression patterns. For exam- and/or chemotherapy are used for the removal of residual
ple, in TIL trials it was shown that melanoma specific and micro-metastatic disease. Concurrently, construct
TIL, following reinfusion, were also capable of killing vaccines and perform safety testing and deliver the
normal melanocytes [46]. Thus, it has become impor- vaccines in prime and boost strategies when the patient’s
tant to understand T cell specificity for tumors within immune system has recovered. This approach should
the context of mechanisms which control peripheral generate “sentinel” lymphocytes which can seek out and
tolerance. Emphasis now is to better understand toler- destroy small pockets of metastatic tumor which escape
ance, apply principals of tolerance to tumor models and recognition or become resistant to conventional therapies.
John R. Yannelli 507

The inherent nature of specific lymphocytes also Unfortunately, the therapy utilizing a short exposure of
predicts the development of memory to protect against leukapheresis products to high concentrations of IL-2
future tumor recurrence. Thus, the combination of (Pulsed-LAK; [61]) did not engender the enthusiasm
standard therapies with immunotherapies may provide which was originally intended. Thus, LAK cell therapy
valuable data and an exciting future in cancer therapies overall was considered too cumbersome a therapy to
[52–57]. This strategy of combined chemotherapy and routinely deliver. Today, with the renewed interest in the
immunotherapy is being investigated by our group innate immune response to cancer, an improved form of
(Hirschowitz and Yannelli, 2008 unpublished observa- this therapy may receive more attention and be utilized
tions). A clinical trial of maintenance chemotherapy in combination with more specific cellular approaches.
in non small cell lung cancer using Pemetrexed + or − Whether this approach is successful remains to be seen.
allogeneic tumor cell vaccine is being tested in our In the mid 1980s, the TIL trials began with the hope
center. It is early in the study but using stage III and IV that more durable clinical successes in a wider range of
NSCLC patients who possess bulky disease; we are tumor types could be realized. However, the enthusiasm
monitoring time to progression as well as immunologic that greeted the Science report by Rosenberg et al. in
responses to the vaccine. Both clinical evaluation and 1986 [60] detailing the murine TIL studies was not real-
immunologic monitoring taken together will provide ized in the early human clinical trials of TIL. In hind-
more impetus for moving forward with vaccines and a sight, the reason for this can be attributed to a number of
better understanding of much needed improvements. issues. Immunogenicity of the tumors being studied,
Finally, it also remains to be seen, however, whether clinical use of bulk TIL populations containing nonspe-
vaccines can be used as a defense against the initial cific T cell populations, and the failure to co-deliver
development of tumors. These studies are best tested in effective cytoreductive and/or immunosuppressive
murine models of spontaneous disease. Once we gain chemotherapy were some of the reasons for the disap-
experience with these models and novel vaccines are pointing results. Additionally, TIL were being used to
proven efficacious, populations at risk for certain forms treat bulky disease. We now realize the tumor burden
of cancer due to environmental factors or genetic dispo- contributes to a significant tumor and/or host derived
sition should be considered as potential candidates. suppressive environment. In retrospect, all of these
factors served to dampen the initial enthusiasm.
A breakthrough occurred in the early 1990s following
Use of LAK and TIL a retrospective analysis of the accumulated clinical and
laboratory data. A 1991 report by Aebersold et al. [24]
in the Treatment of Cancer, discussed characteristics of effector cells that were
important for clinical responses. For instance, age of the
are they gone Forever? TIL, doubling time of TIL, and the ability of the TIL to
In the early 1990s, frustration arose concerning CI trials immunologically recognize autologous or MHC
such as LAK [1, 3, 4] and TIL [22, 58–60]. These stud- matched tumor all proved to be predictors of clinical
ies had been ongoing since 1984 but examination of the responses. In two follow-up reports by this group, both
clinical responses made investigators consider more Rosenberg et al. [60] and Schwartzentruber et al. [62]
closely the cost to benefit ratio. While LAK was work- extended the observations following an analysis of 5
ing in some melanoma patients, it was a difficult therapy years of laboratory and clinical data. Interestingly, the
to deliver. The priming steps with large doses of recom- tumor most responsive to immunotherapy was mela-
binant IL-2, the repeated leukapheresis procedures, the noma. Other tumors such as breast, colon and lung can-
large-scale cell culture, and the requirement for large cer for instance did not appear to be responsive to TIL
numbers (>1011) of LAK cells in multiple cycles made therapy. The 30% or so of melanoma patients that
this a challenging therapy. In addition, the large doses of showed clinical responses all demonstrated the ability
IL-2 required for LAK cell maintenance made this clini- of their TIL to either kill in chromium release assays or
cally difficult for physicians, support staff and patients. specifically secrete cytokines following incubation with
The requirement for hospitalization, many times care in autologous or MHC matched melanoma tumor cells.
the ICU, made LAK therapy a challenge and doomed its The concept of shared tumor associated antigens pre-
spread to the general practice of Oncology. In addition, sented by common MHC antigens had already been
complete and durable responses were not the norm. described in reports by Topalian et al. [6], and Muul
The financial costs associated with this therapy were et al. [5, 39]. These investigators had shown that mela-
also daunting for both the clinicians and the patients. noma TIL could recognize autologous tumor cells and
508 Cellular immunotherapy (CI), where have we been and where are we going?

in many cases melanoma tumor cell lines expressing The frequency of successes, however, using standard TIL
shared MHC class I antigens (HLA-A2 for example). culture conditions was far less than seen in melanoma. In
These observations were later extended to MHC class II the majority of cases, nonspecific TIL or TIL with no
antigens [28, 29, 34]. functional reactivity were derived. This result may reflect
The relatively simple melanoma-TIL culture conditions an inherent inability to lyse fresh tumor cells other than
and the frequency of clinical responses make melanoma melanoma. But, even when tested against stable MHC
an ideal tumor model to study CI. While culture of enzy- matched cell lines, no specific lysis or cytokine release
matically digested tumor biopsies in high concentrations was usually observed. Based on the predictions made by
of IL-2 resulted in tumor specific CD4 and/or CD8 T the Investigators at the NCI [24, 60, 62], the infrequent
cells from about 40% of melanoma biopsies tested [35], generation of specific T cells from non-melanoma tumor
it is also one of the easier 1tumors from which to derive biopsies could explain the overall disappointing clinical
stable long term tumor cell lines [47, 63]. Stable TC results obtained in the early TIL trials. This assessment is
lines make detailed immunologic analysis easier. strengthened by other published reports [81, 85].
Interestingly, in the early studies, site of melanoma Despite these difficulties, novel cell culture method-
tumor biopsy appeared to predict whether specific T ologies have been developed since the early TIL trials
cells were obtained. That was, cutaneous and visceral were begun and improved results in non-melanoma T
lesions were more likely to provide anti-tumor specific- cell cultures. These studies showed that tumor specific T
ity than were TIL derived from tumor involved lymph cells could be derived from the peripheral blood of can-
nodes [35]. A recent report however, showed melanoma cer patients with a variety of cancers. Specific immuno-
reactive TIL could be derived from 39% of tumor logic reagents were then made available for clinical and
involved lymph nodes tested [64]. further immunologic studies. Interestingly, however, the
A recent TIL report reiterates the enormous potential proliferative potential of these T cells is not the same as
of this therapy, particularly in melanoma. With improve- T cells derived from solid tumor. TIL appear to grow
ments to both laboratory TIL procedures and pre-treat- more consistently and to higher levels than specific T
ment of patients with targeted chemotherapy, increased cells obtained from peripheral blood. TIL can prolifer-
clinical effectiveness of the approach was shown [46, ate in the presence of IL-2 alone and often do not need
65–67]. Use of highly selected, antigen specific mela- antigen restimulation. In the TIL studies, over 1011 T
noma reactive TIL following pretreatment of patients cells were often derived from less than 1 × 106 cells [23,
with the immuno-depleting drugs fludarabine and cyclo- 35]. In our own laboratory, we generated over 1011 effec-
phosphamide resulted in 6 of 13 objective clinical tor T cells from as few as 100 T cells. It is now evident
responses with 4 additional mixed responses. The that the growth and differentiation signals vary for T
infused TIL were shown to proliferate in vivo, traffic to cells derived from different anatomic sites including
tumor sites, and displayed marked functional reactivity. peripheral blood, solid and liquid tumors, and tumor
Compared to the early TIL trials, these results were involved or tumor free lymph nodes. These differences
markedly improved. Thus, in melanoma, TIL remains a reflect the maturational state of the T cells that are con-
very viable treatment option and continues to provide tained in these sites including differences in the expres-
the most interesting clinical results of all immunothera- sion of the chemokine receptor CCR7 and the isoforms
py’s tested to date. This group continues to lead in the of CD45 [86–88]. Thus, concerning peripheral blood,
laboratory aspects of CI with a study published online. factors such as when the blood sample is obtained may
In the most recent report, evidence is presented that very be critical. There are likely differences in what propor-
early TIL cultures are the most efficacious in terms of tions of immature versus mature T cells are contained in
anti-tumor reactivity. These T cells contained in early the sample which is most probably influenced by the
culture, while possessing the largest number of anti- overall tumor status and trafficking patterns of T cells. It
tumor T cell clones, do not suffer from long periods of is then unknown what is required for optimal expansion
time in cell culture where important clones are over- including: antigenic stimuli, frequency of restimulation,
grown or critical cell surface receptors and costimula- growth factors, and costimulation. With this said, the
tory molecules are down-regulated [68]. removal of the T cells from the potential suppressive
Examples of specific TIL derived from other tumor environment of a tumor bearing host [89–95] for in vitro
histologies include: colon cancer [69, 70]; esophageal expansion should be advantageous.
cancer [71]; breast cancer [72–75]; follicular lymphomas Autologous or allogeneic melanoma tumor cells were
[76]; ovarian cancer [77, 78]; renal cell cancer [79–82]; used as stimulators in mixed lymphocyte tumor cell cul-
and non small cell lung cancer [83, 84] to name a few. tures (MLTCs) and MHC class I restricted specific CTL
John R. Yannelli 509

were sometimes observed [96–99]. Peripheral blood to the future improvement of CI. Acknowledging the
from both normal donors and cancer patients were used critical importance of the discovery of LDTAs;
in these studies. Celis et al. [100] derived CTL from (Reviewed in: [43–45, 114]), the remainder of this
the peripheral blood of normal donors using antigenic Chapter will focus on the new approaches to CI utilizing
peptides derived from melanoma LDTAs. The culture defined LDTAs as targets for therapy.
conditions in these studies relied on low concentrations
of IL-2 and in some cases IL-7 provided at multiple
intervals during the stimulation and growth phases. More
recent methodologies used gene transfer of costimulatory Lymphocyte Defined Tumor
molecules such as CD80 into fresh or cultured tumor
cell lines. In these studies, tumor specific T cells against
Associated Antigens (LDTAs)
melanoma [38, 101] and other solid tumors were Lymphocyte defined tumor antigens (LDTAs) have been
observed including NSCLC [102], and renal cell cancer identified using a variety of molecular techniques (See
[103–105]. Interestingly, our studies (Bixby and Yannelli Reviews: [37, 43–45, 115]. There have been more than
[102] and [84] have shown that NSCLC specific CTL 70 identified at the writing of this Chapter. The tech-
could only be generated following exposure of T cells to niques used to identify antigens include: (1) cDNA
CD80 gene modified tumor cells confirming a require- cloning (described below) [26, 116, 117]; (2) a “reverse
ment for costimulation. immunology” approach which generate specific T cells
Finally, gene transfer as a tool to improve T cell reac- against peptides derived from sequences of serologically
tivity has gained recent interest after a series of early defined tumor antigens [118–123]; (3) peptide stripping
problems [106–110]. Our group at the NCI performed and HPLC [124–127]; and the newer more advanced
early studies using retroviral mediated gene transfer of molecular techniques including SEREX [128–130],
TIL with genes encoding the cytokines IL-2 and TNF- Proteomics [48, 131, 132], and Microarray analysis
alpha [111, 112]. The novelty of using IL-2 was associ- [133, 134]. The technique initially used to describe a
ated with the desire of infusing anti-tumor T cells that number of antigens in melanoma and other histologies
had an inherent capacity to release IL-2 in order to pro- which is still in use is cDNA-cloning [25–27, 30–33, 37,
vide growth stimulus to the T cells in vivo. Of course an 116, 135–137]. A distinct advantage of this technique is
issue with IL-2 release is controlling the endogenous that it is based on the availability of already defined
IL-2 production thus preventing the growth of T cells tumor specific T cells. It does not speculate as to their
with other specificities, particularly self reactive T lym- existence as some of the newer techniques do, thus
phocytes. TNF-alpha, however, was very intriguing bringing into question the relevance of the LDTA. The
during the early days of gene therapy. While we produced reagents required are tumor specific CTL, cDNA gener-
autologous melanoma tumor cell vaccines secreting ated from a tumor cell line recognized in an MHC
both IL-2 and TNF-alpha [63], much effort was made to restricted fashion by the T cells, and an indicator cell
transduce T cells with the same retroviral vectors. In line which can be transfected with both MHC antigens
murine models TNF-alpha delivered at the tumor site and pools of cDNAs. A cDNA library is prepared, pools
was observed to be a very potent modulator of tumor of cDNA are transfected into 293 human kidney cells or
response, speculated to be the result of tumor vascular COS cells which express the restriction element recog-
destruction. These results led to limited human trials nized by the T cells. The CTL must be tumor specific as
which were eventually halted by the FDA due to safety defined by extensive specificity analysis, and should
issues encountered early on [113]. Coupled with the specifically release cytokines which can be measured
understanding of lymphocyte defined tumor antigens using standard ELISA assays. In addition, a reasonable
(LDTAs) and the characterization of tumor reactive T number of T cells are required (generally 5 × 108 to 1 ×
cell receptors {Cole, 1994 #28}{Shilyansky, 1997 #26; 109). When pools of cDNA are identified which confer
Cole, 1997 #25; Cole, 1995 #27; Shilyansky, 1994 #30; recognition of the indicator cells by the CTL, the experi-
Nishimura, 1994 #29}{Roszkowski, 2005 #9}, most ments are repeated by screening dilutions of the cDNA
recent attempts are proving fruitful in the clinical setting pools until a single cDNA is identified. The cDNA is
[106–110]. Unfortunately, the laboratory requirements sequenced, the protein identified and characterized for
in expertise and molecular probes along with regulatory expression on tumors of the same histology, tumors of
issues make this approach less applicable to the general different histologies, and normal cells.
academic and oncologic setting. None the less, the The antigens discovered thus far by all these techniques
knowledge of TCRs and particularly LDTAs is critical are classified into three categories based on histologic
510 Cellular immunotherapy (CI), where have we been and where are we going?

expression of the genes. The first group identified are of a potent anti-tumor immune response. Vaccines con-
termed cancer testes antigens and are expressed in stitute an active form of immunotherapy since they are
normal testes, melanoma, and other tumor histologies not dependent on the injection of large numbers of in
including breast, lung, bladder, and squamous carcino- vitro derived effector cells. Rather, in most cases, the
mas. These antigens include the MAGE gene family: vaccine is delivered in hopes that the patient’s own
MAGE-1 [116], MAGE-2 [138] MAGE-3 [139] and immune system generates the effector cells in vivo.
BAGE [140]. Additional cancer testes antigens identified Thus, patients enrolled in vaccine trials must be immuno-
include MAGE-12 [141], GAGE [142], and NY-ESO-1 competent. Ideally, a cancer vaccine should: (1) exhibit
[130]. The second group includes melanocyte lineage minimal toxicity, (2) induce the appropriate tumor-
proteins which are expressed on normal melanocytes as specific immune responses against primary tumor and
well as tumor cells. These antigens include gp100 [26], metastatic lesions, (3) induce a memory response to
MART-1/Melan-A [25], tyrosinase [143] tyrosinase related protect against tumor recurrence and (4) be designed so
protein-1 (TRP-1) [33], tyrosinase related protein-2 [32] that it is affordable by cancer patients. The overall goal
and melanocyte-stimulating hormone receptor (MC1R) of course is to achieve a measurable therapeutic benefit,
[144]. The third group are mutated normal proteins which most likely derived from both the cellular and humoral
generate unique epitopes, such as the case with Beta- arms of the immune system. In a Review, Bodey et al.
catenin [137] and others including MUM-1 [145], CDK4 [170] summarizes the potential pitfalls to the approach.
[146], caspase-8 [147], and KIA0205 [148]. Certain of Since vaccines rely on specific B and T cell immunity,
these melanoma antigens such as tyrosinase, have also many factors must be considered including: (1) loss of
been shown to be recognized by class II restricted T cells, MHC expression by tumor cells, (2) low immunogenicity
one such antigen is tyrosinase [29, 34]. The questions of many of the LDTAs, (3) LDTA down-regulation, (4)
now which remain unanswered are which antigens are secretion of immuno-regulatory molecules by tumor
most biologically relevant to the cancer patient and which and infiltrating host cells, and (5) impaired antigen
peptides derived from each protein are the most immuno- presentation by Dendritic cells (DCs). These points
genic? Once these answers are obtained, it is possible that taken alone or in combination can predispose many
combinations of peptides, both CD4 and CD8 specific, vaccines to failure. However, in light of these difficul-
and/or antigens will be shown to be more efficacious as ties, successes have still been reported.
targets for vaccine strategies. In general, cancer vaccines can be divided into three
There are also a number of antigens, fewer defined at categories. These include purified LDTAs, whole
the moment, which have been identified in other cancers autologous or allogeneic tumor cell vaccines, and anti-
such as MUC-1 [149, 150], CEA [151], p53 [152] Her-2/ gen pulsed dendritic cells (DCs). Each has its advan-
neu [153, 154], SART-1 [155], ART-1 [156] and ART-4 tages and disadvantages and will not be discussed in
[157]. As populations of specific T cells are being iden- any particular order of importance. The first category
tified, so are the antigens they recognize. Our lab, utilizing is based on recently identified LDTAs and utilizes
specific T cells generated against CD80 gene modified these molecules as immunizing agents. This includes
NSCLC lines, identified two antigens which include a natural or synthetic proteins, protein derived peptides,
myoinositol monophosphatase protein (IMPA) [158, RNA, “naked” DNA, or DNA encoded in delivery
159]and a GTP binding protein termed GNAS [160]. vehicles such as recombinant viruses, plasmids, or
Unfortunately, based on normal tissue distribution, both recombinant bacteria (See Reviews above). Liposomes
appear to be minor histocompatibility antigens which have also been used for the delivery of a variety of
limit their eventual clinical use [161]. However, cDNA immunogens [171]. In the development of this type of
cloning strategy remains a very powerful tool for LDTA vaccine, melanoma again has served as the prototype
identification. tumor. Similar advantages have been taken of the
established immunogenicity, thus, melanoma vaccine
development has proceeded rapidly in the past 10 years.
Vaccines have been generated using the defined LDTAs
Cancer Vaccines including MART-1, gp100, gp75, MAGE-1, MAGE-3,
The major thrust of CI over the past 10 years, has been BAGE, GAGE, tyrosinase, GM2 and GD2 [162]. In
the development of vaccines for cancer treatment (See addition, as antigens are being discovered in other
Reviews [162–169]. Regardless of what constitutes the histologies, vaccine development is also proceeding
vaccine, the desired result is to provide tumor antigen in rapidly. This is evidenced by the use of carcinoembryonic
a stimulatory manner which allows the in vivo generation antigen (CEA), MUC-1, Her-2neu and Prostate specific
John R. Yannelli 511

membrane antigen (PSMA) as immunogens in clinical that can be a challenge depending upon the histology
trials [163, 164]. Overall, this approach is promising being studied. Some histologies such as melanoma are
for many cancer patients since it is based on sound easier to derive a cell line from than other histologies.
immunologic principals we have learned in bacterial The development of a stable tumor cell line requires
and viral systems. careful attention to growth medium, serum concentra-
The ability to apply LDTAs as a vaccine strategy is tion, growth factors, and substrate making some histolo-
very dependent on the knowledge of the patient’s tumor gies more difficult but certainly not impossible to derive
antigen expression. This can only easily be determined stable long term lines [63, 175–178] (Yannelli et al.,
by the availability of autologous tumor. Unfortunately, Lung Cancer, submitted 2009 still in review). As a solu-
this is not always possible since some patients are unre- tion, short term tumor lines are often attempted and used
sectable, or previously resected tumor lesions are for study [179–181]. Sometimes, however, one has to
unavailable at the time of the trial. In addition, since the question the integrity of the lines in regard to tumor cell
vaccine relies on priming the patient against tumor pro- content. The difficulty is that tumor biopsies are not
teins and MHC presented peptide antigens, MHC and 100% tumor cells. The tumor cell content is variable
LDTA expression by the tumor cells is critical. Decreased and biopsies are mixed with fibroblasts and host infil-
expression of these proteins by the tumor will hamper trating leukocytes. In addition, the viability of tumor
the effectiveness of the vaccine. There is much evidence cells can be a problem. Often, viable tumor cells exist
in the literature for the decreased expression of MHC on the periphery surrounding dead necrotic tissue in the
antigens, particularly HLA-A locus antigens by tumor center of the lesion. Following dissection and enzyme
cells [172–174]. One solution is combining the vaccine digestion the tumor cells often die in the processing.
with agents designed to maintain or increase MHC Hence, what appears to be a short term tumor cell
expression, such as gamma interferon. This will work line is in many cases normal cells such as fibroblasts
assuming that genetic changes have not occurred and/or macrophages. In the vaccine, what has the
affecting MHC class I or Beta-2 microglobulin produc- patient actually been exposed to, tumor cells or adher-
tion, assembly or transport to the cell surface. Secondly, ent normal cells? Many of these studies fail to fully
the inclusion of multiple antigens in the vaccine, a characterize the short term lines before use. Thus, clin-
multivalent approach, increases the expression of at ical results can be misleading. It is critical to evaluate
least one of the antigens if the other is lost by mutations the line for tumor antigen expression, MHC expression
or mechanisms of protein down-regulation exercised by and certainly cell viability. In our own studies with
selected cells in the tumor. Monitoring expression is NSCLC, we observe tumor colonies forming early in the
also important during the course of treatment to insure cell culture period. With time, however, these colonies
that changes do not occur affecting the potential long fail to increase in size and are overgrown by fibroblasts
term success of the vaccine. (Yannelli et al., Submitted to Lung Cancer, 2008).
A second vaccine strategy is a whole cellular vaccine None the less, clinical studies conducted by Berd et al.
using autologous or allogeneic fresh tumor cells or [182] utilized autologous whole-cell tumor vaccines in
tumor cell lines. Autologous tumor cell vaccines attempt stage IV melanoma patients. Their nonrandomized trial
in a controlled fashion to immunize the patient against demonstrated a 12.5% response rate, as measured by the
all the antigens that are endogenously expressed by their development of delayed-type hypersensitivity against
tumor. The use of autologous tumor cells to make indi- autologous melanoma tumor cells. In a subsequent
vidualized vaccines is certainly optimal; however, it study, they conjugated the hapten dinitrophenyl to the
does have its drawbacks. To accommodate multiple or autologous vaccine and observed striking inflammatory
even a single vaccination requires a reasonable amount responses in metastatic lesions of patients. In addition,
of fresh tumor biopsy. This in itself selects against a an increase in disease-free survival was achieved [183].
number of patients. Some patients have small lesions, Use of autologous tumor cell vaccines has also been
the tumor is inaccessible or the patient is inoperable. attempted in other tumor histologies with some degree
This is a particular problem in patients with NSCLC. of success. Vaccination in renal cell cancer also demon-
Widespread tumors in the thoracic cavity are many strated an anti-tumor immune response and a survival
times too difficult to obtain and it becomes an issue of benefit in treated patients [184]. More recently, Dillman
cost versus benefit to the patient. In some cases the et al. [181] showed that short term lines could be gener-
tumor cell content of the biopsy obtained is variable or ated 43% of the time in a study including 125 cancer
the tumor cells are necrotic. An alternative strategy patients of varying histologies. In this study, patients
is the development of an autologous tumor cell line but who developed positive delayed type hypersensitivity
512 Cellular immunotherapy (CI), where have we been and where are we going?

(DTH) responses to autologous tumor cell vaccines two. It is possible to be misled. What appears to be an
showed improved survival. anti-tumor response may actually have simply been an
For the difficulties described using autologous tumor, allo response. The vaccine was rejected with little to no
many investigators have chosen to study stable long impact on the patient’s tumor. Clinical ineffectiveness
term allogeneic tumor cell lines as vaccines due to their may be due to a faulty vaccine. On the other hand, there
availability and the presence of shared tumor antigens. has been literature over the years suggesting that the
Allogeneic lines are normally standardized in terms of generation of a third party allo-immune response can be
growth conditions and are amenable to manipulations like beneficial to the overall development of an anti-tumor
gene transfer. Use of these lines decreases the variability response [192].
in vaccine preparations used for multiple immunizations. An additional approach to tumor cell vaccines has
These lines also reduce the variability between patients. been to genetically modify the autologous or allogeneic
Again, however, it is critical to characterize antigen tumor cells to express agents designed to enhance the
expression by patient’s tumor and be sure the antigen or anti-tumor immune response [63, 193–196]. Immune
antigens are expressed by the allogeneic line chosen for stimulating molecules such as the cytokines TNF-alpha,
the vaccine. It is also important to match at least one or GM-CSF, gamma interferon or IL-2 for example have
more MHC class I antigens between the cell line and been used to aid in the development of immune
patient to insure the tumor derived peptides are pre- responses. In some cases the tumor cells have been
sented properly. The availability of over 30 NSCLC gene-modified to deliver lymphocyte co-stimulatory
lines in our lab makes an allogeneic tumor cell vaccine molecules such as CD80 [197–199]. Allogeneic mela-
very plausible. We have characterized: (1) the growth noma cell lines engineered to secrete IL-2 were admin-
characteristics of the lines, (2) tumor antigen expression istered to melanoma patients in separate studies [200,
of the lines, and (3) MHC expression of the lines [185]. 201] and each produced clinical responses, albeit weak
These are critical parameters in the development of ones. Haight et al. [202]demonstrated an antibody
allogeneic tumor cell vaccines. In a published study, a response to IL-2 expressing allogeneic neuroblastoma
polyvalent whole cell melanoma vaccine, prepared cell lines in six patients with neuroblastoma. Additional
with three allogeneic cell lines, produced a higher studies have tested a large number of cytokine genes for
median survival than historical controls and resulted in efficacy. Using a murine melanoma model, Dranoff
both cellular and humoral immune responses against et al. [203]compared ten genes for their ability to enhance
melanoma antigens [186, 187]. Evaluation of a vaccine tumor cell immunogenicity. Of all the genes tested,
(Melacine) consisting of lysates from two allogeneic retroviral transduction with GM-CSF produced potent,
cell lines in the adjuvant Detox found it comparable to long-lasting specific anti-tumor immunity. GM-CSF
chemotherapy in prolonging survival while causing was first recognized for its ability to stimulate the growth
much less toxicity [188–190]. However, while the and differentiation of myeloid lineage hematopoietic
clinical results appear promising in melanoma [191], progenitor cells. GMCSF is produced by T cells, mac-
there should be caution when using allogeneic reagents rophages, endothelial cells and fibroblasts in response to
for immunotherapy. These tumor cells, while potentially immune stimuli, which acts in a paracrine fashion to
expressing shared tumor antigens, also express alloge- recruit neutrophils, monocytes and lymphocytes for
neic MHC class I and in some cases class II antigens host defense [24–26]. Central to its use as an immune
which can give rise to potent allogeneic immune responses. adjuvant in vaccines, GMCSF is known to enhance
These vaccines may be quickly rejected before the macrophage activity and increase dendritic cell (DC)
tumor response is allowed to develop (direct response maturation and function, thereby augmenting antigen
involving T cell recognition of allogeneic MHC antigens processing and presentation [204]. Subsequent studies
on tumor cells). Alternatively, a slower developing revealed this immunostimulatory cytokine exerts pleio-
allogeneic response (indirect response involving pro- tropic effects on the immune system. Importantly, it
cessing and presentation of allogeneic MHC antigens promotes the differentiation and stimulation of dendritic
by host DCs), may develop along with the anti-tumor cells (DCs) which are professional antigen presenting
response. The response to allogeneic antigens may cells capable of sensitizing naïve T lymphocytes and
provide additional levels of critical cytokines necessary eliciting a primary T cell response. Interestingly, early
for the overall development of the anti-tumor response TIL trials in melanoma demonstrated that the specific
(bystander effect). It is important, however, to have the release of GM-CSF by TIL often correlated with significant
reagents available to decipher the response and deter- clinical responses [62]. One can speculate that the CD14+
mine if it is an anti-tumor, an allogeneic, or a mix of the cells recruited to the tumor site through inflammatory
John R. Yannelli 513

mechanisms become stimulated by the GM-CSF, receiving his or her tumor-derived peptides in the
undergo differentiation and accomplish tumor antigen context of self-MHC. The primary disadvantage of
presentation to T cells in draining lymph nodes. Thus, utilizing DCs as a vaccine, however, is the potential
GM-CSF may be an especially promising cytokine for development of immune responses against “self” anti-
gene-modified vaccines. gens shared by tumor and normal cells. Ludewig et al.
Synergy between antigenic protein and GMCSF has [219]reported on the induction of severe autoimmune
been shown in several clinical studies [205–209]. In disease with dendritic cell (DC) immunotherapy. With
NSCLC, Nemunatis et al. evaluated the effects of that stated, however, DCs may still be the most power-
GM-CSF gene-modified autologous cancer cells with ful tool in immunotherapy to date.
encouraging biologic and clinical results [210, 211].
Similarly, an autologous melanoma vaccine secreting
GMCSF reported in 1997 produced anti-tumor immune
responses associated with a partial but temporary
Dendritic Cells and Vaccines
clinical benefit [197, 212]. A Phase I clinical trial inves- DCs can be derived from normal donors and patients
tigating the biologic activity of these vaccines found with cancer including: B cell lymphoma [220, 221];
infiltration of T cells, DCs, macrophages and eosino- breast [222]; CML [223]; colon [224]; gliomas [225];
phils at the immunization sites in all 21 patients evalu- gynecologic malignancies [226]; medullary thyroid
ated [213, 214]. In addition, autologous tumor-reactive carcinoma [227]; melanoma [10, 228–230]; myeloma
CTL were detected following vaccination. GM-CSF- [231]; NSCLC [232]; prostate [233–235]; renal cell
secreting autologous tumor vaccines have also been uti- cancer [236, 237]; and various leukemias [238];
lized in clinical trials for renal cell cancer and prostate (additional references in [218]). Dendritic cells are
cancer [195, 215–217], and similar anti-tumor immune capable of presenting peptides processed from tumor
responses were observed. Together, these results dem- protein antigens to CD4 helper and CD8 cytotoxic T
onstrate the feasibility and safety of this vaccination cells. In addition, through cytokine release DCs can
strategy, the low level of toxicity to patients, and regulate the function of NK and NKT cells.
confirm the bioactivity of this reagent. In the laboratory, DCs are routinely generated from
Thirdly and the most recent approach is the use of CD14+ adherent monocytes obtained from either
antigen-loaded DCs as immunogens [218]. While all the peripheral blood draws or leukapheresis products. In
results are not in at the time of the preparation of this some cases, CD34+ precursors are used and are obtained
Chapter, we would speculate that in a future review the following mobilization using G-CSF (granulocyte
clinical use of DCs will have been proven in one appli- colony stimulating factor) infusions. The potency of
cation or another. We anticipate that DCs will constitute DCs to orchestrate T cell immune responses and the
a critical component of vaccines delivered to not only technologies recently developed to generate large numbers
cancer patients but also to many patients having a vari- has made them an attractive component of immuno-
ety of immunologic disorders. DCs are the most potent therapy protocols. Therefore it is not surprising that
antigen-presenting cells in the body. It is speculated in researchers have focused on their utilization in the
many cases that an inherent problem in tumor immunol- development of novel vaccine strategies for the treat-
ogy is the inadequate presentation of tumor antigens to ment of cancer [218, 239].
lymphocyte precursors. This vaccine approach is CD34+ DC progenitors differentiate into CD14+/
designed to remove DC precursors from the patient, dif- HLA-DR+/CD11+ intermediates that circulate systemi-
ferentiate them in an environment free of the tumor- cally and come to reside in peripheral tissues [240, 241].
induced suppression, and pulse them with relevant The differentiation of DCs from CD14+ precursors is a
tumor antigen before injection. The high expression of complex process which results in a heterogeneous popu-
costimulatory molecules (CD80, 86, 40) and MHC class lation of cells characterized by phenotypically defined
I and II molecules; the DCs ability to process and subtypes. In addition, depending upon the mode of
present relevant tumor derived peptides; and their abil- cellular and/or cytokine stimulation, these DC subtypes
ity to secrete IL-12p70 makes them very attractive as may acquire distinct and opposing functions in regard to
immunogens. Interestingly, DC based vaccines do not the cellular immune response (stimulatory, DC1; inhibi-
require that tumor antigens be fully characterized. This tory, DC2; See Reviews [218, 242–244]. This is the
approach can be utilized in the treatment of tumors with topic of much debate when considering which differ-
primarily undefined antigens. If autologous tumor cell entiation agents to use in the generation of DCs for
preparations are used to pulse the DCs, the patient is clinical trials.
514 Cellular immunotherapy (CI), where have we been and where are we going?

CD14+ cells removed from peripheral blood are capable of the differentiation process. These investigators
less than stimulatory for cellular immune responses showed that passage across an endothelial cell layer
due to the large amounts of IL-10 secreted following induced the expression of DC phenotypic characteristics
stimulation with soluble agents (LPS for instance) including costimulatory molecule expression, ingestion
(Yannelli et al., unpublished observation). Thus, of particulate matter, and expression of CD83, considered
proper differentiation of CD14+ cells to DCs is criti- by many to be a marker of DC maturation.
cal for T cell stimulation. The differentiation steps DCs can be matured from CD14+ precursors in vitro
most likely begin at the site of inflammation. in order to study DC biology and provide cells for clini-
Interestingly, it is the final maturation of DCs which cal studies. The clinical trials currently on-going use
appears to be critical to the development of a potent DCs pulsed with antigen in a variety of forms ranging
Th1 cellular immune response. Immature DCs are from purified peptides, crude cell extracts, necrotic or
considerably less stimulatory and may actually be apoptotic tumor cells, and RNA or DNA through gene
inhibitory toward the development of the desired cell transfer techniques. There are a number of good refer-
mediated immune response [245]. These maturation ences detailing the generation of DCs for clinical trials
steps can be monitored using appropriate in vitro [218, 247–249]. The initial cytokine requirements for
analysis. in vitro derivation of DCs from CD14+ cells was reported
CD14+ cells begin the differentiation process to by Romani et al. [250] and Sallusto and Lanzavecchia
“immature DCs”, that is, cells that are highly phago- [247]. These Investigators demonstrated that the culture
cytic, express moderate levels of MHC class I and II of adherent CD14+ cells in GM-CSF and IL-4 resulted
antigens and relatively low levels of adhesion and T cell in cells that lost CD14 expression, were CD1a+ and
costimulatory molecules, including CD40, CD80, and capable of phagocytosis or antigen uptake. The matura-
CD86 (See Review [243, 244]). At the site of inflamma- tion of DCs in vitro following 5–7 days of culture can be
tion (bacterial or viral infection, growing tumor nodule), accomplished by exposing the DCs to either: (1) monocyte
the immature DCs phagocytize particulate matter conditioned medium (MCM) [248]; (2) T cell conditioned
derived from damaged tissue including viral infected medium [251]; (3) CD4s bearing CD40L or anti-CD40
cells, bacteria, and necrotic or apoptotic tumor cell bod- mAb [252]; (4) exposure to combinations of cytokines
ies. At this point, presumably in response to local including IL-1Beta/IL-6/TNF-alpha and PGE-2 [223];
cytokine signals, the DCs begin the final differentiation or (5) TNF-alpha/Poly (IC) [253]. In considering which
to mature DCs. These signals can be replicated in vitro maturation factor to use for immunotherapy trials one
as will be discussed below. In vivo, it is thought that must consider (1) the need to recover a high percentage of
passage across an endothelial cell layer and a basement viable DCs, (2) the secretion of high amounts of IL-12p70
membrane, serves to further differentiate the DCs. These with lesser amounts of IL-10, (3) the expression of high
mature DCs traffic to the paracortical region of draining levels of T cell costimulatory molecules, and (4) insure
lymph nodes via afferent lymphatics. “Mature DCs”, at a minimum that the DCs are stimulatory for T cells in
having stopped phagocytosis, process the proteins into allogeneic mixed DC lymphocyte cultures (MLDCC).
peptides which are presented in both MHC class I and/ More importantly, studies should be conducted to dem-
or II molecules at the cell surface. The DCs change their onstrate that the mature DCs can indeed present tumor
cytokine secretion pattern from IL-10 production char- antigen to T cell precursors.
acteristic of CD14+ cells to the release of IL-12p70 A number of clinical trials are currently underway
(Th1 supporting cytokine). At this point, the mature studying DCs in a variety of tumor histologies (Reviewed
DCs up regulate the proteins necessary for antigen pre- in Steinman and Dhodapkar [218, 254]. One of the first
sentation. In the paracortical regions of the lymph nodes, reported DC clinical trials involved the vaccination of
the DCs present peptides derived from tumor antigen, autologous antigen pulsed DCs into patients with malig-
for instance, to both CD4 and CD8 precursor T cells nant B cell lymphoma [220]. A total of ten patients were
which circulate through the node. The antigen stimu- treated and a majority developed T cell mediated anti-
lated T cells express IL-2 receptors, proliferate and idiotype proliferative responses. Moreover, two patients
mature to effector cells. These antigen specific T cells experienced complete tumor regression, one had a partial
leave the node and circulate to the site of inflammation response, and three each stable disease or disease progres-
or the tumor bed to provide effector function. Randolf sion. This group subsequently initiated trials in patients
et al. [246] details what may occur in vivo with the mat- with multiple myeloma [231] and prostate cancer
uration of DCs from CD14+ precursors. In this elegant [255, 256]. One study examined the effects of different
report the investigators demonstrated that mature DCs routes of DC administration on the ability to induce
do not rely on proliferation since irradiated cells were immune responses in patients, and antigen-specific
John R. Yannelli 515

T cell responses were observed in all patients regardless In these studies, autologous DCs were generated from
of the route utilized [256]. However, the quality of the adherent or Miltenyi bead purified CD14+ cells obtained
responses was variable. A second group has been utiliz- from leukapheresis products. The DCs were pulsed with
ing autologous DCs pulsed with HLA-A2 binding apoptotic bodies derived from an allogeneic NSCLC
peptides derived from prostate-specific membrane line, 1650-TC. Following safety testing, DCs were used
antigen (PSMA). In the initial Phase I study, seven par- in prime and boost doses consisting of 100 million anti-
tial responders out of 51 participants had decreased serum gen pulsed DCs per indradermal/subcutaneous injec-
PSA levels. In a subsequent Phase II trial, GM-CSF was tion. Our results, focused on immune assessment,
included as an adjuvant to DC vaccination and 31% of showed that an immune responses was observed in
the evaluable patient’s experienced partial or complete 24/35 patients receiving the vaccine (69%). Importantly,
responses [232, 257–259]. Another trial reported the while the sample size was small, we showed that
immunization of patients with advanced ovarian or immune responses were generated regardless of prior
breast carcinoma with Her-2/neu or muc-1 peptide- conventional therapy or stage of the disease treated,
pulsed DCs. Peptide-specific CTL responses were although the final evaluation of the data indicated that
detected in five of ten patients, as assessed by intracel- early stage patients responded more favorably. None the
lular gamma-IFN staining and in microcytotoxicity less, even stages III and IV benefited. In our Center,
assays [150]. Kugler et al. [260] reported responses in 7 attention has now shifted from the DC trial to a new
of 17 patients with metastatic renal cell carcinoma vaccine we have constructed consisting of the apoptotic
following vaccination with tumor cell-dendritic cell bodies derived from 1650-TC along with recombinant
hybrids. There has been a recent study in patients with GMCSF. The simplicity of this approach has allowed
advanced colorectal cancer [224]. Of 15 patients vacci- the immunization of patients in four clinical centers
nated with autologous DCs pulsed with tumor RNA and across Kentucky, strengthening the value of the approach
keyhole limpet hemocyanin (KLH), 11 developed a in that it is a transferable technology. An evaluation of
positive skin test reaction to KLH and a decrease in the pilot data from ten patients has shown that similar
CEA levels were observed in seven of these patients. immunologic results are being obtained as determined
DC vaccination approaches have been examined in using gamma interferon ELISPOT assay. Thus, our
the setting of melanoma as well. In one study, patients present focus, combining vaccines with chemotherapy,
received DCs pulsed with either tumor cell lysate or a utilize this approach.
cocktail composed of different peptides [228]. Delayed- All in all, these clinical trials verify: (1) the ability to
type hypersensitivity reactions to peptide loaded DCs generate significant numbers of antigen pulsed DCs for
were observed in 11/16 patients and peptide-specific clinical trials, (2) the route of administration needed
CTL were identified, indicating the vaccination was for DC vaccination, and (3) in some cases significant
successful in generating an antigen-specific immune clinical responses. Thus, DCs have already proven their
response. Additionally, 5/16 patients exhibited demon- worth as a treatment option for patients with cancer.
strable regression of organ metastases yielding two A major question that remains, similar to the LAK and
complete and three partial responses. A recent Phase I TIL trials, is whether these cells can be routinely generated
clinical trial administered intravenously, tyrosinase and in small clinical centers. If these cells are only available
gp100 peptide-pulsed DCs derived from PBMC to at the NCI and large Cancer Centers, many patients will
melanoma patients [261]. In the 16 patients treated, one not benefit because they will never gain access to the
patient had a complete remission of metastatic disease, therapy. As we have demonstrated in our approach,
two had stable disease and two had mixed responses. making DCs is a cumbersome expensive process which
In another trial, DCs obtained from CD34+ progenitor requires FDA oversite. As discussed above, while our
cells were pulsed with melanoma peptides derived from results were fruitful and consistent with others in the
Melan-A/MART-1, tyrosinase, MAGE-3 and gp100, as literature, focus is shifting to easier tumor cell based
well as control antigens, and administered subcutane- vaccines.
ously to 18 patients [262]. An immune response was
generated against the control antigens in 16/18 patients. Treg Cells in Cancer: Biology and Potential
Ten of these patients exhibited responses to more than
two of the melanoma antigens, and seven of these had
Role in Tumor Immunity
regression of metastatic disease. A review of the immunologic literature over the past 4
In our own Center at the University of Kentucky, decades has suggested the role of immune cells in
Markey Cancer Center, we have concluded our clinical controlling the immune response. Early on, the focus
trial of antigen pulsed DCs in NSCLC [263, 264]. was on suppression. It was suggested that the immune
516 Cellular immunotherapy (CI), where have we been and where are we going?

response to tumors which appear to grow uncontrolla- effector function through surface or secreted TGF-beta
bly even in the presence of an apparent immune response [278]. While Tregs act directly on lymphocytes, it is
was greatly affected by their presence and function. In believed their activation may follow interaction with DCs
the past decade, the biology of suppressor cells, and [279] or in some cases tumor cells. Elegant studies by
their increased role in maintaining immunologic homeo- Rong Fu Wang have been the first to document antigen
stasis, has been uncovered. specificity of Tregs in a human TIL system, suggesting
The control of immune responses, that is limiting that tumor peptides may be directly recognized [280].
hyper-responsiveness and maintaining tolerance, has It is well documented that the percentage of Tregs is
been shown in part to be controlled by a subset of CD4 higher in cancer patients tumor tissue, blood, pleural effu-
helper cells called regulatory T cells or Tregs [265]. sions, and in draining lymph nodes [16, 275, 281–283].
Early work by Sakaguchi [266, 267] demonstrated that Interestingly, CD4+CD25+FOXP3+ cells were found to
thymectomized newborn mice developed autoimmu- be selectively drawn to tumor sites (tumor Tregs) by the
nity. The transfer of CD4+CD25+ T cells reversed expression of CCL22 by tumor macrophages and tumor
the effects. In rodent models, Treg depletions lead to cells [284]. This accumulation was correlated with poor
autoimmunity and increased allo and tumor reactivity clinical outcome. While murine studies have shown that
leading to rejection of transplanted tumors [268, 269]. the depletion of Tregs results in maximal tumor rejection
Since tumor antigens are in most cases normal antigens, [285] and, depletion of human Tregs enhances CD8 T cell
increased numbers of Tregs during anti-tumor responses responses following DNA immunization [286], there are
could inhibit anti-tumor reactivity. limited reports on the effects of tumor antigen vaccines
There are different phenotypes reported for human on levels of Treg cells before and after vaccination. We
Tregs. A naturally occurring form (nTreg) co-expresses have been examining this question in our DC vaccine
CD4, CD25, CTLA4, the newly described glucocor- study and are summarizing results for publication at this
ticoid induced receptor (GITR) and FoxP3 [270, 271]. time (Yannelli et al., 2008).
It is difficult to assess suppressive activity to all
CD4+CD25+ cells since CD25 is expressed by activated
CD4s. However, FoxP3, a transcriptional regulator,
appears to be the more attractive marker since induction
Immunological Monitoring
of FoxP3 on non-regulatory T cells by gene transfer In all the discussion presented, the key to understanding
induces a suppressor phenotype [270, 271]. Mutations the immune response which is a direct result of the
in the FoxP3 gene causes the absence of Treg cells in immune intervention is the development of immune
both mice and humans [272]. Most recently, it was assessment assays which are reliable, reproducible, and
reported that Foxp3 acts as both a transcriptional acti- can be utilized from site to site to insure comparative
vator and repressor [273] and that FoxP3 binds to the results. Prior to the immunologic assay, however, is the
promoters of well characterized regulators of T cell handling, preparation and cryopreservation of PBMC.
activation and function [274]. These issues will be discussed below.
Phenotypic analysis of normal donor peripheral blood In terms of collecting and handling of PBMC for fur-
reveals that 5–10% [275]of all peripheral CD4 cells are ther immunologic testing, it is standard to subject the
Tregs. Cancer patients can have as high as 20–25% [276] peripheral blood product or leukapheresis product to
Tregs in circulation. O’Garra and Vieira in Nature Ficoll Hypaque gradient separation in order to obtain
Medicine [277], described two types of Tregs, the natu- enriched mononuclear cells including monocytes, T
rally occurring nTregs (CD4+CD25+) and Treg produc- cells, B cells and NK cells. Within the T cell population
ing IL-10 (IL-10Treg, also secrete TGF-Beta). These two of course are contained the cytolytic cells, the helper
populations can take different names including natural cells and the regulatory T cells or Tregs. It is from these
(CD4+CD25+ arising in the thymus) and inducible cell types that many of the immune effector cells can be
(CD4+CD25− arising in the periphery, iTregs). A third routinely generated including CTL, DCs, and Tregs. In
population has also been described which is CD25− but most cases, immune assessment studies are not done
still expresses FoxP3 [277] Tregs have complex func- immediately. It is beneficial to wait until the patients
tions which result in both nonspecific or specific immune have undergone all of the immunizations and a reason-
suppression. Tregs act on naïve T cells as well as acti- able follow up time has passed.
vated T cells and function depends on both cell:cell The issue of cryopreparation and eventual thaw is a
contact or through cytokine release. Tregs can inhibit critical point that does not always appear to receive the
IL-2 production and may have inhibitory effects on CTL attention it should. Recently the NCI has requested
John R. Yannelli 517

uniformity in this approach and has funded a number of peripheral blood. Fluorescent labeled tetramers are con-
studies to consolidate approaches and provide a mean- structed to bind a unique MHC-peptide complex to the
ingful, easy technique for cryopreservation. In our studies, TCR, the tetramer tagged T cells are analyzed using a flow
we were struck by the use of human serum in cryo- cytometer. The majority of reagents currently available
preservation and its cost. We determined that the fluid are for class I peptide complexes although class II com-
replacement medium Plasmalyte A could be substituted plexes are becoming more available with time. Since
for either human serum or fetal bovine serum in the tetramers are specific for a unique MHC-peptide complex,
freezing of human PBMC [287]. The average cost of a this assay is most useful for the study of HLA-restricted
liter of Plasmalyte A is around $7. This provides signifi- peptide vaccines, although tetramers are currently not
cant savings over conventional serum. available for all antigens and all HLA types.
In order to assess immune function following vaccine The coupling of immune assessment assays and clinical
administration, most investigators examine cytotoxicity, responses is critical for an overall understanding of the
proliferation and cytokine production; all of which benefit of CI. While examples exist showing that certain
monitor antigen specific responsiveness of T cells. immune assessment assays correlate with tumor response,
Unfortunately, as detailed in Keilholz et al., 2002, short- this has not been a consistent finding. The difficulty at the
comings exist for each of the analyses mentioned [288]. moment is identifying the best in vitro assay which shows
In most cases, one is interested in the increase in spe- the greatest correlation across a range of studies done at
cific lymphocyte precursor frequency following immune different clinical centers. There is much focus in this area
intervention compared to that found before the vaccine and what are needed are standardization guidelines and
is delivered. An issue of course is using peripheral blood consistent controls to allow comparison across both study
as a source of T cells which may not represent the time points and across different geographic sites. When
immune response in lymph nodes and tumor tissue. this is accomplished it is likely that immune assays such
However, because of its universal availability, periph- as ELISPOT will gain favor as a primary endpoint analy-
eral blood remains an accepted anatomic site for study. sis in clinical immunotherapy studies.
Cytotoxicity as performed using chromium release is
not sensitive and relies on expansion of T cells from
the original sample. Proliferation assays are highly
variable and cytokine release assays also lack sensitivity.
Summary
Interestingly, an assay which has received much favor has As stated in the beginning of this Review, there has been
been the enzyme linked immunospot assay or ELISPOT. tremendous progress made in the field of CI since the
Many investigators utilize this approach and if stan- initial reports using LAK and TIL. Therapies have
dardization of immune assessment is to be made, it is progressed from using nonspecific killer cells to highly
likely in this assay. Lymphocytes to be analyzed are cul- specific T cells over the course of almost 20 years. The
tured with antigen presenting cells on membranes con- techniques used to generate these cells have been
taining cytokine capture antibodies in a 96 well format. improved to the point where it is possible for any labo-
Captured cytokine is visualized using color reagents ratory with immunologic expertise to participate in
and quantitated as spots indicating single cytokine pro- cellular immunotherapy clinical trials. This is true of
ducing cells. The results indicate an increase in “spots” course as long as the participating lab follows GLP
which correlate an increase in the relative number of procedures for the generation of therapeutic cells. The
cytokine producing cells. In our own studies, we have techniques exist for safe production and efficient moni-
observed increases in gamma interferon producing cells toring of cellular products for bacterial and viral contami-
following vaccine therapy, usually at multiple time nation. The field has progressed even further with the
points correlating with the prime and the boost vaccine discovery and molecular characterization of a variety of
[262, 263]. The single cell cytokine assays can also be LDTAs. The knowledge of these antigens has allowed
performed using flow cytometry with considerable sen- their use in generating more specific populations of T
sitivity. This assay can be sued to further delineate T cell cells for infusion into patients. In addition, these LDTAs
subsets secreting cytokine. An issue with this assay is have been used as targets for vaccines. Finally, the
the routine availability of a flow cytometer, although elucidation of the differentiation steps of DCs and the
ELISPOT analysis requires a reader system which can knowledge of large scale culture requirements has intro-
also be very expensive to purchase. duced DCs as another major component of CI. Most
Finally, tetramer staining is a non-functional, quantita- importantly, these cells have allowed the application of
tive measure of antigen specific T cell frequency in the immunotherapy to tumors other than melanoma.
518 Cellular immunotherapy (CI), where have we been and where are we going?

In closing, however, no matter what is accomplished 7. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the
adoptive immunotherapy of cancer with tumor-infiltrating lympho-
over the next 10 years we need additional insight into cytes. Science 1986 Sep 19;233(4770):1318–21.
the mechanisms where-by tumors evade immune 8. Pockaj BA, Sherry RM, Wei JP, Yannelli JR, Carter CS, Leitman
responses. While tumor and host induced suppression SF, et al. Localization of 111indium-labeled tumor infiltrating lym-
phocytes to tumor in patients receiving adoptive immunotherapy:
has been a major area of study over the years, it must Augmentation with cyclophosphamide and correlation with
receive continued and more focused attention in order response. Cancer 1994 Mar 15;73(6):1731–7.
for these therapies to work either alone or in the adju- 9. Kammula US, Lee KH, Riker AI, Wang E, Ohnmacht GA,
Rosenberg SA, et al. Functional analysis of antigen-specific T lym-
vant setting. It is especially important to determine why phocytes by serial measurement of gene expression in peripheral
the immune system is failing to recognize developing blood mononuclear cells and tumor specimens. J Immunol 1999
tumors in the micro-metastatic stage, that is, before the Dec 15;163(12):6867–75.
10. Thurner B, Haendle I, Roder C, Dieckmann D, Keikavoussi P,
tumor vasculature is developed. Thus, there needs to be Jonuleit H, et al. Vaccination with mage-3A1 peptide-pulsed
a better understanding of what the exact mechanisms mature, monocyte-derived dendritic cells expands specific cytotoxic
are where-by tumor cells are evading the immune sys- T cells and induces regression of some metastases in advanced
stage IV melanoma. J Exp Med 1999 Dec 6;190(11):1669–78.
tem and its recognition properties. If we understood this 11. Yee C, Thompson JA, Roche P, Byrd DR, Lee PP, Piepkorn M,
in the first place, strategies could be implemented to et al. Melanocyte destruction after antigen-specific immunotherapy
of melanoma: direct evidence of t cell-mediated vitiligo. J Exp
strengthen the early immune response in patients at risk Med 2000 Dec 4;192(11):1637–44.
and correct the deficits so the tumor never takes hold nor 12. Apostolopoulos V, Yu M, Corper AL, Teyton L, Pietersz GA,
has the opportunity to metastasize. It is hoped that at the McKenzie IF, et al. Crystal structure of a non-canonical low-affinity
peptide complexed with MHC class I: a new approach for vaccine
next revision of this Chapter, a complete understanding design. J Mol Biol 2002 May 17;318(5):1293–305.
of tumor and host induced suppression is obtained and it 13. Reinherz EL, Tan K, Tang L, Kern P, Liu J, Xiong Y, et al. The
is coupled with accepted and proven CI approaches to crystal structure of a T cell receptor in complex with peptide and
MHC class II. Science 1999 Dec 3;286(5446):1913–21.
cancer treatment. 14. Jardetzky T. Not just another Fab: the crystal structure of a TcR-
MHC-peptide complex. Structure 1997 Feb 15;5(2):159–63.
Acknowledgement This Chapter is dedicated to the 15. Stern LJ, Brown JH, Jardetzky TS, Gorga JC, Urban RG,
Strominger JL, et al. Crystal structure of the human class II MHC
many patients who have given themselves for these protein HLA-DR1 complexed with an influenza virus peptide.
studies. Additionally, on a local level, this Chapter is Nature 1994 Mar 17;368(6468):215–21.
dedicated to Dr. Bonnie Sigafus whose tireless dedica- 16. Woo EY, Chu CS, Goletz TJ, Schlienger K, Yeh H, Coukos G, et al.
Regulatory CD4(+)CD25(+) T cells in tumors from patients with
tion to patients with lung cancer has increased funding early-stage non-small cell lung cancer and late-stage ovarian can-
to scientists in need. Dr. Sigafus recently passed on but cer. Cancer Res 2001 Jun 15;61(12):4766–72.
17. Baecher-Allan C, Viglietta V, Hafler DA. Inhibition of human
her memory will always remain. CD4(+)CD25(+high) regulatory T cell function. J Immunol 2002
Dec 1;169(11):6210–7.
18. Lieberman J. Mechanisms of granule-mediated cytotoxicity. Curr
Opin Immunol 2003 Oct;15(5):513–5.
References 19. Clark R, Griffiths GM. Lytic granules, secretory lysosomes and
disease. Curr Opin Immunol 2003 Oct;15(5):516–21.
1. Rosenberg SA, Lotze MT, Muul LM, Leitman S, Chang AE, 20. Catalfamo M, Henkart PA. Perforin and the granule exocytosis
Ettinghausen SE, et al. Observations on the systemic administra- cytotoxicity pathway. Curr Opin Immunol 2003 Oct;15(5):522–7.
tion of autologous lymphokine-activated killer cells and recombi- 21. Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L,
nant interleukin-2 to patients with metastatic cancer. N Engl J Med Powell CA. Non-small-cell lung cancer molecular signatures reca-
1985 Dec 5;313(23):1485–92. pitulate lung developmental pathways. Am J Pathol 2003
2. Yannelli JR. The preparation of effector cells for use in the adop- Nov;163(5):1949–60.
tive cellular immunotherapy of human cancer. J Immunol Methods 22. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian
1991 May 17;139(1):1–16. SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and
3. West WH, Tauer KW, Yannelli JR, Marshall GD, Orr DW, Thurman interleukin-2 in the immunotherapy of patients with metastatic
GB, et al. Constant-infusion recombinant interleukin-2 in adoptive melanoma. A preliminary report. N Engl J Med 1988 Dec
immunotherapy of advanced cancer. N Engl J Med 1987 Apr 22;319(25):1676–80.
9;316(15):898–905. 23. Maleckar JR, Friddell CS, Sferruzza A, Thurman GB, Lewko WM,
4. Rosenberg SA, Lotze MT, Muul LM, Chang AE, Avis FP, Leitman West WH, et al. Activation and expansion of tumor-derived acti-
S, et al. A progress report on the treatment of 157 patients with vated cells for therapeutic use. J Natl Cancer Inst 1989 Nov
advanced cancer using lymphokine-activated killer cells and inter- 1;81(21):1655–60.
leukin-2 or high-dose interleukin-2 alone. N Engl J Med 1987 Apr 24. Aebersold P, Hyatt C, Johnson S, Hines K, Korcak L, Sanders M,
9;316(15):889–97. et al. Lysis of autologous melanoma cells by tumor-infiltrating
5. Muul LM, Spiess PJ, Director EP, Rosenberg SA. Identification of lymphocytes: association with clinical response. J Natl Cancer Inst
specific cytolytic immune responses against autologous tumor in 1991 Jul 3;83(13):932–7.
humans bearing malignant melanoma. J Immunol 1987 Feb 25. Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Rivoltini L,
1;138(3):989–95. Topalian SL, et al. Cloning of the gene coding for a shared
6. Topalian SL, Solomon D, Rosenberg SA. Tumor-specific cytolysis human melanoma antigen recognized by autologous T cells
by lymphocytes infiltrating human melanomas. J Immunol 1989 infiltrating into tumor. Proc Natl Acad Sci U S A 1994 Apr
May 15;142(10):3714–25. 26;91(9):3515–9.
John R. Yannelli 519

26. Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Sakaguchi K, 43. Renkvist N, Castelli C, Robbins PF, Parmiani G. A listing of human
Appella E, et al. Identification of a human melanoma antigen tumor antigens recognized by T cells. Cancer Immunol Immunother
recognized by tumor-infiltrating lymphocytes associated with in 2001 Mar;50(1):3–15.
vivo tumor rejection. Proc Natl Acad Sci U S A 1994 Jul 44. Rosenberg SA. Progress in human tumour immunology and immu-
5;91(14):6458–62. notherapy. Nature 2001 May 17;411(6835):380–4.
27. Kawakami Y, Eliyahu S, Sakaguchi K, Robbins PF, Rivoltini L, 45. Stevanovic S. Identification of tumour-associated T-cell epitopes
Yannelli JR, et al. Identification of the immunodominant peptides for vaccine development. Nat Rev Cancer 2002 Jul;2(7):514–20.
of the MART-1 human melanoma antigen recognized by the major- 46. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P,
ity of HLA-A2-restricted tumor infiltrating lymphocytes. J Exp Schwartzentruber DJ, Topalian SL, Sherry R, Restifo NP, Hubicki
Med 1994 Jul 1;180(1):347–52. AM, Robinson MR, Raffeld M, Duray P, Seipp CA, Rogers-Freezer
28. Topalian SL. MHC class II restricted tumor antigens and the role of
L, Morton KE, Mavroukakis SA, White DE, Rosenberg SA, Cancer
CD4+ T cells in cancer immunotherapy. Curr Opin Immunol 1994
Oct;6(5):741–5. regression and autoimmunity in patients after clonal repopulation
29. Topalian SL, Rivoltini L, Mancini M, Markus NR, Robbins PF, with antitumor lymphocytes. Science 2002 Sep 19;298:850–854.
Kawakami Y, et al. Human CD4+ T cells specifically recognize a 47. Houghton AN, Gold JS, Blachere NE. Immunity against cancer:
shared melanoma-associated antigen encoded by the tyrosinase lessons learned from melanoma. Curr Opin Immunol 2001
gene. Proc Natl Acad Sci U S A 1994 Sep 27;91(20):9461–5. Apr;13(2):134–40.
30. Robbins PF, el-Gamil M, Li YF, Topalian SL, Rivoltini L, 48. Lee SY, Jeoung D. The reverse proteomics for identification of
Sakaguchi K, et al. Cloning of a new gene encoding an antigen tumor antigens. J Microbiol Biotechnol 2007 Jun;17(6):879–90.
recognized by melanoma-specific HLA-A24-restricted tumor-infil- 49. Parmiani G, Russo V, Marrari A, Cutolo G, Casati C, Pilla L, et al.
trating lymphocytes. J Immunol 1995 Jun 1;154(11):5944–50. Universal and stemness-related tumor antigens: potential use in cancer
31. Wang RF, Robbins PF, Kawakami Y, Kang XQ, Rosenberg SA. immunotherapy. Clin Cancer Res 2007 Oct 1;13(19):5675–9.
Identification of a gene encoding a melanoma tumor antigen recog- 50. Elsner HA, Eiz-Vesper B, Blasczyk R, Bade-Doding C. Allele-
nized by HLA-A31-restricted tumor-infiltrating lymphocytes. J specific peptide presentation of human leukocyte antigens: impli-
Exp Med 1995 Feb 1;181(2):799–804. cations for tumor immunotherapy. Anticancer Res 2007 Jul–Aug;
32. Wang RF, Appella E, Kawakami Y, Kang X, Rosenberg SA. 27(4A):2075–7.
Identification of TRP-2 as a human tumor antigen recognized by 51. Adler AJ. Mechanisms of T cell tolerance and suppression in
cytotoxic T lymphocytes. J Exp Med 1996 Dec 1;184(6):2207–16. cancer mediated by tumor-associated antigens and hormones. Curr
33. Wang RF, Parkhurst MR, Kawakami Y, Robbins PF, Rosenberg Cancer Drug Targets 2007 eb;7(1):3–14.
SA. Utilization of an alternative open reading frame of a normal 52. Tarhini AA, Kirkwood JM, Gooding WE, Moschos S, Agarwala
gene in generating a novel human cancer antigen. J Exp Med 1996 SS, A phase 2 trial of sequential temozolomide chemotherapy fol-
Mar 1;183(3):1131–40. lowed by high-dose interleukin 2 immunotherapy for metastatic
34. Topalian SL, Gonzales MI, Parkhurst M, Li YF, Southwood S, melanoma. Cancer 2008 Aug 21;113:1632–1640.
Sette A, et al. Melanoma-specific CD4+ T cells recognize nonmu- 53. Zhang L, Dermawan KT, Jin ML, Xiong SD, Chu YW, Does chemo-
tated HLA-DR-restricted tyrosinase epitopes. J Exp Med 1996 therapy augment anti-tumor immunotherapy by preferential impair-
May 1;183(5):1965–71.
ment of regulatory T cells? Med Hypotheses 2008 Aug 6;71:802–804.
35. Yannelli JR, Hyatt C, McConnell S, Hines K, Jacknin L, Parker L,
et al. Growth of tumor-infiltrating lymphocytes from human solid 54. Chareyron S, Alamir M, Mixed immunotherapy and chemotherapy
cancers: summary of a 5-year experience. Int J Cancer 1996 Feb of tumors: feedback design and model updating schemes. J Theor
8;65(4):413–21. Biol 2009;258:444–454.
36. Yannelli JR, McConnell S, Parker L, Nishimura M, Robbins P, Yang 55. Ramakrishnan R, Antonia S, Gabrilovich DI. Combined modality
J, et al. Melanoma tumor-infiltrating lymphocytes derived from four immunotherapy and chemotherapy: a new perspective. Cancer
distinct anatomic sites obtained from a single patient: comparison of Immunol Immunother 2008 Oct;57(10):1523–9.
functional reactivity and melanoma antigen recognition. J 56. Bellone M, Mondino A, Corti A. Vascular targeting, chemotherapy
Immunother Emphasis Tumor Immunol 1995 Nov;18(4):263–71. and active immunotherapy: teaming up to attack cancer. Trends
37. Van den Eynde BJ, van der Bruggen P. T cell defined tumor anti- Immunol 2008 May;29(5):235–41.
gens. Curr Opin Immunol 1997 Oct;9(5):684–93. 57. Royer PJ, Bougras G, Ebstein F, Leveque L, Tanguy-Royer S,
38. Yang S, Darrow TL, Seigler HF. Generation of primary tumor- Simon T, et al. Efficient monocyte-derived dendritic cell genera-
specific cytotoxic T lymphocytes from autologous and human lym- tion in patients with acute myeloid leukemia after chemotherapy
phocyte antigen class I-matched allogeneic peripheral blood treatment: application to active immunotherapy. Exp Hematol
lymphocytes by B7 gene-modified melanoma cells. Cancer Res 2008 Mar;36(3):329–39.
1997 Apr 15;57(8):1561–8. 58. Dillman RO, Oldham RK, Barth NM, Cohen RJ, Minor DR, Birch
39. Hom SS, Topalian SL, Simonis T, Mancini M, Rosenberg SA. Common R, et al. Continuous interleukin-2 and tumor-infiltrating lympho-
expression of melanoma tumor-associated antigens recognized by cytes as treatment of advanced melanoma. A national biotherapy
human tumor infiltrating lymphocytes: analysis by human lymphocyte study group trial. Cancer 1991 Jul 1;68(1):1–8.
antigen restriction. J Immunother 1991 Jun;10(3):153–64. 59. Oldham RK, Dillman RO, Yannelli JR, Barth NM, Maleckar JR,
40. Hom SS, Schwartzentruber DJ, Rosenberg SA, Topalian SL. Sferruzza A, et al. Continuous infusion interleukin-2 and tumor-derived
Specific release of cytokines by lymphocytes infiltrating human activated cells as treatment of advanced solid tumors: a National
melanomas in response to shared melanoma antigens. J Immunother Biotherapy Study Group Trial. Mol Biother 1991 Jun;3(2):68–73.
1993 Jan;13(1):18–30. 60. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber
41. Frederiksen KS, Lundsgaard D, Freeman JA, Hughes SD, Holm DJ, Weber JS, et al. Treatment of patients with metastatic mela-
TL, Skrumsager BK, et al. IL-21 induces in vivo immune activa- noma with autologous tumor-infiltrating lymphocytes and interleu-
tion of NK cells and CD8(+) T cells in patients with metastatic kin 2. J Natl Cancer Inst 1994 Aug 3;86(15):1159–66.
melanoma and renal cell carcinoma. Cancer Immunol Immunother 61. Horton SA, Oldham RK, Yannelli JR. Generation of human lym-
2008 Oct;57(10):1439–49. phokine-activated killer cells following brief exposure to high dose
42. Bercovici N, Haicheur N, Massicard S, Vernel-Pauillac F, Adotevi interleukin 2. Cancer Res 1990 Mar 15;50(6):1686–92.
O, Landais D, et al. Analysis and characterization of antitumor 62. Schwartzentruber DJ, Hom SS, Dadmarz R, White DE, Yannelli
T-cell response after administration of dendritic cells loaded JR, Steinberg SM, et al. In vitro predictors of therapeutic response
with allogeneic tumor lysate to metastatic melanoma patients. in melanoma patients receiving tumor-infiltrating lymphocytes and
J Immunother 2008 Jan;31(1):101–12. interleukin-2. J Clin Oncol 1994 Jul;12(7):1475–83.
520 Cellular immunotherapy (CI), where have we been and where are we going?

63. Yannelli JR, Hyatt C, Johnson S, Hwu P, Rosenberg SA. 79. Brouwenstijn N, Gaugler B, Kruse KM, van der Spek CW, Mulder
Characterization of human tumor cell lines transduced with A, Osanto S, et al. Renal-cell carcinoma-specific lysis by cytotoxic
the cDNA encoding either tumor necrosis factor alpha (TNF- T-lymphocyte clones isolated from peripheral blood lymphocytes
a) or interleukin-2 (IL-2). J Immunol Methods 1993 May and tumor-infiltrating lymphocytes. Int J Cancer 1996 Oct
5;161(1):77–90. 9;68(2):177–82.
64. Benlalam H, Labarriere N, Linard B, Derre L, Diez E, Pandolfino 80. Finke JH, Rayman P, Hart L, Alexander JP, Edinger MG, Tubbs
MC, et al. Comprehensive analysis of the frequency of recognition RR, et al. Characterization of tumor-infiltrating lymphocyte
of melanoma-associated antigen (MAA) by CD8 melanoma infil- subsets from human renal cell carcinoma: specific reactivity defined
trating lymphocytes (TIL): implications for immunotherapy. Eur J by cytotoxicity, interferon-gamma secretion, and proliferation.
Immunol 2001 Jul;31(7):2007–15. J Immunother Emphasis Tumor Immunol 1994 Feb;15(2):91–104.
65. Dudley ME, Wunderlich JR, Shelton TE, Even J, Rosenberg SA. 81. Steger GG, Pierce WC, Figlin R, Czernin J, Kaboo R, DeKernion
Generation of tumor-infiltrating lymphocyte cultures for use in JB, et al. Patterns of cytokine release of unselected and CD8+
adoptive transfer therapy for melanoma patients. J Immunother selected renal cell carcinoma tumor-infiltrating lymphocytes (TIL).
2003 Jul–Aug;26(4):332–42. Evidence for enhanced specific killing of tumor necrosis factor-
66. Dudley ME, Wunderlich JR, Yang JC, Hwu P, Schwartzentruber secreting/IL-6 nonsecreting TIL in vitro and correlation with com-
DJ, Topalian SL, et al. A phase I study of nonmyeloablative chemo- plete response in vivo. Clin Immunol Immunopathol 1994
therapy and adoptive transfer of autologous tumor antigen-specific Aug;72(2):237–47.
T lymphocytes in patients with metastatic melanoma. J Immunother 82. Koo AS, Tso CL, Shimabukuro T, Peyret C, deKernion JB,
2002 May–Jun;25(3):243–51. Belldegrun A. Autologous tumor-specific cytotoxicity of tumor-
67. Dudley ME, Wunderlich JR, Yang JC, Sherry RM, Topalian SL, infiltrating lymphocytes derived from human renal cell carcinoma.
Restifo NP, et al. Adoptive cell transfer therapy following non- J Immunother 1991 Oct;10(5):347–54.
myeloablative but lymphodepleting chemotherapy for the treat- 83. Seki N, Hoshino T, Kikuchi M, Hayashi A, Itoh K. HLA-A locus-
ment of patients with refractory metastatic melanoma. J Clin Oncol restricted and tumor-specific CTLs in tumor-infiltrating lympho-
2005 Apr 1;23(10):2346–57. cytes of patients with non-small cell lung cancer. Cell Immunol
68. Tran KQ, Zhou J, Durflinger KH, Langhan MM, Shelton TE, 1997 Feb 1;175(2):101–10.
Wunderlich JR, Robbins PF, Rosenberg SA, Dudley ME, Minimally 84. Yannelli JR, Hirscowitz E, Wroblewski JM. Growth and functional
cultured tumor-infiltrating lymphocytes display optimal character- reactivity of lymphocytes obtained from three anatomic compart-
ments in patients with non-small-cell lung cancer (NSCLC).
istics for adoptive cell therapy. J Immunother 2008 Sep
Cancer Biother Radiopharm 2003 Oct;18(5):735–49.
5;31:742–751. 85. Dreno B, Nguyen JM, Khammari A, Pandolfino MC, Tessier MH,
69. Gohara R, Nakao M, Ogata Y, Isomoto H, Oizumi K, Itoh K. Bercegeay S, et al. Randomized trial of adoptive transfer of mela-
Histocompatibility leukocyte antigen-A2402-restricted cytotoxic T noma tumor-infiltrating lymphocytes as adjuvant therapy for stage
lymphocytes recognizing adenocarcinoma in tumor-infiltrating III melanoma. Cancer Immunol Immunother 2002
lymphocytes of patients with colon cancer. Jpn J Cancer Res 1997 Nov;51(10):539–46.
Feb;88(2):198–204. 86. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two sub-
70. Hom SS, Rosenberg SA, Topalian SL. Specific immune recogni- sets of memory T lymphocytes with distinct homing potentials and
tion of autologous tumor by lymphocytes infiltrating colon carci- effector functions. Nature 1999 Oct 14;401(6754):708–12.
nomas: analysis by cytokine secretion. Cancer Immunol
87. Geginat J, Lanzavecchia A, Sallusto F, Proliferation and differen-
Immunother 1993;36(1):1–8.
71. Toh U, Yamana H, Nakao M, Imai Y, Seki N, Takasu H, et al. HLA tiation potential of human CD8+ memory T-cell subsets in response
class I-restricted and tumor-specific cytotoxic T lymphocytes from to antigen or homeostatic cytokines. Blood 2003 Feb
metastatic lymph nodes of esophageal cancers. Cell Immunol 1997 6;101:4260–4266.
May 1;177(2):137–43. 88. Geginat J, Sallusto F, Lanzavecchia A. Cytokine-driven proliferation
72. Schwartzentruber DJ, Solomon D, Rosenberg SA, Topalian SL. and differentiation of human naive, central memory, and effector
Characterization of lymphocytes infiltrating human breast cancer: memory CD4(+) T cells. J Exp Med 2001 Dec 17;194(12):1711–9.
specific immune reactivity detected by measuring cytokine secre- 89. Young MR, Wright MA, Lozano Y, Matthews JP, Benefield J,
tion. J Immunother 1992 Jul;12(1):1–12. Prechel MM. Mechanisms of immune suppression in patients with
73. Linehan DC, Goedegebuure PS, Peoples GE, Rogers SO, Eberlein head and neck cancer: influence on the immune infiltrate of the
TJ. Tumor-specific and HLA-A2-restricted cytolysis by tumor- cancer. Int J Cancer 1996 Jul 29;67(3):333–8.
associated lymphocytes in human metastatic breast cancer. J 90. Spellman JE, Gollnick SO, Zhang PJ, Tomasi TB. Cytokine
Immunol 1995 Nov 1;155(9):4486–91. production by human soft tissue sarcomas: implications for
74. Baxevanis CN, Dedoussis GV, Papadopoulos NG, Missitzis I, immunosuppression within the tumour bed. Surg Oncol 1996
Stathopoulos GP, Papamichail M. Tumor specific cytolysis by Oct–Dec;5(5–6):237–44.
tumor infiltrating lymphocytes in breast cancer. Cancer 1994 Aug 91. Avradopoulos K, Mehta S, Blackinton D, Wanebo HJ. Interleukin-10
15;74(4):1275–82. as a possible mediator of immunosuppressive effect in patients
75. Dadmarz R, Sgagias MK, Rosenberg SA, Schwartzentruber DJ. with squamous cell carcinoma of the head and neck. Ann Surg
CD4+ T lymphocytes infiltrating human breast cancer recognise Oncol 1997 Mar;4(2):184–90.
autologous tumor in an MHC-class-II restricted fashion. Cancer 92. Hishii M, Nitta T, Ishida H, Ebato M, Kurosu A, Yagita H, et al.
Immunol Immunother 1995 Jan;40(1):1–9. Human glioma-derived interleukin-10 inhibits antitumor immune
76. Schultze JL, Seamon MJ, Michalak S, Gribben JG, Nadler LM. responses in vitro. Neurosurgery 1995 Dec;37(6):1160–6; discus-
Autologous tumor infiltrating T cells cytotoxic for follicular sion 6–7.
lymphoma cells can be expanded in vitro. Blood 1997 May 93. Armstrong L, Jordan N, Millar A. Interleukin 10 (IL-10) regulation
15;89(10):3806–16. of tumour necrosis factor alpha (TNF-alpha) from human alveolar
77. Freedman RS, Platsoucas CD. Immunotherapy for peritoneal ovar- macrophages and peripheral blood monocytes. Thorax 1996
ian carcinoma metastasis using ex vivo expanded tumor infiltrating Feb;51(2):143–9.
lymphocytes. Cancer Treat Res 1996;82:115–46. 94. Dix AR, Brooks WH, Roszman TL, Morford LA. Immune defects
78. Apiranthitou-Drogari M, Paganin C, Bernasconi S, Losa G, Maneo observed in patients with primary malignant brain tumors. J
A, Colombo N, et al. In search of specific cytotoxic T lymphocytes Neuroimmunol 1999 Dec;100(1–2):216–32.
infiltrating or accompanying human ovarian carcinoma. Cancer 95. Steinbrink K, Graulich E, Kubsch S, Knop J, Enk AH. CD4(+) and
Immunol Immunother 1992;35(4):289–95. CD8(+) anergic T cells induced by interleukin-10-treated human
John R. Yannelli 521

dendritic cells display antigen-specific suppressor activity. Blood cDNA for the gene therapy of cancer in humans. J Immunol 1993
2002 Apr 1;99(7):2468–76. May 1;150(9):4104–15.
96. Crowley NJ, Darrow TL, Seigler HF. Generation of autologous 112. Treisman J, Hwu P, Yannelli JR, Shafer GE, Cowherd R, Samid
tumor-specific cytotoxic T-cells using HLA-A region matched D, et al. Upregulation of tumor necrosis factor-alpha produc-
allogeneic melanoma. Curr Surg 1989 Sep–Oct;46(5):393–6. tion by retrovirally transduced human tumor-infiltrating
97. Crowley NJ, Slingluff CL, Jr., Darrow TL, Seigler HF. Generation lymphocytes using trans-retinoic acid. Cell Immunol 1994
of human autologous melanoma-specific cytotoxic T-cells using Jul;156(2):448–57.
HLA-A2-matched allogeneic melanomas. Cancer Res 1990 Feb 113. Rosenberg SA, Anderson WF, Blaese M, Hwu P, Yannelli JR,
1;50(3):492–8. Yang JC, et al. The development of gene therapy for the treatment
98. Crowley NJ, Darrow TL, Quinn-Allen MA, Seigler HF. MHC- of cancer. Ann Surg 1993 Oct;218(4):455–63; discussion 63–4.
restricted recognition of autologous melanoma by tumor-specific 114. van den Eynde B. [Identification of cancer antigens of relevance
cytotoxic T cells. Evidence for restriction by a dominant HLA-A for specific cancer immunotherapy]. Bull Mem Acad R Med Belg
allele. J Immunol 1991 Mar 1;146(5):1692–9. 2001;156(10–12):548–55.
99. Stevens EJ, Jacknin L, Robbins PF, Kawakami Y, el Gamil M, 115. Van den Eynde BJ, Boon T. Tumor antigens recognized by T
Rosenberg SA, et al. Generation of tumor-specific CTLs from lymphocytes. Int J Clin Lab Res 1997;27(2):81–6.
melanoma patients by using peripheral blood stimulated with 116. van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen
allogeneic melanoma tumor cell lines. Fine specificity and E, Van den Eynde B, et al. A gene encoding an antigen recognized
MART-1 melanoma antigen recognition. J Immunol 1995 Jan by cytolytic T lymphocytes on a human melanoma. Science 1991
15;154(2):762–71. Dec 13;254(5038):1643–7.
100. Celis E, Tsai V, Crimi C, DeMars R, Wentworth PA, Chesnut 117. Coulie PG, Weynants P, Lehmann F, Herman J, Brichard V,
RW, et al. Induction of anti-tumor cytotoxic T lymphocytes in Wolfel T, et al. Genes coding for tumor antigens recognized by
normal humans using primary cultures and synthetic peptide human cytolytic T lymphocytes. J Immunother 1993
epitopes. Proc Natl Acad Sci U S A 1994 Mar 15;91(6):2105–9. Aug;14(2):104–9.
101. Sule-Suso J, Arienti F, Melani C, Colombo MP, Parmiani G. A 118. Noppen C, Levy F, Burri L, Zajac P, Remmel E, Schaefer C, et al.
B7–1-transfected human melanoma line stimulates proliferation Naturally processed and concealed HLA-A2.1-restricted epitopes
and cytotoxicity of autologous and allogeneic lymphocytes. Eur from tumor-associated antigen tyrosinase-related protein-2. Int
J Immunol 1995 Oct;25(10):2737–42. J Cancer 2000 Jul 15;87(2):241–6.
102. Bixby DL, Yannelli JR. CD80 expression in an HLA-A2-positive 119. Dannull J, Diener PA, Prikler L, Furstenberger G, Cerny T,
human non-small cell lung cancer cell line enhances tumor- Schmid U, et al. Prostate stem cell antigen is a promising candi-
specific cytotoxicity of HLA-A2-positive T cells derived from a date for immunotherapy of advanced prostate cancer. Cancer Res
normal donor and a patient with non-small cell lung cancer. Int J 2000 Oct 1;60(19):5522–8.
Cancer 1998 Dec 9;78(6):685–94. 120. Arai J, Yasukawa M, Ohminami H, Kakimoto M, Hasegawa A,
103. Bain C, Merrouche Y, Puisieux I, Duc A, Colombo MP, Favrot Fujita S. Identification of human telomerase reverse tran-
M. B7.1 gene transduction of human renal-cell-carcinoma cell scriptase-derived peptides that induce HLA-A24-restricted
lines restores the proliferative response and cytotoxic function of antileukemia cytotoxic T lymphocytes. Blood 2001 May
allogeneic T cells. Int J Cancer 1996 Sep 17;67(6):769–76. 1;97(9):2903–7.
104. Schendel DJ, Frankenberger B, Jantzer P, Cayeux S, Nobetaner 121. Kao H, Marto JA, Hoffmann TK, Shabanowitz J, Finkelstein SD,
E, Willimsky G, et al. Expression of B7.1 (CD80) in a renal cell Whiteside TL, et al. Identification of cyclin B1 as a shared human
carcinoma line allows expansion of tumor-associated cytotoxic T epithelial tumor-associated antigen recognized by T cells. J Exp
lymphocytes in the presence of an alloresponse. Gene Ther 2000 Med 2001 Nov 5;194(9):1313–23.
Dec;7(23):2007–14. 122. Vonderheide RH, Anderson KS, Hahn WC, Butler MO, Schultze
105. Wang YC, Zhu L, McHugh R, Graham SD, Jr., Hillyer CD, JL, Nadler LM. Characterization of HLA-A3-restricted cytotoxic
Dillehay D, et al. Induction of autologous tumor-specific cyto- T lymphocytes reactive against the widely expressed tumor anti-
toxic T-lymphocyte activity against a human renal carcinoma cell gen telomerase. Clin Cancer Res 2001 Nov;7(11):3343–8.
line by B7–1 (CD8O) costimulation. J Immunother Emphasis 123. Schroers R, Huang XF, Hammer J, Zhang J, Chen SY.
Tumor Immunol 1996 Jan;19(1):1–8. Identification of HLA DR7-restricted epitopes from human
106. Heemskerk B, Liu K, Dudley ME, Johnson LA, Kaiser A, telomerase reverse transcriptase recognized by CD4+ T-helper
Downey S, et al. Adoptive cell therapy for patients with cells. Cancer Res 2002 May 1;62(9):2600–5.
melanoma, using tumor-infiltrating lymphocytes genetically 124. Engelhard VH, Appella E, Benjamin DC, Bodnar WM, Cox AL,
engineered to secrete interleukin-2. Hum Gene Ther 2008 Chen Y, et al. Mass spectrometric analysis of peptides associated
May;19(5):496–510. with the human class I MHC molecules HLA-A2.1 and HLA-B7
107. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. and identification of structural features that determine binding.
Adoptive cell transfer: a clinical path to effective cancer immu- Chem Immunol 1993;57:39–62.
notherapy. Nat Rev Cancer 2008 Apr;8(4):299–308. 125. Slingluff CL, Jr., Cox AL, Henderson RA, Hunt DF, Engelhard
108. Johnson LA, Heemskerk B, Powell DJ, Jr., Cohen CJ, Morgan VH. Recognition of human melanoma cells by HLA-A2.1-
RA, Dudley ME, et al. Gene transfer of tumor-reactive TCR restricted cytotoxic T lymphocytes is mediated by at least six
confers both high avidity and tumor reactivity to nonreactive shared peptide epitopes. J Immunol 1993 Apr 1;150(7):2955–63.
peripheral blood mononuclear cells and tumor-infiltrating 126. Slingluff CL, Jr., Cox AL, Stover JM, Jr., Moore MM, Hunt DF,
lymphocytes. J Immunol 2006 Nov 1;177(9):6548–59. Engelhard VH. Cytotoxic T-lymphocyte response to autologous
109. Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, human squamous cell cancer of the lung: epitope reconstitution
Sherry RM, et al. Cancer regression in patients after transfer of with peptides extracted from HLA-Aw68. Cancer Res 1994 May
genetically engineered lymphocytes. Science 2006 Oct 15;54(10):2731–7.
6;314(5796):126–9. 127. Skipper JC, Gulden PH, Hendrickson RC, Harthun N, Caldwell
110. Hughes MS, Yu YY, Dudley ME, Zheng Z, Robbins PF, Li Y, et JA, Shabanowitz J, et al. Mass-spectrometric evaluation of HLA-
al. Transfer of a TCR gene derived from a patient with a marked A*0201-associated peptides identifies dominant naturally pro-
antitumor response conveys highly active T-cell effector func- cessed forms of CTL epitopes from MART-1 and gp100. Int J
tions. Hum Gene Ther 2005 Apr;16(4):457–72. Cancer 1999 Aug 27;82(5):669–77.
111. Hwu P, Yannelli J, Kriegler M, Anderson WF, Perez C, Chiang Y, 128. Park S, Lim Y, Lee D, Cho B, Bang YJ, Sung S, et al. Identification
et al. Functional and molecular characterization of tumor-infil- and characterization of a novel cancer/testis antigen gene CAGE-
trating lymphocytes transduced with tumor necrosis factor-alpha 1. Biochim Biophys Acta 2003 Jan 27;1625(2):173–82.
522 Cellular immunotherapy (CI), where have we been and where are we going?

129. Nakatsura T, Senju S, Ito M, Nishimura Y, Itoh K. Cellular and 146. Wolfel T, Hauer M, Schneider J, Serrano M, Wolfel C, Klehmann-
humoral immune responses to a human pancreatic cancer anti- Hieb E, et al. A p16INK4a-insensitive CDK4 mutant targeted by
gen, coactosin-like protein, originally defined by the SEREX cytolytic T lymphocytes in a human melanoma. Science 1995
method. Eur J Immunol 2002 Mar;32(3):826–36. Sep 1;269(5228):1281–4.
130. Jager E, Chen YT, Drijfhout JW, Karbach J, Ringhoffer M, Jager 147. Mandruzzato S, Brasseur F, Andry G, Boon T, van der Bruggen
D, et al. Simultaneous humoral and cellular immune response P. A CASP-8 mutation recognized by cytolytic T lymphocytes on
against cancer-testis antigen NY-ESO-1: definition of human his- a human head and neck carcinoma. J Exp Med 1997 Aug
tocompatibility leukocyte antigen (HLA)-A2-binding peptide 29;186(5):785–93.
epitopes. J Exp Med 1998 Jan 19;187(2):265–70. 148. Gueguen M, Patard JJ, Gaugler B, Brasseur F, Renauld JC,
131. Levy F, Burri L, Morel S, Peitrequin AL, Levy N, Bachi A, et al. The Van Cangh PJ, et al. An antigen recognized by autologous
final N-terminal trimming of a subaminoterminal proline-containing CTLs on a human bladder carcinoma. J Immunol 1998 Jun
HLA class I-restricted antigenic peptide in the cytosol is mediated by 15;160(12):6188–94.
two peptidases. J Immunol 2002 Oct 15;169(8):4161–71. 149. Brossart P, Heinrich KS, Stuhler G, Behnke L, Reichardt VL,
132. Le Naour F. Contribution of proteomics to tumor immunology. Stevanovic S, et al. Identification of HLA-A2-restricted T-cell
Proteomics 2001 Oct;1(10):1295–302. epitopes derived from the MUC1 tumor antigen for broadly appli-
133. Wang T, Hopkins D, Schmidt C, Silva S, Houghton R, Takita H, cable vaccine therapies. Blood 1999 Jun 15;93(12):4309–17.
et al. Identification of genes differentially over-expressed in lung 150. Brossart P, Wirths S, Stuhler G, Reichardt VL, Kanz L, Brugger
squamous cell carcinoma using combination of cDNA subtraction W. Induction of cytotoxic T-lymphocyte responses in vivo after
and microarray analysis. Oncogene 2000 Mar 16;19(12):1519–28. vaccinations with peptide-pulsed dendritic cells. Blood 2000
134. Mathiassen S, Lauemoller SL, Ruhwald M, Claesson MH, Buus Nov 1;96(9):3102–8.
S. Tumor-associated antigens identified by mRNA expression 151. Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton JM,
profiling induce protective anti-tumor immunity. Eur J Immunol Schlom J. Generation of human cytotoxic T cells specific for
2001 Apr;31(4):1239–46. human carcinoembryonic antigen epitopes from patients immu-
135. Brichard V, Van Pel A, Wolfel T, Wolfel C, De Plaen E, Lethe B, nized with recombinant vaccinia-CEA vaccine. J Natl Cancer
et al. The tyrosinase gene codes for an antigen recognized by Inst 1995 Jul 5;87(13):982–90.
autologous cytolytic T lymphocytes on HLA-A2 melanomas. J 152. Ropke M, Hald J, Guldberg P, Zeuthen J, Norgaard L, Fugger L,
Exp Med 1993 Aug 1;178(2):489–95. et al. Spontaneous human squamous cell carcinomas are killed by
136. Coulie PG, Brichard V, Van Pel A, Wolfel T, Schneider J, a human cytotoxic T lymphocyte clone recognizing a wild-type
Traversari C, et al. A new gene coding for a differentiation anti- p53-derived peptide. Proc Natl Acad Sci U S A 1996 Dec
gen recognized by autologous cytolytic T lymphocytes on HLA- 10;93(25):14704–7.
A2 melanomas. J Exp Med 1994 Jul 1;180(1):35–42. 153. Fisk B, Savary C, Hudson JM, O’Brian CA, Murray JL, Wharton
137. Robbins PF, El-Gamil M, Li YF, Kawakami Y, Loftus D, Appella JT, et al. Changes in an HER-2 peptide upregulating HLA-A2
E, et al. A mutated beta-catenin gene encodes a melanoma-spe- expression affect both conformational epitopes and CTL recogni-
cific antigen recognized by tumor infiltrating lymphocytes. J Exp tion: implications for optimization of antigen presentation and
Med 1996 Mar 1;183(3):1185–92. tumor-specific CTL induction. J Immunother Emphasis Tumor
138. Visseren MJ, van der Burg SH, van der Voort EI, Brandt RM, Immunol 1995 Nov;18(4):197–209.
Schrier PI, van der Bruggen P, et al. Identification of HLA- 154. Fisk B, Blevins TL, Wharton JT, Ioannides CG. Identification of
A*0201-restricted CTL epitopes encoded by the tumor-specific an immunodominant peptide of HER-2/neu protooncogene rec-
MAGE-2 gene product. Int J Cancer 1997 Sep 26;73(1):125–30. ognized by ovarian tumor-specific cytotoxic T lymphocyte lines.
139. Gaugler B, Van den Eynde B, van der Bruggen P, Romero P, J Exp Med 1995 Jun 1;181(6):2109–17.
Gaforio JJ, De Plaen E, et al. Human gene MAGE-3 codes for an 155. Shichijo S, Nakao M, Imai Y, Takasu H, Kawamoto M, Niiya F,
antigen recognized on a melanoma by autologous cytolytic T et al. A gene encoding antigenic peptides of human squamous cell
lymphocytes. J Exp Med 1994 Mar 1;179(3):921–30. carcinoma recognized by cytotoxic T lymphocytes. J Exp Med
140. Boel P, Wildmann C, Sensi ML, Brasseur R, Renauld JC, Coulie 1998 Feb 2;187(3):277–88.
P, et al. BAGE: a new gene encoding an antigen recognized on 156. Nishizaka S, Gomi S, Harada K, Oizumi K, Itoh K, Shichijo S. A
human melanomas by cytolytic T lymphocytes. Immunity 1995 new tumor-rejection antigen recognized by cytotoxic T lympho-
Feb;2(2):167–75. cytes infiltrating into a lung adenocarcinoma. Cancer Res 2000
141. Panelli MC, Bettinotti MP, Lally K, Ohnmacht GA, Li Y, Robbins Sep 1;60(17):4830–7.
P, et al. A tumor-infiltrating lymphocyte from a melanoma metas- 157. Kawano K, Gomi S, Tanaka K, Tsuda N, Kamura T, Itoh K,
tasis with decreased expression of melanoma differentiation anti- et al. Identification of a new endoplasmic reticulum-resident
gens recognizes MAGE-12. J Immunol 2000 Apr protein recognized by HLA-A24-restricted tumor-infiltrating
15;164(8):4382–92. lymphocytes of lung cancer. Cancer Res 2000 Jul 1;60(13):
142. Van den Eynde B, Peeters O, De Backer O, Gaugler B, Lucas S, 3550–8.
Boon T. A new family of genes coding for an antigen recognized 158. Fauroux CM, Lee M, Cullis PM, Douglas KT, Gore MG, Freeman
by autologous cytolytic T lymphocytes on a human melanoma. J S. Stereochemistry at phosphorus of the reaction catalyzed by
Exp Med 1995 Sep 1;182(3):689–98. myo-inositol monophosphatase. J Med Chem 2002 Mar
143. Kang X, Kawakami Y, el-Gamil M, Wang R, Sakaguchi K, 14;45(6):1363–73.
Yannelli JR, et al. Identification of a tyrosinase epitope recog- 159. Yoshikawa T, Padigaru M, Karkera JD, Sharma M, Berrettini
nized by HLA-A24-restricted, tumor-infiltrating lymphocytes. J WH, Esterling LE, et al. Genomic structure and novel variants of
Immunol 1995 Aug 1;155(3):1343–8. myo-inositol monophosphatase 2 (IMPA2). Mol Psychiatry 2000
144. Salazar-Onfray F, Nakazawa T, Chhajlani V, Petersson M, Karre Mar;5(2):165–71.
K, Masucci G, et al. Synthetic peptides derived from the melano- 160. Rao VV, Schnittger S, Hansmann I. G protein Gs alpha (GNAS
cyte-stimulating hormone receptor MC1R can stimulate HLA- 1), the probable candidate gene for Albright hereditary osteo-
A2-restricted cytotoxic T lymphocytes that recognize naturally dystrophy, is assigned to human chromosome 20q12-q13.2.
processed peptides on human melanoma cells. Cancer Res 1997 Genomics 1991 May;10(1):257–61.
Oct 1;57(19):4348–55. 161. Wroblewski JM, Yannelli JR. Identification of HLA-CW3,
145. Chiari R, Foury F, De Plaen E, Baurain JF, Thonnard J, Coulie PG. GNAS and IMPA as cytotoxic T-lymphocyte (CTL) target anti-
Two antigens recognized by autologous cytolytic T lymphocytes gens using an allogeneic mixed lymphocyte tumor cell culture
on a melanoma result from a single point mutation in an essential (MLTC) system and subsequent cDNA library screening. Cancer
housekeeping gene. Cancer Res 1999 Nov 15;59(22):5785–92. Biother Radiopharm 2007 Apr;22(2):206–22.
John R. Yannelli 523

162. Hemmila MR, Chang AE. Clinical implications of the new biol- notherapy: correlation of survival with skin testing by autologous
ogy in the development of melanoma vaccines. J Surg Oncol tumor cells. Cancer Immunol Immunother 1990;32(1):62–6.
1999 Apr;70(4):263–74. 185. Wroblewski JM, Bixby DL, Borowski C, Yannelli JR. Characteri-
163. Moingeon P. Cancer vaccines. Vaccine 2001 Jan 8;19(11–12): zation of human non-small cell lung cancer (NSCLC) cell lines for
1305–26. expression of MHC, co-stimulatory molecules and tumor-associ-
164. Jager E, Jager D, Knuth A. Clinical cancer vaccine trials. Curr ated antigens. Lung Cancer 2001 Aug–Sep;33(2–3):181–94.
Opin Immunol 2002 Apr;14(2):178–82. 186. Morton DL, Foshag LJ, Hoon DS, Nizze JA, Famatiga E, Wanek
165. Apostolopoulos V, Weiner DB, Gong J. Cancer vaccines: methods LA, et al. Prolongation of survival in metastatic melanoma after
for inducing immunity. Expert Rev Vaccines 2008 Sep;7(7):861–2. active specific immunotherapy with a new polyvalent melanoma
166. Chekhun VF. Cancer vaccines. Exp Oncol 2008 Jun;30(2):90. vaccine. Ann Surg 1992 Oct;216(4):463–82.
167. Emens LA. Cancer vaccines: on the threshold of success. Expert 187. Morton DL, Hoon DS, Nizze JA, Foshag LJ, Famatiga E, Wanek
Opin Emerg Drugs 2008 Jun;13(2):295–308. LA, et al. Polyvalent melanoma vaccine improves survival of
168. de Gruijl TD, van den Eertwegh AJ, Pinedo HM, Scheper RJ. patients with metastatic melanoma. Ann N Y Acad Sci 1993 Aug
Whole-cell cancer vaccination: from autologous to allogeneic 12;690:120–34.
tumor- and dendritic cell-based vaccines. Cancer Immunol 188. Elliott GT, McLeod RA, Perez J, Von Eschen KB. Interim results
Immunother 2008 Oct;57(10):1569–77. of a phase II multicenter clinical trial evaluating the activity of a
169. Reichert JM, Wenger JB. Development trends for new cancer thera- therapeutic allogeneic melanoma vaccine (theraccine) in the
peutics and vaccines. Drug Discov Today 2008 Jan;13(1–2):30–7. treatment of disseminated malignant melanoma. Semin Surg
170. Bodey B, Bodey B, Jr., Siegel SE, Kaiser HE. Failure of cancer Oncol 1993 May–Jun;9(3):264–72.
vaccines: the significant limitations of this approach to immuno- 189. Mitchell MS. Combinations of anticancer drugs and immunother-
therapy. Anticancer Res 2000 Jul–Aug;20(4):2665–76. apy. Cancer Immunol Immunother 2003 Nov;52(11):686–92.
171. Razzaque A, Dye E, Puri RK. Characterization of tumor vaccines 190. Sondak VK, Sosman JA. Results of clinical trials with an allo-
during product development. Vaccine 2000 Nov 8;19(6):644–7. genic melanoma tumor cell lysate vaccine: Melacine. Semin
172. Restifo NP, Esquivel F, Kawakami Y, Yewdell JW, Mule JJ, Cancer Biol 2003 Dec;13(6):409–15.
Rosenberg SA, et al. Identification of human cancers deficient in 191. Chan AD, Morton DL. Active immunotherapy with allogeneic
antigen processing. J Exp Med 1993 Feb 1;177(2):265–72. tumor cell vaccines: present status. Semin Oncol 1998
173. Restifo NP, Kawakami Y, Marincola F, Shamamian P, Taggarse Dec;25(6):611–22.
A, Esquivel F, et al. Molecular mechanisms used by tumors to 192. Newton DA, Acierno PM, Brescia FJ, Brown EA, Gattoni-Celli
escape immune recognition: immunogenetherapy and the cell S. Semi-allogeneic vaccines for patients with cancer and AIDS. J
biology of major histocompatibility complex class I. J Immunother Immunother 2002 Jul–Aug;25(4):334–41.
1993 Oct;14(3):182–90. 193. Rosenberg SA. Gene therapy for cancer. Jama 1992 Nov
174. Restifo NP, Marincola FM, Kawakami Y, Taubenberger J, 4;268(17):2416–9.
Yannelli JR, Rosenberg SA. Loss of functional beta 2-micro- 194. Nelson WG, Simons JW, Mikhak B, Chang JF, DeMarzo AM,
globulin in metastatic melanomas from five patients receiving Carducci MA, et al. Cancer cells engineered to secrete granulo-
immunotherapy. J Natl Cancer Inst 1996 Jan 17;88(2):100–8. cyte-macrophage colony-stimulating factor using ex vivo gene
175. Lewko WM, Ladd P, Hubbard D, He YJ, Vaghmar R, Husseini S, transfer as vaccines for the treatment of genitourinary malignan-
et al. Tumor acquisition, propagation, and preservation. The culture cies. Cancer Chemother Pharmacol 2000;46 Suppl:S67–72.
of human colorectal cancer. Cancer 1989 Oct 15;64(8):1600–7. 195. Tani K, Nakazaki Y, Hase H, Takahashi K, Azuma M, Ohata J,
176. Lewko WM, Vaghmar R, Hubbard D, Moore M, He YJ, Chang L, et al. Progress reports on immune gene therapy for stage IV renal
et al. Cultured cell lines from human breast cancer biopsies and cell cancer using lethally irradiated granulocyte-macrophage
xenografts. Breast Cancer Res Treat 1990 Dec;17(2):121–9. colony-stimulating factor-transduced autologous renal cancer
177. Pass HI, Stevens EJ, Oie H, Tsokos MG, Abati AD, Fetsch PA, cells. Cancer Chemother Pharmacol 2000;46(Suppl):S73–6.
et al. Characteristics of nine newly derived mesothelioma cell 196. Qian HN, Liu GZ, Cao SJ, Feng J, Ye X. The experimental
lines. Ann Thorac Surg 1995 Apr;59(4):835–44. study of ovarian carcinoma vaccine modified by human B7–1
178. Oie HK, Russell EK, Carney DN, Gazdar AF. Cell culture meth- and IFN-gamma genes. Int J Gynecol Cancer 2002 Jan–Feb;
ods for the establishment of the NCI series of lung cancer cell 12(1):80–5.
lines. J Cell Biochem Suppl 1996;24:24–31. 197. O’Rourke MG, Schmidt CW, O’Rourke TR, Ellem KA.
179. Sgagias MK, Nieroda C, Yannelli JR, Cowan KH, Danforth Immunotherapy, including gene therapy, for metastatic mela-
DN, Jr. Upregulation of DF3, in association with ICAM-1 and noma. Aust N Z J Surg 1997 Dec;67(12):834–41.
MHC class II by IFN-gamma in short-term human mammary 198. Schadendorf D, Paschen A, Sun Y. Autologous, allogeneic tumor
carcinoma cell cultures. Cancer Biother Radiopharm 1996 cells or genetically engineered cells as cancer vaccine against
Jun;11(3):177–85. melanoma. Immunol Lett 2000 Sep 15;74(1):67–74.
180. Lahn M, Kohler G, Schmoor C, Dengler W, Veelken H, 199. Antonia SJ, Seigne J, Diaz J, Muro-Cacho C, Extermann M,
Brennscheidt U, et al. Processing of tumor tissues for vaccination Farmelo MJ, et al. Phase I trial of a B7–1 (CD80) gene modified
with autologous tumor cells. Eur Surg Res 1997;29(4):292–302. autologous tumor cell vaccine in combination with systemic
181. Dillman RO, Beutel LD, Barth NM, de Leon C, O’Connor AA, interleukin-2 in patients with metastatic renal cell carcinoma. J
DePriest C, et al. Irradiated cells from autologous tumor cell lines Urol 2002 May;167(5):1995–2000.
as patient-specific vaccine therapy in 125 patients with metastatic 200. Arienti F, Sule-Suso J, Belli F, Mascheroni L, Rivoltini L, Melani
cancer: induction of delayed-type hypersensitivity to autologous C, et al. Limited antitumor T cell response in melanoma patients
tumor is associated with improved survival. Cancer Biother vaccinated with interleukin-2 gene-transduced allogeneic mela-
Radiopharm 2002 Feb;17(1):51–66. noma cells. Hum Gene Ther 1996 Oct 20;7(16):1955–63.
182. Berd D, Maguire HC, Jr., McCue P, Mastrangelo MJ. Treatment 201. Belli F, Arienti F, Sule-Suso J, Clemente C, Mascheroni L,
of metastatic melanoma with an autologous tumor-cell vaccine: Cattelan A, et al. Active immunization of metastatic melanoma
clinical and immunologic results in 64 patients. J Clin Oncol patients with interleukin-2-transduced allogeneic melanoma
1990 Nov;8(11):1858–67. cells: evaluation of efficacy and tolerability. Cancer Immunol
183. Berd D, Kairys J, Dunton C, Mastrangelo MJ, Sato T, Maguire Immunother 1997 Jun;44(4):197–203.
HC, Jr. Autologous, hapten-modified vaccine as a treatment for 202. Haight AE, Bowman LC, Ng CY, Vanin EF, Davidoff AM.
human cancers. Semin Oncol 1998 Dec;25(6):646–53. Humoral response to vaccination with interleukin-2-expressing
184. McCune CS, O’Donnell RW, Marquis DM, Sahasrabudhe DM. allogeneic neuroblastoma cells after primary therapy. Med
Renal cell carcinoma treated by vaccines for active specific immu- Pediatr Oncol 2000 Dec;35(6):712–5.
524 Cellular immunotherapy (CI), where have we been and where are we going?

203. Dranoff G, Jaffee E, Lazenby A, Golumbek P, Levitsky H, Brose 218. Steinman RM, Dhodapkar M. Active immunization against can-
K, et al. Vaccination with irradiated tumor cells engineered to cer with dendritic cells: the near future. Int J Cancer 2001
secrete murine granulocyte-macrophage colony-stimulating factor Nov15;94(4):459–73.
stimulates potent, specific, and long-lasting anti-tumor immunity. 219. Ludewig B, Ochsenbein AF, Odermatt B, Paulin D, Hengartner H,
Proc Natl Acad Sci U S A 1993 Apr 15;90(8):3539–43. Zinkernagel RM. Immunotherapy with dendritic cells directed
204. Arellano M, E KW. Granulocyte-macrophage-colony-stimulating against tumor antigens shared with normal host cells results in severe
factor and other cytokines: as adjuncts to cancer immunotherapy, autoimmune disease. J Exp Med 2000 Mar 6;191(5):795–804.
stem cell transplantation, and vaccines. Curr Hematol Rep 2004 220. Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B,
Nov;3(6):424–31. et al. Vaccination of patients with B-cell lymphoma using
205. Powell A, Creaney J, Broomfield S, Van Bruggen I, Robinson B. autologous antigen-pulsed dendritic cells. Nat Med 1996
Recombinant GM-CSF plus autologous tumor cells as a Jan;2(1):52–8.
vaccine for patients with mesothelioma. Lung Cancer 2006 221. Timmerman JM, Czerwinski DK, Davis TA, Hsu FJ, Benike C,
May;52(2):189–97. Hao ZM, et al. Idiotype-pulsed dendritic cell vaccination for
206. Leong SP, Enders-Zohr P, Zhou YM, Stuntebeck S, Habib FA, B-cell lymphoma: clinical and immune responses in 35 patients.
Allen RE, Jr., et al. Recombinant human granulocyte mac- Blood 2002 Mar 1;99(5):1517–26.
rophage-colony stimulating factor (rhGM-CSF) and autologous 222. Triozzi PL, Khurram R, Aldrich WA, Walker MJ, Kim JA,
melanoma vaccine mediate tumor regression in patients with Jaynes S. Intratumoral injection of dendritic cells derived in
metastatic melanoma. J Immunother 1999 Mar;22(2):166–74. vitro in patients with metastatic cancer. Cancer 2000 Dec
207. Murata S, Ladle BH, Kim PS, Lutz ER, Wolpoe ME, Ivie SE, et al. 15;89(12):2646–54.
OX40 costimulation synergizes with GM-CSF whole-cell vaccina- 223. Padley DJ, Dietz AB, Gastineau DA, Vuk-Pavlovic S. Mature
tion to overcome established CD8+ T cell tolerance to an endoge- myeloid dendritic cells for clinical use prepared from CD14+
nous tumor antigen. J Immunol 2006 Jan 15;176(2):974–83. cells isolated by immunomagnetic adsorption. J Hematother
208. Barrio MM, de Motta PT, Kaplan J, von Euw EM, Bravo AI, Stem Cell Res 2001 Jun;10(3):427–9.
Chacon RD, et al. A phase I study of an allogeneic cell vaccine 224. Rains N, Cannan RJ, Chen W, Stubbs RS. Development of a
(VACCIMEL) with GM-CSF in melanoma patients. J Immunother dendritic cell (DC)-based vaccine for patients with advanced
2006 Jul–Aug;29(4):444–54. colorectal cancer. Hepatogastroenterology 2001 Mar–Apr;48
209. Ullenhag GJ, Frodin JE, Jeddi-Tehrani M, Strigard K, Eriksson (38):347–51.
E, Samanci A, et al. Durable carcinoembryonic antigen (CEA)- 225. Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK, et
specific humoral and cellular immune responses in colorectal al. Vaccination of malignant glioma patients with peptide-pulsed
carcinoma patients vaccinated with recombinant CEA and granu- dendritic cells elicits systemic cytotoxicity and intracranial T-cell
locyte/macrophage colony-stimulating factor. Clin Cancer Res infiltration. Cancer Res 2001 Feb 1;61(3):842–7.
2004 May 15;10(10):3273–81. 226. Hernando JJ, Park TW, Kubler K, Offergeld R, Schlebusch H,
210. Nemunaitis J, Sterman D, Jablons D, Smith JW, 2nd, Fox B, Bauknecht T. Vaccination with autologous tumour antigen-pulsed
Maples P, et al. Granulocyte-macrophage colony-stimulating fac- dendritic cells in advanced gynaecological malignancies: clinical
tor gene-modified autologous tumor vaccines in non-small-cell and immunological evaluation of a phase I trial. Cancer Immunol
lung cancer. J Natl Cancer Inst 2004 Feb 18;96(4):326–31. Immunother 2002 Mar;51(1):45–52.
211. Nemunaitis J. GVAX (GMCSF gene modified tumor vaccine) in 227. Bachleitner-Hofmann T, Stift A, Friedl J, Pfragner R, Radelbauer
advanced stage non small cell lung cancer. J Control Release K, Dubsky P, et al. Stimulation of autologous antitumor T-cell
2003 Aug 28;91(1–2):225–31. responses against medullary thyroid carcinoma using tumor
212. Ellem KA, O’Rourke MG, Johnson GR, Parry G, Misko IS, lysate-pulsed dendritic cells. J Clin Endocrinol Metab 2002
Schmidt CW, et al. A case report: immune responses and clini- Mar;87(3):1098–104.
cal course of the first human use of granulocyte/macrophage- 228. Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, et
colony-stimulating-factor-transduced autologous melanoma al. Vaccination of melanoma patients with peptide- or tumor
cells for immunotherapy. Cancer Immunol Immunother 1997 lysate-pulsed dendritic cells. Nat Med 1998 Mar;4(3):328–32.
Mar;44(1):10–20. 229. Thurner B, Roder C, Dieckmann D, Heuer M, Kruse M, Glaser
213. Dranoff G, Soiffer R, Lynch T, Mihm M, Jung K, Kolesar K, et A, et al. Generation of large numbers of fully mature and stable
al. A phase I study of vaccination with autologous, irradiated dendritic cells from leukapheresis products for clinical applica-
melanoma cells engineered to secrete human granulocyte-mac- tion. J Immunol Methods 1999 Feb 1;223(1):1–15.
rophage colony stimulating factor. Hum Gene Ther 1997 Jan 230. Panelli MC, Wunderlich J, Jeffries J, Wang E, Mixon A, Rosenberg
1;8(1):111–23. SA, et al. Phase 1 study in patients with metastatic melanoma of
214. Soiffer R, Lynch T, Mihm M, Jung K, Rhuda C, Schmollinger JC, immunization with dendritic cells presenting epitopes derived
et al. Vaccination with irradiated autologous melanoma cells from the melanoma-associated antigens MART-1 and gp100. J
engineered to secrete human granulocyte-macrophage colony- Immunother 2000 Jul–Aug;23(4):487–98.
stimulating factor generates potent antitumor immunity in 231. Reichardt VL, Okada CY, Liso A, Benike CJ, Stockerl-Goldstein
patients with metastatic melanoma. Proc Natl Acad Sci U S A KE, Engleman EG, et al. Idiotype vaccination using dendritic
1998 Oct 27;95(22):13141–6. cells after autologous peripheral blood stem cell transplantation
215. Simons JW, Mikhak B, Chang JF, DeMarzo AM, Carducci MA, for multiple myeloma – a feasibility study. Blood 1999 Apr
Lim M, et al. Induction of immunity to prostate cancer antigens: 1;93(7):2411–9.
results of a clinical trial of vaccination with irradiated autologous 232. Yannelli JR, Sturgill J, Foody T, Hirschowitz E, The large scale
prostate tumor cells engineered to secrete granulocyte-mac- generation of dendritic cells for the immunization of patients
rophage colony-stimulating factor using ex vivo gene transfer. with non-small cell lung cancer (NSCLC). Lung Cancer
Cancer Res 1999 Oct 15;59(20):5160–8. 2005;47:337–350.
216. Simons JW. Bioactivity of human GM-CSF gene therapy in met- 233. Murphy G, Tjoa B, Ragde H, Kenny G, Boynton A. Phase I
astatic renal cell carcinoma and prostate cancer. Hinyokika Kiyo clinical trial: T-cell therapy for prostate cancer using autologous
1997 Nov;43(11):821–2. dendritic cells pulsed with HLA-A0201-specific peptides
217. Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, from prostate-specific membrane antigen. Prostate 1996 Dec;29
Carducci MA, et al. Bioactivity of autologous irradiated renal (6):371–80.
cell carcinoma vaccines generated by ex vivo granulocyte-mac- 234. Murphy GP, Tjoa BA, Simmons SJ, Jarisch J, Bowes VA, Ragde
rophage colony-stimulating factor gene transfer. Cancer Res H, et al. Infusion of dendritic cells pulsed with HLA-A2-specific
1997 Apr 15;57(8):1537–46. prostate-specific membrane antigen peptides: a phase II prostate
John R. Yannelli 525

cancer vaccine trial involving patients with hormone-refractory monocyte-derived dendritic cells for clinical use. Cancer
metastatic disease. Prostate 1999 Jan 1;38(1):73–8. Immunol Immunother 2001 Oct;50:417–427.
235. Murphy GP, Tjoa BA, Simmons SJ, Ragde H, Rogers M, Elgamal 254. Santegoets SJ, van den Eertwegh AJ, van de Loosdrecht AA,
A, et al. Phase II prostate cancer vaccine trial: report of a study Scheper RJ, de Gruijl TD. Human dendritic cell line models for
involving 37 patients with disease recurrence following primary DC differentiation and clinical DC vaccination studies. J Leukoc
treatment. Prostate 1999 Apr 1;39(1):54–9. Biol 2008 Jul 29.
236. Holtl L, Rieser C, Papesh C, Ramoner R, Herold M, Klocker H, 255. Fong L, Brockstedt D, Benike C, Breen JK, Strang G, Ruegg CL,
et al. Cellular and humoral immune responses in patients with et al. Dendritic cell-based xenoantigen vaccination for prostate
metastatic renal cell carcinoma after vaccination with antigen cancer immunotherapy. J Immunol 2001 Dec 15;167(12):7150–6.
pulsed dendritic cells. J Urol 1999 Mar;161(3):777–82. 256. Fong L, Brockstedt D, Benike C, Wu L, Engleman EG. Dendritic
237. Merad M, Angevin E, Wolfers J, Flament C, Lorenzi I, Triebel cells injected via different routes induce immunity in cancer
F, et al. Generation of monocyte-derived dendritic cells from patients. J Immunol 2001 Mar 15;166(6):4254–9.
patients with renal cell cancer: modulation of their functional 257. Tjoa BA, Erickson SJ, Bowes VA, Ragde H, Kenny GM, Cobb
properties after therapy with biological response modifiers OE, et al. Follow-up evaluation of prostate cancer patients
(IFN-alpha plus IL-2 and IL-12). J Immunother 2000 infused with autologous dendritic cells pulsed with PSMA pep-
May–Jun;23(3):369–78. tides. Prostate 1997 Sep 1;32(4):272–8.
238. Claxton DF, McMannis J, Champlin R, Choudhury A. Therapeutic 258. Tjoa BA, Simmons SJ, Bowes VA, Ragde H, Rogers M, Elgamal
potential of leukemia-derived dendritic cells: preclinical and A, et al. Evaluation of phase I/II clinical trials in prostate cancer
clinical progress. Crit Rev Immunol 2001;21(1–3):147–55. with dendritic cells and PSMA peptides. Prostate 1998 Jun
239. Nencioni A, Brossart P. New perspectives in dendritic cell-based 15;36(1):39–44.
cancer immunotherapy. BioDrugs 2001;15(10):667–79. 259. Tjoa BA, Murphy GP. Development of dendritic-cell based pros-
240. Steinman R, Hoffman L, Pope M. Maturation and migration of tate cancer vaccine. Immunol Lett 2000 Sep 15;74(1):87–93.
cutaneous dendritic cells. J Invest Dermatol 1995 Jul;105(1 260. Kugler A, Stuhler G, Walden P, Zoller G, Zobywalski A, Brossart
Suppl):2S–7S. P, et al. Regression of human metastatic renal cell carcinoma
241. Mosca PJ, Hobeika AC, Clay TM, Nair SK, Thomas EK, Morse after vaccination with tumor cell-dendritic cell hybrids. Nat Med
MA, et al. A subset of human monocyte-derived dendritic cells 2000 Mar;6(3):332–6.
expresses high levels of interleukin-12 in response to combined 261. Lau R, Wang F, Jeffery G, Marty V, Kuniyoshi J, Bade E, et al.
CD40 ligand and interferon-gamma treatment. Blood 2000 Nov Phase I trial of intravenous peptide-pulsed dendritic cells in
15;96(10):3499–504. patients with metastatic melanoma. J Immunother 2001
242. Hart DN. Dendritic cells: unique leukocyte populations which Jan–Feb;24(1):66–78.
control the primary immune response. Blood 1997 Nov 262. Banchereau J, Palucka AK, Dhodapkar M, Burkeholder S, Taquet
1;90(9):3245–87. N, Rolland A, et al. Immune and clinical responses in patients
243. Satthaporn S, Eremin O. Dendritic cells (II): Role and thera- with metastatic melanoma to CD34(+) progenitor-derived den-
peutic implications in cancer. J R Coll Surg Edinb 2001 dritic cell vaccine. Cancer Res 2001 Sep 1;61(17):6451–8.
Jun;46(3):159–67. 263. Hirschowitz EA, Foody T, Hidalgo GE, Yannelli JR, Immunization
244. Satthaporn S, Eremin O. Dendritic cells (I): Biological functions. of NSCLC patients with antigen-pulsed immature autologous
J R Coll Surg Edinb 2001 Feb;46(1):9–19. dendritic cells. Lung Cancer 2007 May 15;57:365–372.
245. Dhodapkar MV, Steinman RM, Krasovsky J, Munz C, Bhardwaj 264. Hirschowitz EA, Foody T, Kryscio R, Dickson L, Sturgill J,
N. Antigen-specific inhibition of effector T cell function in Yannelli J. Autologous dendritic cell vaccines for non-small-cell
humans after injection of immature dendritic cells. J Exp Med lung cancer. J Clin Oncol 2004 Jul 15;22(14):2808–15.
2001 Jan 15;193(2):233–8. 265. Jonuleit H, Schmitt E, Steinbrink K, Enk AH. Dendritic cells as a
246. Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller tool to induce anergic and regulatory T cells. Trends Immunol
WA. Differentiation of monocytes into dendritic cells in a 2001 Jul;22(7):394–400.
model of transendothelial trafficking. Science 1998 Oct 266. Sakaguchi S. Naturally Arising CD4+ Regulatory T Cells for
16;282(5388):480–3. Immunologic Self-Tolerance and Negative Control of Immune
247. Sallusto F, Lanzavecchia A. Efficient presentation of soluble Responses. Annu Rev Immunol 2004 Apr;22:531–62.
antigen by cultured human dendritic cells is maintained by granu- 267. Sakaguchi S. Naturally arising CD4+ regulatory t cells for immu-
locyte/macrophage colony-stimulating factor plus interleukin 4 nologic self-tolerance and negative control of immune responses.
and downregulated by tumor necrosis factor alpha. J Exp Med Annu Rev Immunol 2004;22:531–62.
1994 Apr 1;179(4):1109–18. 268. Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immu-
248. Bender A, Sapp M, Schuler G, Steinman RM, Bhardwaj N. nity by removing CD25+CD4+ T cells: a common basis between
Improved methods for the generation of dendritic cells from non- tumor immunity and autoimmunity. J Immunol 1999 Nov
proliferating progenitors in human blood. J Immunol Methods 15;163(10):5211–8.
1996 Sep 27;196(2):121–35. 269. Sakaguchi S, Sakaguchi N, Shimizu J, Yamazaki S, Sakihama T,
249. Zhou LJ, Tedder TF. Human blood dendritic cells selectively Itoh M, et al. Immunologic tolerance maintained by CD25+
express CD83, a member of the immunoglobulin superfamily. J CD4+ regulatory T cells: their common role in controlling auto-
Immunol 1995 Apr 15;154(8):3821–35. immunity, tumor immunity, and transplantation tolerance.
250. Romani N, Gruner S, Brang D, Kampgen E, Lenz A, Immunol Rev 2001 Aug;182:18–32.
Trockenbacher B, et al. Proliferating dendritic cell progenitors in 270. Hori S, Sakaguchi S. Foxp3: a critical regulator of the develop-
human blood. J Exp Med 1994 Jul 1;180(1):83–93. ment and function of regulatory T cells. Microbes Infect 2004
251. Kato K, Takaue Y, Wakasugi H. T-cell-conditioned medium effi- Jul;6(8):745–51.
ciently induces the maturation and function of human dendritic 271. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the
cells. J Leukoc Biol 2001 Dec;70(6):941–9. development and function of CD4+CD25+ regulatory T cells.
252. Ebner S, Ratzinger G, Krosbacher B, Schmuth M, Weiss A, Nat Immunol 2003 Apr;4(4):330–6.
Reider D, et al. Production of IL-12 by human monocyte-derived 272. Ramsdell F. Foxp3 and natural regulatory T cells: key to a cell
dendritic cells is optimal when the stimulus is given at the onset lineage? Immunity 2003 Aug;19(2):165–8.
of maturation, and is further enhanced by IL-4. J Immunol 2001 273. Zheng Y, Josefowicz SZ, Kas A, Chu TT, Gavin MA, Rudensky
Jan 1;166(1):633–41. AY, Genome-wide analysis of Foxp3 target genes in develop-
253. Spisek R, Bretaudeau L, Barbieux I, Meflah K, Gregoire M, ing and mature regulatory T cells. Nature 2007 Jan 21;445:
Standardized generation of fully mature p70 IL-12 secreting 936–940.
526 Cellular immunotherapy (CI), where have we been and where are we going?

274. Marson A, Kretschmer K, Frampton GM, Jacobsen ES, Polansky pancreas or breast adenocarcinoma. J Immunol 2002 Sep
JK, Macisaac KD, et al. Foxp3 occupancy and regulation of key 1;169(5):2756–61.
target genes during T-cell stimulation. Nature 2007 Jan 21. 283. Marshall NA, Christie LE, Munro LR, Culligan DJ, Johnston
275. Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck- PW, Barker RN, et al. Immunosuppressive regulatory T cells are
Loebenstein B. Increase of regulatory T cells in the peripheral abundant in the reactive lymphocytes of Hodgkin lymphoma.
blood of cancer patients. Clin Cancer Res 2003 Feb;9(2):606–12. Blood 2004 Mar 1;103(5):1755–62.
276. Woo EY, Yeh H, Chu CS, Schlienger K, Carroll RG, Riley JL, et 284. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P,
al. Cutting edge: Regulatory T cells from lung cancer patients et al. Specific recruitment of regulatory T cells in ovarian carcinoma
directly inhibit autologous T cell proliferation. J Immunol 2002 fosters immune privilege and predicts reduced survival. Nat Med
May 1;168(9):4272–6. 2004 Sep;10(9):942–9.
277. O’Garra A, Vieira P. Regulatory T cells and mechanisms of 285. Sutmuller RP, van Duivenvoorde LM, van Elsas A, Schumacher
immune system control. Nat Med 2004 Aug;10(8):801–5. TN, Wildenberg ME, Allison JP, et al. Synergism of cytotoxic
278. Chen ML, Pittet MJ, Gorelik L, Flavell RA, Weissleder R, von T lymphocyte-associated antigen 4 blockade and depletion of
Boehmer H, et al. Regulatory T cells suppress tumor-specific CD25(+) regulatory T cells in antitumor therapy reveals alter-
CD8 T cell cytotoxicity through TGF-beta signals in vivo. Proc native pathways for suppression of autoreactive cytotoxic T
Natl Acad Sci U S A 2005 Jan 11;102(2):419–24. lymphocyte responses. J Exp Med 2001 Sep 17;194(6):
279. Cederbom L, Hall H, Ivars F. CD4+CD25+ regulatory T cells 823–32.
down-regulate co-stimulatory molecules on antigen-presenting 286. Furuichi Y, Tokuyama H, Ueha S, Kurachi M, Moriyasu F,
cells. Eur J Immunol 2000 Jun;30(6):1538–43. Kakimi K. Depletion of CD25+CD4+T cells (Tregs) enhances
280. Wang HY, Peng G, Guo Z, Shevach EM, Wang RF. Recognition the HBV-specific CD8+ T cell response primed by DNA immu-
of a new ARTC1 peptide ligand uniquely expressed in tumor nization. World J Gastroenterol 2005 Jun 28;11(24):3772–7.
cells by antigen-specific CD4+ regulatory T cells. J Immunol 287. Best A, Hidalgo G, Mitchell K, Yannelli JR, Issues concerning
2005 Mar 1;174(5):2661–70. the large scale cryopreservation of peripheral blood mononuclear
281. Javia LR, Rosenberg SA. CD4+CD25+ suppressor lymphocytes cells (PBMC) for immunotherapy trials. Cryobiology 2007 Feb
in the circulation of patients immunized against melanoma anti- 28;54:294–297.
gens. J Immunother 2003 Jan–Feb;26(1):85–93. 288. Keilholz U, Weber J, Finke JH, Gabrilovich DI, Kast WM, Disis
282. Liyanage UK, Moore TT, Joo HG, Tanaka Y, Herrmann V, ML, et al. Immunologic monitoring of cancer vaccine therapy:
Doherty G, et al. Prevalence of regulatory T cells is increased in results of a workshop sponsored by the Society for Biological
peripheral blood and tumor microenvironment of patients with Therapy. J Immunother 2002 Mar–Apr;25(2):97–138.
16 Growth and differentiation factors as cancer
therapeutics
KAPIL MEHTA, ROBERT K. OLDHAM, AND BULENT OZPOLAT

Abbreviations used ADCC, antibody-dependent cyto- further division and are destined to undergo programmed
toxicity; ANLL, acute non-lymphoblastic leukemias; cell death. Another line of evidence suggesting that growth
APL, acute promyelocytic leukemia; AML, acute myel- and maturation factors may be operative in some form of
oid leukemia; ATRA, all-trans-retinoic acid; CML, cancers is the observation that patients with neuroblas-
chronic myelogenous leukemia; CSF, colony-stimulating toma and germ cell tumors can show maturational effects
factor; DBD, DNA-binding domain; DMSO, dimethyl- in the tumor biopsies post treatment. It is not known
sulfoxide; HMBA, hexamethylenebisacetamide; IL-1, -2, whether this relates to the treatment since occasional
etc., interleukin-1, -2, etc.; LBD, ligand-binding domain; spontaneous maturation had been seen prior to effective
MDS, myelodysplastic syndromes; PKC, protein kinase therapy for these cancers. These lines of evidence have led
C; RAR, retinoic recepotr; RXR, retinoid X receptor; some researchers and clinicians to speculate that cancer-
TNF, tumor necrosis factor ous growth remains under some degree of control by dif-
ferentiation and growth factors and that these processes
might be augmented or altered therapeutically.
Differentiation therapy for the treatment of malignant dis- A number of observations suggest that differentiation of
orders offers an attractive alternative to the conventional human myeloid leukemic cells occurs in vivo. The most
cytotoxic chemotherapy. The concept of differentiation obvious example is chronic myeloid leukemia (CML),
therapy is based on the principal of ‘reform’ rather than where glucose-6-phosphate dehydrogenase (G6PD) poly-
‘retaliation’. The perception that malignant cell results morphism revealed the derivation of mature granulocytes,
from a block in the differentiation pathway has led to a red cells, and platelets from the leukemic clone. Similar
conceptual strategy to remove this block and to re-estab- analysis revealed differentiation of acute myeloid leukemia
lish normal homeostasis. Differentiation therapy, in gen- (AML) clones to red blood cells and platelets in some
eral, is associated with a growth arrest and long-term patients [95], and the presence of Auer rods in mature neu-
commitment of the cell to die via apoptosis or senescence. trophils of AML patients in remission and persistence of
The conventional cytotoxic therapy contrasts from the cytogenetic abnormalities in the absence of normal meta-
differentiation therapy in that there is no attempt to restore phases in patients with AML in clinical remission [26, 89].
normal homeostasis in the former case and it is accompa- DNA recombinant technology has also demonstrated
nied by immediate cell death. The treatment of acute pro- leukemic cell differentiation in AML and preleukemia/
myelocytic leukemia (APL) by all-trans-retinoic acid myelodysplastic syndrome (MDS). DNA restriction frag-
(ATRA) revolutionized the treatment of the cancer and ment-length polymorphism in heterozygous individuals
provided the first example and proof of concept of dif- has provided strong evidence for in vivo differentiation
ferentiation therapy. Therefore, the differentiation therapy [95, 417]. HPRT gene analysis, quantitative analysis of
may offer the opportunity for the use of the new, rela- chromosome 8 trisomy and chromosome 7 monosomy,
tively non-toxic agents as well as the use of current che- and immunoglobin (Ig) gene rearrangement analysis
motherapeutic agents at doses significantly lower than showed that five of six patients with AML had maturation
those maximally tolerated. Combining low dose of che- to mature granulocytes in the active phase of the disease
motherapy with one or more differentiation agents may even in aneuploid leukemia. Another approach has been
also be of particular interest in the management of disor- to label myeloblasts in vivo with bromodeoxyuridine
ders resistant to conventional drug therapy. (BUdR), which allowed the demonstration of BUdR in
Interest in the therapeutic use of differentiation agents mature granulocytes of AML patients [326].
has resurged following the discovery of the dramatic Premature chromosome condensation has been used
effects of retinoic acid in the treatment of acute promyelo- to detect the presence of abnormal cytogenetic clones in
cytic leukemia (APL). Treatment with all-trans retinoic mature cells. Fusion of mitotic tissue culture cells with
acid (ATRA) induces differentiation of APL cells into the mature cell in question (e.g., granulocytes) results in
mature phenotype such that they are no longer capable of immediate chromosome condensation of the chromatin

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 527
© Springer Science + Business Media B.V. 2009
528 Growth and differentiation factors as cancer therapeutics

of interphase nuclei into discrete chromosomes. This the use of leukemic cell lines of myeloid lineage capable
permitted karyotype analysis of nondividing cells of 13 of terminal maturation.
patients with CML, and with MDS or AML, after low- The human myeloid leukemic cell lines HL-60, NB4
and high-dose chemotherapy [159]. Twelve of these and U937 that can undergo myeloid differentiation by
cases showed evidence of maturation of cytogenetically all-trans-retinoic acid (ATRA) or other differentiation
abnormal leukemic clones in vivo. inducing agents have been studied most extensively.
Cell lines such as the human myelomonocytic leuke- HL-60 was isolated from a patient with acute promyelo-
mia HL-60 cells are promiscuous in undergoing drug- cytic leukemia (APL), and it retains a promyelocytic
induced differentiation. Indeed, over 80 distinct morphology but does not express translocation t(15;17)
compounds (not including an even greater number of represent M2 type of AML (FAB classification) [110].
analogues) have been shown to induce differentiation of NB4 cells was isolated from long-term cultures of leu-
the HL-60 cell line (vide infra). Many of these agents kemia blast cells on bone-marrow stromal fibroblasts of
may operate via common pathways; nevertheless they a patient with APL [430]. NB4 cells (M3-AML) are true
belong to distinct categories of compounds and can be APL cells harboring t(15;17); thus expressing PML-
divided into the following groups: RARα fusion protein, leading to a differentiation block
at the promyelocytic stage. U937 (M4/M5-AML) was
1. Vitamins, including retinoids and vitamin D3
established from the pleural fluid of a patient with true
[1,(25-(OH)2 D3), and vitamin E.
macrophage-type diffuse histiocytic lymphoma, and has
2. Polar/planar compounds such as dimethylsulfoxide
an immature monoblast-macrophage morphology [365].
(DMSO), hexamethylene bisacetamide (HMBA),
These lines are cloned, readily maintained in culture
N-methylformamide (NMF), cotylenin A
and, while resembling immature cells of their lineage,
3. Cytokines and hematopoietic growth factors (e.g.,
can be induced to terminal neutrophil, eosinophil, and
Erythropoietin (Epo), G-CSF, GM-CSF, IL-1, IL-3,
macrophage (HL-60) or monocyte-macrophage (U937)
IL-6, LIF, TNF, TGFβ, IFNα, IFNβ, IFNγ)
differentiation. NB4 cells have bileneage potential and
4. Phorbol esters (e.g., phorbol myristate acetate)
undergo phenotypic change along monocytoid/mac-
5. Chemotherapeutic agents that interfere with DNA
rophage lines with phorbol ester induction and granulo-
synthesis (e.g., cytosine arabinoside, tiazofurin,
cytic differentiation by retinoic acid [431].
6-thioguanine)
The histochemistry and morphology of these cell lines
6. Chemotherapeutic agents that influence topoi-
typify them as immature cells of myelomonocytic lin-
somerase II
eage. The HL-60 cell line exists in many variants with
7. Chemotherapeutic agents that inhibit DNA methyltrans-
differing doubling times and extents of spontaneous dif-
ferase (e.g., 5-azacytidine, 5-aza-2′-deoxycytidine)
ferentiation [123]. The cells weakly phagocytize latex
8. Hormones (steroids-dexamethasone and predniso-
particles or yeast in the uninduced state and possess low
lone) [429].
numbers of Fc C3b, insulin, and chemotactic fMLP recep-
9. Chromatine remodeling: Histone deaceltilase inhib-
tors, whose expression can be greatly enhanced following
itors (HDAC) (butyrates, valproic acid)
exposure to differentiation-inducing agents [4, 286].
10. Others: adenosine analogs (neplanocin A, deoxyco-
HL-60 cells are positive to myeloperoxidase, ASD
formycin), arsenic trioxide, peroxisosome prolifer-
chloracetate esterase, and Sudan black B, but unlike nor-
ator-activated recptor gamma (PPARγ)
mal promyelocytes they do not stain for alkaline phos-
phatase, and unlike macrophages they are essentially
acid phosphatase-negative [110, 410]. Activated forms
Human Leukemic Cell Lines of beta-glucuronidase, lysozyme, and G6PD, found at
high levels in the granules of normal granulocytes, are
as Models for Differentiation present at low concentrations in HL-60 and are mini-
Therapy mally involved in degranulation. It has been proposed
that these enzymes are already synthesized in HL-60 and
Much of the history of the development of an effective stored within the granules as zymogens, but are only
chemotherapy for leukemia was based on the availability converted to active enzymes upon stimulation [204,
of cell lines, or transplantable leukemias, with sensitivity 422]. Uninduced HL-60 cells have a marginal capacity
to cell cycle-specific agents. The development of biologi- to produce H2O2 and O2−, have a low level of HMPS
cal response modification therapy and the recognition of activity [284], and have a greatly impaired ability to kill
leukemic cell differentiation as an obtainable goal led to staphylococcus and other microorganisms [110].
Kapil Mehta et al. 529

The U937 cell line as originally reported grew slowly; or in the segmented neutrophils of the majority of patients
however, later passages of the line have an accelerated with leukemia [38]. Under normal conditions, lactofer-
population doubling of 20–48 h. The cells have a mono- rin is active as a suppressor molecule, and inhibits the
blastic morphology and histochemical profile (strong production and/or release of granulocyte-macrophage
ANAE, NASDAE, and beta-glucuronidase positivity colony-stimulating factors from monocytes and mac-
and weak acid and alkaline phosphatase) [365, 410]. rophages at concentrations as low as 10−15 M. In vivo
U937 cells also secrete lysozyme, and neutral protease lactoferrin inhibits myelopoiesis in mice at concentra-
elastase is present within the cells; these are monocyte- tions as low as 10−4 mg/mouse [34]. Therefore, detection
specific characteristics [323, 365]. The cells bear few of the synthesis of lactoferrin in HL-60 cells, even at
Fc, C3, and chemotactic peptide receptors when com- low concentrations, would be of importance, especially
pared with normal monocytes, but histamine and insulin if the synthesis could be increased by differentiation-
receptors are expressed [5, 365]. Only a small percentage inducing agents and if the lactoferrin so induced was
of U937 cells are phagocytic, and weakly produce H2O2 functionally active. We have detected lactoferrin in
and O2− and are incapable of killing microorganisms or HL-60 cells at very low levels, using radioimmunoas-
tumor cells [204, 324, 355]. say; in biosynthesis studies, using autoradiographic gel
Upon induction of differentiation, morphological analysis (3H) leucine incorporated into material immu-
changes involve significant increases in substratum noprecipitated with lactoferrin antibody. Following
adherence. The HL-60 cell line changes morphology induction of differentiation with DMSO or retinoic acid,
from promyelocytic to later stages of granulocyte a two- to fourfold increase in lactoferrin has been noted.
degranule formation when treated with 2% DMSO and This is still much lower than the 10−12 g per segmented
retinoic acid [37, 41, 110], whereas phorbol esters trans- neutrophil reported in normal peripheral blood, indicat-
form the cells to monocyte-macrophages with the disap- ing a persisting abnormality in the differentiated HL-60
pearance of azurophilic granules and appearance of neutrophil, comparable to that noted in neutrophils of
pseudopodia and a cerebriform nucleus [337]. There is patients with leukemia [38].
evidence that these cells can differentiate to eosinophils The isozyme pattern of lactate dehydrogenase in
spontaneously, and in response to stimulatory activities HL-60 cells is reported to change after induction with
in human placenta-conditioned media [222]. U937 cells, DMSO; however, the resultant pattern is not character-
in response to differentiation stimuli, increase in size istic of mature granulocytes or macrophages but rather
and acquire lobated nuclei, and cytoplasmic granules of more immature stages [303]. Simultaneous expres-
are replaced by vacuoles, duplicating monoblast to sion of other granulocytic and monocytic specific mark-
monocyte transformation. Various granulocyte lysozyme, ers may also be the result of incomplete or asynchronous
acid phosphatase, and beta-glucuronidase activities maturation. Of particular interest is the observation that
have been measured in induced cells; when HL-60 is cell division is not required for the induction of many of
induced with phorbol ester (TPA), the activity of these these characteristics and may account for their uncoor-
enzymes is increased up to 20-fold [337]. Similar dinated expression [37].
increases in activity are usually seen after induction Messenger RNA species have been isolated from
with all agents used for differentiation induction. The HL-60 cells translated in vitro and analyzed by two-
distinctive monocyte lysosomal enzyme, acid phos- dimensional SDS-PAGE after phorbol ester or DMSO
phatase, can be detected after exposure of HL-60 cells induction. The phorbol-induced protein pattern differed
to phorbol ester for several days. The elaboration of this significantly from the DMSO pattern and was character-
enzyme into surrounding media is also a monocyte- ized as monocytoid rather than granulocytic [61]. Poly
associated characteristic and accompanies the phorbol- ADP-ribose metabolism has been linked to the maturation
induced maturation of these cells. Myeloperoxidase, an of granulocyte-macrophage progenitors. ADP-ribosyl
enzyme specific to the myelomonocytic lineage, is con- transferase is a chromatin-bound enzyme catalyzing the
stitutively expressed in HL-60. However, activity is lost transfer of ADP-ribose to chromatin proteins. The activity
after induction with phorbol esters, lymphokines, or of this enzyme increases during the CSF-stimulated dif-
retinoic acid, but is unaffected when DMSO or cAMP- ferentiation of marrow precursors to monocytes [104].
inducing agents are used [338]. Using DNA array, suppression-subtractive hybridization,
Lactoferrin, a metal-binding glycoprotein, is present in and differential-display PCR techniques, the expression
normal granulocytes in the secondary (specific) granules of about 169 genes was shown to be modulated in
and is one of the most specific markers for the neutrophil ATRA-treated human APL (NB4) cells [230]. Of these
lineage. It has not been detected in HL-60 cells [284], 32 genes appeared to be the direct targets of the ATRA
530 Growth and differentiation factors as cancer therapeutics

signaling pathway since their transcriptional regulation stimulation [200], and differed from peripheral blood
was not blocked by cyclohexamide. A similar detailed monocytes and macrophages in this respect. However,
analysis of genes regulated by ATRA-treatment in mouse if supplied with exogenous arachidonic acid, the cells
APL cells (MPRO) was reported [223]. Both the studies can release PGE2 [60]. The failure to release esterified
suggested the involvement of several transcription regu- arachidonic acid to the cyclooxygenase pathway was
latory factors during the process of ATRA-induced believed to be a property shared by U937 cells and certain
granulocytic differentiation. macrophage subsets.
One of the most frequently employed and convenient The production of certain biologically active macro-
qualitative measures of reactive oxygen intermediates as molecules such as, interleukin 1 (IL-1) (endogenous
important microbicidal products or granulocytes of mac- pyrogen, LAF) can be induced by treatment of U937
rophages is the reduction of NBT to a black formizan cells with agents like endotoxin or phorbol esters [302].
precipitate. A variety of factors, both physiologic and IL-2 is probably the factor released by U937 that
pharmacologic, can induce HL-60 production of H2O2 increases the production of collagenase and PGE2 in
[141, 117, 294]. The microbicidal capacity of granulo- cultured synovial cells [9]. U937 cells also secrete
cytes and HL-60 cells is metabolically linked to an hematopoietic growth-stimulating factors that may act
increase in the hexose monophosphate shunt activity and as autostimulatory growth factors [14]. The release of
consequent production of superoxide anion activity. such factors can be suppressed, as in normal myelopoi-
Arachidonic acid metabolism has been analyzed after esis, by agents such as lactoferrin [38]. U937 also pro-
induction of maturation with several agents, and recent duces the second component of complement, and the
results suggest that there is enhanced synthesis and release release of C2 can be augmented sevenfold by inducing
of prostoglandins of the E and F2-alpha type [142]. maturation of U937 with phorbol ester or a lymphokine
Maturation of U937 cells is usually characterized by [302, 313]. Similarly, treatment of U937 cells with Con
quantitative rather than qualitative changes, since the A has been shown to induce the synthesis of a 65 kDa
cells constitutively produce a variety of enzymes and heat resistant lymphocyte proliferation inhibitor that is
growth factors. Because U937 cells are of monocytic distinct from lymphotoxin or interferons [405].
origin, agents that effect a maturational change may also The qualitative and quantitative changes in the
be potent immunoadjuvants. Blood monocytes and expression of certain plasma membrane constituents in
U937 cells are characterized by their content of fluoride- HL-60 and U937 have been associated with differentia-
inhabitable esterase, alkaline and acid phosphatase, and tion. Induction of HL-60 with DMSO or phorbol ester,
β-glucuronidase. These enzyme levels are elevated by for example, modifies the expression of “fast-eluting”
exposure of normal or neoplastic cells to agents known glycopeptides [387]. Comparison of the elution profiles
to influence monocytic activation, such as vitamin D of fucosyl-labeled glycopeptides with normal granulo-
metabolites, phorbol esters, gamma-interferon, and cytes and macrophages revealed that HL-60 glycopep-
lymphokines [141, 142, 147]. Other enzymatic activi- tides were larger and more complex than those on a
ties that have been studied in U937 cells include elastase, normal band of polymorphonuclear neutrophils. DMSO
collagenase, and plasminogen activator. U937 elastase or phorbol treatment of HL-60 did not induce a normal-
is not released constitutively and is not readily modu- ization of the elution profile to a mature conformation
lated [287, 353]. [385]. Induction of cell-surface glycoproteins on HL-60,
The production of reactive oxygen species and the particularly as seen in the time of appearance of gp130
percentage of NBT-positive cells are increased in U937 following DMSO treatment, was concomitant with
cells by a variety of agents, such as phorbol esters, development of chemotactic ability [109]. In this con-
DMSO, lymphokines, α- and β-interferons, retinoic text, patients with impaired granulocyte chemotaxis
acid, 1,25-dihydroxy vitamin D3, prostaglandin (PG) E have greatly reduced amounts of GP130 in their granu-
and other cAMP-inducing agents, cytosine arabinoside locytes [342].
(ara-C), and protein DIFs [2, 34, 141, 292, 293]. On a Cytoskeletal proteins of HL-60 have also been studied.
more quantitative basis, the activity of the hexose mono- During induced maturation, there is increased synthesis of
phosphate shunt has been examined. Notably, db-cAMP vimentin and other structural proteins. As may be expected,
and PGE are able to increase activity of the shunt, but the profiles of cytoskeletal proteins induced by DMSO
cannot induce other characteristics of maturity in U937 resemble those of granulocytes and, following phorbol
[291]. Under certain conditions, U937 cells did not appear ester induction, resemble those of macrophages [27].
to produce prostaglandin E2 in response to lipopolysac- Modulation of expression of several cell surface anti-
charide (LPS), endotoxin, or concanavalin A (Con A) gens has been characterized by the use of monoclonal
Kapil Mehta et al. 531

antibodies. In one study, two monoclonal antibodies, protein of molecular weight 17,000, termed tumor
B9.8.1 (specific for monocytes and metamyelocytes), necrosis factor (TNF) [6, 309]. This activity is inhibi-
were used to characterize maturational changes [310]. tory to neoplastic cell proliferation, including that of
Treatment of HL-60 with granulocyte- or macrophage- leukemic cells and normal hematopoietic stem cells.
inducing agents resulted in enhanced antigen expres- The intriguing possibility of autoregulation of leukemic
sion. In a similar study, reactivity with the monoclonal cells by production of an endogenous growth-inhibitory
antibodies Mo1 and Mo2, recognizing determinants molecule (TNF) is suggested.
specific to the myelomonocytic lineage, could be More than 80% of cells from a human promyelocytic
induced with phorbol esters, as well as protein inducers leukemic cell line (HL-60) possess the capacity for self-
of differentiation [69, 379]. renewal as evidenced by their ability to form large pri-
HL-60 constitutively expresses HLA-A and -B anti- mary colonies in semisolid medium and the presence
gens, and induction of HLA-DR determinants has been within these colonies of cells capable of subsequent
reported [69]. Normal myelomonocytic precursors colony formation. Colony development is independent
express the DR antigens twice: transiently during early of colony-stimulating factor. The observed autostimula-
stages (CFU-GM) [98], and later as monocytes. tion suggests the production of specific growth promot-
HLA-DR determinants of HL-60 have been detected ers by the cells. Differentiation, either to mature
following treatment with inducers of monocyte mac- granulocytes or to macrophages, induced by various
rophage differentiation, but not with agents that induce agents is associated with reduced cloning potential.
granulocyte maturation. Nevertheless, colonies containing differentiated cells
The expression of Fc and C3 receptors has been studied can be developed either by cloning cells in the presence
extensively in U937. The receptor for IgG1 can be induced of suboptimal concentrations of inducer or by adding
by a variety of agents, both physiologic and pharmaco- inducers to colonies developed in its absence. The loss
logic [141, 200]. The receptor for IgE is also present, of self-renewal was found to be one of the early proper-
but its modulation has not been studied. The C3B receptor ties that changed following the initiation of differentia-
(as recognized by EAC or Mac-1 monoclonal antibody) tion. The loss preceded not only the overt expression of
is inducible in a similar manner, as are Mac-1 and mac- maturation-specific functions but also cellular commit-
rophage-restricted Mac-3 cell-surface antigens [325]. ment to terminal differentiation; shorter contact with the
The chemotactic receptor for the potent chemotactic inducer is required to cause loss of self-renewal than to
peptide fMLP is weakly expressed in U937 (7,505 sites/ induce an irreversible transition to differentiation. This
cell). Within 3 days of exposure to phorbol esters, dex- results in cells that lose their self-renewal potential
amethasone, or protein inducers of differentiation without being able to complete their program of differ-
(excluding native or recombinant γ-interferon), up to entiation [96]. Increased migration of tumor cells upon
fivefold increase in fMLP receptors is seen [142, 313]. differentiation to form diffuse colonies may be the result
U937 cells are generally considered devoid of any Ia of increased mobility and chemotactic responsiveness.
antigen on their surface; however, small numbers of In a modified chemotaxis assay, HL-60 cells increased
cells react with antibodies to HLA-DR and HLA-DS/ their migration in response to fMLP after a 5-day of
CD molecules. It has proved possible to clone such induction with db-cAMP or DMF [99, 388].
Ia-positive cells and develop constitutively Ia antigen- Phagocytosis of latex beads or opsonized yeast and
positive variant lines of U937 [121]. Induction of the capacity to kill microorganisms are readily induced
HLA-DR determinants in response to treatment with by a wide variety of differentiation-inducing agents.
gamma-interferon and PG has been reported [311]. Phagocytosis is one of the earliest acquired effector
Beta2-microglobulin expression has also been shown to functions of both granulocytes and monocytes. Phorbol
increase in response to protein inducers of differentia- ester or DMSO-induced HL-60 cells effectively kill
tion [287]. Insulin may play an important role in the staphylococcus [190]. It has been demonstrated that dif-
growth regulation of U937 cells, since the number of ferentiation-induced HL-60 cells can mediate monocyte
insulin receptors is reduced after incubation with differ- ADCC-like reaction against antibody-coated chicken
entiation-inducing agents, and this may have a causal erythrocytes [69].
role in the observed growth inhibition. The association The U937 cell line is capable of being induced to
between differentiation induction and growth inhibition mediate antibody-dependent cellular cytotoxicity
of U937 and HL-60 cells may also be linked to the ability (ADCC) effector function. First reports of ADCC capacity
of potent inducers of macrophage differentiation, such against tumor markers used lymphokine, interferon, and
as phorbol ester, to induce cellular production of the phorbol ester preparations to activate or “induce” the
532 Growth and differentiation factors as cancer therapeutics

cells [147, 324, 365]. ADCC activity of U937 cells genes other than c-myc; however, tests with several other
against erythrocytes can be induced by as little as ten oncogene probes, including N-ras, Ha-ras, Ki-ras, myb,
units of γ-interferon and 300 units of α- and β-interferon. and abl, showed no evidence of amplification or gross
U937 cells have been studied for their ability to support rearrangement of these genes in DNA in any HL-60 line
the intracellular multiplication of Toxoplasma gondii. or subline [65]. High levels of c-fos expression have been
A small portion of U937 cells can spontaneously phago- detected in macrophages, but not in uninduced HL-60
cytize these protozoans, and after 24 h those cells have cells. However, when HL-60 cells were induced to mac-
either lysed or contain numerous trophozoites within their rophages, c-fos expression was readily detectable in the
vacuoles. After a 3-day incubation with lymphokine differentiated cells [272].
preparations there is a significant decrease in the num-
ber of organisms found within the vacuoles, in spite of a
generalized increase in phagocytosis [408].
In an effort to analyze the pathways involved in dif-
Vitamin A Analogues
ferentiation of cell lines such as HL-60, many attempts as Leukemia Differentiation-
have been made to select sublines capable of sustaining
exponential growth in the presence of a single inducer of Inducing Agents
differentiation but not in the presence of different, struc- Retinoids (vitamin A and its analogues) and retinoic acid,
turally unrelated inducers of differentiation. Neutrophilic in particular, are a family of fat soluble compounds that
granulocytic and monocyte/macrophage programs of exert a potent effect on the cell growth and differentiation
HL-60 are mechanistically different and separable, and of various cell types including promyelocytic leukemia,
both agent-specific and common quantitative alterations neuroblastoma, teratocarcinoma, breast cancer, prostate
contribute to the mechanism for resistance to granulo- cancer, melanoma, bladder cancer, squamous cell carcino-
cyte differentiation. mas of cervix, skin cancer, head and neck cancer and rhab-
Numerous studies have demonstrated that the oncogene domyosarcoma [68, 342, 361, 434]. Their antiproliferative
c-myc is amplified 20- to 40-fold in HL-60 cells compared and differentiation-inducing properties have brought them
with normal human DNA and is associated with an ele- under the close scrutiny of oncologists. A major develop-
vated level of cellular myc-mRNA, and a decrease in this ment in the clinical application of retinoids was the dis-
mRNA follows chemically induced differentiation [37, covery that ATRA could induce the mature phenotype in
132]. Within 5 days of addition of DMSO or phorbol ester myeloid leukemia cells thus resulting in complete remis-
to HL-60 cultures, transcription of the c-myc gene was sion in patients with acute promyelocytic leukemia. [46,
markedly reduced when compared with control cultures. 72, 80, 90, 91, 170, 399] In light of high complete remis-
This decrease was not accompanied by alteration in either sion rates achieved (80–90%), ATRA is currently being
the bulk rate of transcription or the c-myc copy number, used as a frontline therapy for treatment of APL patients.
suggesting that decreased cellular myc RNA levels are Retinoids exert their biological effects by interacting
due to decreased transcription of the myc protooncogene with two families of intracellular nuclear receptors: the
[132]. The expression of c-myc in HL-60 does not corre- retinoic acid receptors (RARs), which are activated by
late with the proportion of proliferating cells, and the ATRA and 9-cis retinoic acid, and the retinoid X receptors
kinetics of decrease upon DMSO induction is paralleled (RXRs), which are activated by 9-cis retinoic acid only
closely by an increasing proportion of histochemically [32, 227]. Each of these receptor families consists of three
detected, differentiated myeloid cells and by a decrease in subtypes (- α-β and -γ), and each subtype exists in multiple
clonogenic potential, but not by changes in the proportion isoforms that arise due to alternative splicing and differen-
of proliferating cells [97]. Changes in c-myc expression tial use of two promoters (Table 1).
subsequent to differentiation of HL-60 can therefore be
directly related to the differentiation process rather than to
a cell cycle-related phenomenon. Gallagher et al. [112] Table 1. Retinoic acid (RAR) and retinoid X (RXR) receptors
observed a little change or decrease in the amplification Receptor Isoforms Chromosomal location Ligand
level of the known amplified c-myc gene in various drug-
RARα α1, α2 17θ21.1
resistant sublines in comparison with wild-type HL-60
RARβ β1, β2, β3, β4 3p24 ATRA &
cells and despite the existence of numerous double, min- RARγ γ1, γ2 12q13 9-cis RA
ute chromosomes (indicators of amplified genes) in some RXRα α1, α2 9q34.3
drug-resistant sublines. Differential response to differen- RXRβ β1, β2 6q21.3 9-cis RA
tiation agents could still be related to amplification of RXRγ γ1, γ2 1q22–q23
Kapil Mehta et al. 533

RARs and RXRs share – along with other members receptors but dissociate upon ligand binding (Fig. 1a),
of the nuclear hormone receptor superfamily – a modular and (ii) the co-activators that bind only to the liganded
structure having a highly conserved DNA binding receptors (Fig. 1b). Two corepressors, SMRT and
domain (DBD), a ligand-binding domain, and a less N-CoR, have been identified that associate with unli-
well conserved amino-terminal domain and a hinge ganded retinoid receptors and suppress the basal tran-
region present between the DBD and the LBD. The scriptional activity. Another co-repressor, mSin3A,
DBD has two zinc fingers and is highly conserved which is a homologue of the yeast global-transcriptional
between the members of the superfamily. The LBD is repressor Sin3p, has been shown to interact directly
also involved in dimerization and has a carboxyl-termi- with SMRT and N-CoR1. Sin3A and SMRT, in turn, can
nal activation domain, called AF-2. The AF-2 domain interact with histone deacytylase 1 (HDAC1) to form a
has been shown to function as an autonomous transcrip- multisubunit repressor complex [277].
tional activation domain when fused to a heterologous Similarly, a growing number of coactivator proteins
DBD. The hinge region of the retinoid receptors has have been identified, though their mechanism of action
been shown to constitute the domain responsible for is not well understood except for it has been known that
interaction with some co-repressors [51, 52]. they bind to the liganded receptor at the AF-1 domain
The presence of intermediary factors that link nuclear [208]. One factor that is known to potentiate the tran-
receptors with basal transcriptional factors had been scriptional activation of the retinoid receptors is the steroid
proposed based on transcriptional interference/squelch- receptor coactivator SRC-1 [412]. The formation of a
ing observed between transcriptional activation domains RXR/RAR heterodimer is required for the high- affinity
of different receptors. Two kinds of factors have been binding to specific DNA sequences known as retinoic acid
identified: (i) the corepressors, which bind to unliganded response elements (RAREs) that is critical for subsequent

a +0.1-1 µM ATRA +9-cis RA

RARα RARα
RXRα RXRα
HL - 60 Cell/WT Differentiation Apoptosis

b
1 µM ATRA
Differentiation blocked due to mutation in the RARα gene

HL-60R

c RARα-transfected HL-60 R cells regain


0,1-1 µM ATRA differentiation potential in response to ATRA
treatment

HL-60R/RARα
d
0.1 µM 9-cis-RA RXR -transfected HL-60R cells undergo
apoptosis in response to RXR ligation

HL-60R/RXRa

Figure 1. A model showing the basal repression and retinoid-induced transcriptional regulation of the target gene(s). (a) The
RAR/RXR heterodimer binds to a specific direct-repeat regulatory element that is separated by 5 bp (DR5-RARE) in the regula-
tory site of the target gene. In the absence of ligand (ATRA), the RAR/RXR heterodimer recruits a transcriptional repression
complex that comprises N-CoR, mSin3a, SMRT, and a histone deacetylase (HDAC-1). The HDAC-1 activity helps to suppress
transcriptional activity. (b) Addition of ATRA results in a conformational change in the RAR/RXR heterodimer that leads to the
release of the repressor complex and recruitment of a transcriptional activator complex that has histone acetyltransferase (HAT)
activity. This activity destabilizes nucleosomes and creates a permissive state for promoter activation
534 Growth and differentiation factors as cancer therapeutics

retinoid-induced transcription of target genes. Most TRAIL (tumor necrosis factor related apoptosis induc-
RAREs have been identified in the regulatory regions of ing ligand) expression. Thus, induction of TRAIL-
genes whose transcription is induced by retinoids. These mediated death signaling may contribute to the thera-
cognate response elements consist of direct repeats (DR) peutic value of retinoids [8].
of two core motifs in the form of PUG(G/T)TCA(X) A two-step model has been proposed for induction
nPUG(G/T)TCA or similar but degenerate motifs. The of differentiation where early events anteceding
most common spacing observed in RAREs is 5 bp(DR5); precommitment regulate growth arrest and late events,
however, RAREs containing 1 (DR1) and 2 bp (DR2) are and subsequent to precommitment regulate the choice
also common. of a specific differentiation lineage [414]. Thus the
HL-60 and NB4 cells have been extensively used to lineage specificity of cells treated sequentially with
investigate retinoid-mediated signaling pathways dur- two discrete exposures to alternative inducers depends
ing normal myelopoesis. Like normal granulocytes, on the order of exposure. Retinoic acid exposure of
they undergo terminal differentiation in response to HL-60 for 24–72 h followed by phorbol ester treatment
retinoid treatment. Moreover, like normal granulocytes, produces monocyte-macrophage differentiation, whereas
ATRA-induced HL-60 and NB4 cells have a limited reversing the order of treatment favors granulocyte
in vitro life span and undergo apoptosis in response to differentiation [425]. The precommitment phase has a
certain stimuli [233, 431]. Thus, Both of these cells characteristic duration following retinoic acid exposure,
can also serve as a model to investigate the molecular persisting for several cell cycles despite removal of
mechanisms of apoptosis in terminally differentiated retinoic acid [134]. This precommitment is paralleled
hematopoietic cells. However, the interpretation of by elevations in c-myc. Other events associated with
retinoids’ action on cell growth, differentiation, or apop- retinoid induction of differentiation involve elevation of
tosis becomes complicated in view of the fact that retin- tyrosine kinase activity [74] and a protein kinase C cascade
oids can mediate these effects by binding and activating system. Sphinganine, a potent inhibitor of PKC, enhances
two different types of nuclear receptors: the RARs and differentiation of HL-60 induced by retinoids, and the
the RXRs, which as described above, differ in their granulocytes produced are more fully differentiated as
sequences and exhibit distinct ligand-binding proper- indicated by enhanced superoxide production in response
ties. Since most of the blood cells, including HL-60 to fMLP [363]. A role for topoisomerase II in retinoid-
and NB4, express both types of receptors [144, 327], induced granulocytic differentiation has been suggested
retinoid-induced effects in these cells may be a result of [102]. In HL-60 differentiation, retinoic acid stimulates
activation of either RARs, RXRs or both types of receptors. transient relocation of DNA supercoiling, and this is
We addressed this problem by using a mutant subclone associated with the formation of small numbers of
of the HL-60 cell line (HL-60R) in which retinoid receptor protein-linked DNA breaks (a characteristic of topoi-
function has been abrogated as a result of a -trans dominant somerase reactions). Both events are perturbed by VP16,
negative regulatory point mutation in the ligand-binding which inhibits differentiation [102].
domain of the receptor [333]. HL-60R subclones express- Combinations of differentiation-inducing agents have
ing specific receptors were generated by retrovirus- been explored in vitro with retinoic acid. Twenty-two of
mediated transduction of RARα or RXRα specific 24 patients with ANLL showed differentiation of bone
coding sequences [334]. Our results suggested that the marrow in cultures with a combination of retinoic acid
introduction of RARα into HL-60R cells completely (10−6 M), aclacinomycin A (80 nM), and dimethylforma-
restored their sensitivity to ATRA-induced granulocytic mide (100 mM) [145]. Enhancement of differentiation
differentiation. In contrast, the introduction of RXRα was seen with retinoid combined with interferons-α and
cDNA rendered these cells remarkably sensitive to -β [183, 193, 196], and -&gammabdot; [153, 383].
apoptosis in response to the RXR-specific legends [239]. Tumor necrosis factor (TNF) at 2.5 U/ml inhibited
These observations provided a direct evidence for growth and, synergistically with retinoic acid, induced
RARα involvement in retinoid-induced differentiation differentiation in HL-60 and KG-1 cultures and in mar-
and of RXRα in programmed cell death (Fig. 2). Using row cultures from four to nine patients with ANLL
the receptor-selective retinoids, other investigators [378]. Retinoic acid reversed TNF inhibition of normal
arrived at similar conclusion that specific retinoid recep- marrow myeloid colonies and leukemic growth marrow
tors are involved in the regulation of differentiation and cultures from three to nine patients with ANLL.
apoptotic events in HL-60 cells [278, 386]. In this context, leukemia differentiation-inducing factor
More recently, it was demonstrated that ATRA induces (GM-DF) produced by mitogen-stimulated human leu-
post-maturation apoptosis of APL cells by regulating kocytes acted synergistically with retinoic acid in inducing
Kapil Mehta et al. 535

mSin3a HDAC-1
C Repression
a complex
N-CoR SMRT

LBD LBD
RARα TRANSCRIPTIONAL
Histones REPRESSION
DBD DBD

Closed chromatin
(repressed)

5 bp
AGGTCA AGGTCA
DR-5 RARE

ATRA
b SRCs
CPB/p300
(HATs)
LBD LBD

Ligated RARα TRANSCRIPTIONAL


ACTIVATION
DBD DBD

Ac Ac Ac Ac Open chromatin
Ac (activated)
Ac

5 bp
AGGTCA AGGTCA
DR-5 RARE

Figure 2. Retinoid-mediated differentiation and apoptosis of myeloid leukemia HL-60 cells


(a) The RARα nuclear receptors in HL-60 cells when bind to and are activated by appropriate ligand (e.g. ATRA) results in
granulocytic differentiation of the cells. The differentiation process is associated with induction of several new genes, including
RXRα receptors. It is conceivable that ATRA is isomerized to 9-cis RA in situ, which can then bind and activate RXRα receptors,
leading to the onset of apoptosis in differentiated HL-60 cells. (b) HL-60R cells harbor a functional mutation in the RARα gene
that results in non-functional RARα protein and renders the cells resistant to ATRA. (c) Retrovirally-transduced expression of
functional RARα in HL-60R cells restores the ability of these cells to differentiate in response to ATRA treatment. (d) However,
transduction of RXRα cDNA, renders the HL-60R cells highly sensitive to RXR-selective ligand(s) (such as, 9-cis RA) and
induce massive apoptosis in these cells without morphological differentiation.

maturation of the human leukemic lines U937 and be induced to mature neutrophil differentiation [74] and
HL-60 [292]. T-cell-derived GM-DF was subsequently retinoic acid induces the human malignant monoblast
shown to be due to the synergistic action of IFNγ, lym- line U937 to monocyte-like cells with the capacity to
photoxin, and GM-DSF [154]. Compounds elevating reduce nitroblue tetrazolium [292], the human myeloid
intracellular levels of cAMP, such as dibutryl cAMP, cell lines KG-1 and K562 cannot be induced to differen-
PGE, and choleratoxin, acted synergistically with retinoic tiate [77]. The mouse myeloid leukemia M1 can be
acid to induce maturation of both cell lines. In contrast induced to increased levels of lysomal enzyme produc-
to the requirement for continuous presence of retinoic tion without induction of phagocytosis, locomotive
acid for up to 5 days in order to achieve terminal activity, or morphological maturation. Fresh leukemic
differentiation of HL-60 cells, differentiation proteins cells from patients with various myeloid leukemias have
or cAMP-elevating compounds are active on leukemic also been exposed to retinoic acid in short-term primary
cells primed with retinoic acid within 8–16 h [292]. suspension cultures, and morphological and functional
Retinoid-mediated differentiation of myeloid leuke- maturation was observed only in cases of acute promy-
mia cells is not a universal phenomenon. While the elocytic leukemia [113, 193]. The differential sensitivity
murine myelomonocytic leukemic cell line WEHI-3 can of various leukemias to retinoic acid induction of terminal
536 Growth and differentiation factors as cancer therapeutics

differentiation is not dependent on the presence or acid, might be effective in the therapy of patients with
absence of cellular retinoic acid binding protein [77, 371]. myelodysplastic syndrome because of the possibility
High-affinity retinoic acid receptors (RARs), predom- of prevention of progression to overt leukemia of these
inantly alpha-type, were found in 12 leukemic cell lines preleukemic patients, for whom no other effective
and in marrow blasts from 32 patients with ANLL [203, treatment is currently available.
283, 396]. While some correlation was found between Table 2 lists some of the clinical studies conducted in
RAR expression and retinoid response with four leuke- patients with myelodysplastic syndromes (MDS). These
mic cell lines [203], extensive analysis of marrow cul- studies employed oral doses of retinoids ranging from
tures from a large group of primary AML patients showed 10–100 mg/m2/d for 4 weeks to 5 years of duration
no correlation between receptor expression and ability of [171]. The percentages of response described in these
retinoic acid to inhibit colony formation or induce dif- studies have been encouraging, but toxicity often lim-
ferentiation [396]. The potential for terminal differentia- ited the duration of the therapy. Hepatotoxicity was
tion may be irreversibly lost in many cases of acute dose-limiting but was completely reversible upon cessa-
myeloid leukemia, but this need not negate the therapeu- tion of the therapy. In addition, cheilosis, hyperkerato-
tic value of retinoic acid treatment in a wide range of sis, stomatitits, and increase in serum triglyceride levels
leukemias, since considerable evidence has accumulated were reported. For example, Besa et al. [29] in their
to suggest that retinoids can selectively inhibit leukemic study observed a 47% response including complete
cell self-renewal independently of activation of a differ- remissions in 17 patients with MDS receiving 100 mg/m2
entiation program in the leukemic stem cell. Retinoic 13-cis-retinoic acid with an improved survival in
acid is a potent inhibitor of the clonal growth in vitro of responders of 33 months versus 10 months in the nonre-
myeloid leukemic cells, and a 50% growth inhibition of sponders. However, no beneficial effects were reported
HL-60 was achieved by 2.4 nM retinoic acid. The human with either retinoic acid or isoretinoin in two studies of
myeloid leukemic line KG-1, which is not inducible to 14 MDS patients [146, 164]. N-4-Hydroxyphenyl-
differentiation, was nevertheless extremely sensitive to retinamide (Fenretinide) lacked clinical effect in 15
retinoic acid, with 50% of the colonies inhibited by MDS patients and in some may have enhanced leukemic
2.4 nM concentrations of the drug [77]. progression [115]. In a double-blind, placebo-controlled
Thus, antiproliferative action of retinoids upon leu- trial of 13-cis-retinoic acid in 68 MDS patients with
kemic cells is more general than the incidence of 100 mg/m2 for 6 months, no significant differences were
induction of terminal differentiation, and is seen with observed between treatment groups [195]. Approximately
retinoid concentrations readily attainable in vivo. The 30% of patients in both groups had progression of the
potential efficacy of retinoic acid in the treatment of disease and survival was virtually identical. Ninety per-
human leukemia is further suggested by the observa- cent of the treated patients developed mild to moderate
tion that it enhances colony-stimulating factor (CSF) skin toxicity.
induced clonal growth of normal human myeloid and In view of these generally negative results, MDS
erythroid cells in vitro [78]. In long-term bone marrow therapy with retinoic acid has been extended to combi-
culture it also enhanced progenitor cell production. nations with other agents with potential differentiation-
Maximal stimulation occurred at a retinoic acid con- inducing capacity (Table 2). In a comparative study of
centration of 3 × 10−7 M acid, which increased the such combination therapy in 62 MDS patients, 50%
mean number of colonies by 213 ± 8% over plates con- response was seen with a combination of retinoic acid,
taining CSF alone [31]. Retinoic acid has no direct 1,25(OH)2 D3, and interferon, a response rate compara-
CSF activity, nor does it stimulate CSF production by ble to that obtained with low-dose cytosine arabinoside
the cultured bone marrow cells or marrow stroma. This [151]. Combining all four agents proved too toxic.
stimulation may be mediated by increased responsive- Combining retinoic acid with low-dose ara-C produced
ness of the progenitor cells, possibly by increasing the favorable results in one study [103], but in another study
number of growth factor receptors per cell. Retinoids of 14 MDS patients the response was no better than
are reported to enhance the binding of epidermal either agent alone [160]. Combining a tocopherol
growth factor (EGF) to fibroblasts and epidermal cells (800 mg/m2) with retinoic acid (100 mg/m2) produced a
by increasing the number of EGF receptors per cell 62% response in 13 MDS patients and reduced the
[176, 188, 319]. Enhancement of normal myelopoiesis toxicity involving liver damage, hyperkeratosis, and
[31] and inhibition of myeloid leukemic cell prolifera- mucositis [29].
tion by retinoic acid suggest that 13-cis-retinoic acid, In poor-prognosis acute nonlymphoblastic leukemia,
which is significantly less toxic in vivo than retinoic 13-cis-retinoic acid has some efficacy on its own in one
Kapil Mehta et al. 537

Table 2. Clinical studies with retinoids in MDS patients


Retinod No. of Pts. Dose (mg/m2/day) Duration (weeks) Response (%) Ref.
13-cis RA
15 20–125 7–30 33 [125]
18 50–100 >8 16 [128]
15 100 8 20 [368]
10 100 8 30 [312]
8 20–100 6 50 [183]
33 20 8 9 [58]
30 50 >4 56 [205]
35 100 6 3 [195]
66 100 6 23 [28]
34 10–60 >12 (5 years) 12 [35]
ATRA
2 45 8–10 100 [392]
14 30–90 12 0 [15]
29 10–250 8 3 [15]
23 45 >4 13 [289]
10 45 6 50 [393]
Retinoid + others
13-cis RA + LD-Ara-C 2 [160]
13-cis RA + LD-Ara-C 3 [59]
13-cis RA + VCR 77 [103]
13-cis RA + Vit.D3 + IFNα 44 [151]
13-cis RA + Vit.D3 + LD-Ara-C 26 [152]
ATRA + G-CSF 40 [114]
ATRA + G-CSF + EPO + tocopherol 90 [235]
IFNα2 + thymopentin + LD-Ara-C 75 [391]
13-cis RA + Vit.D3 + 6-TH 52–61 [93]
ATRA + IFNα + G-CSF 23 [165]
6-TH, 6-thyoguanine; LD-Ara-C, Low dose Ara-C

small study [164], and clinical improvements were seen in 99% and >1% of APL patients, respectively. The trans-
using combinations with low-dose ara-C [349] or locations are usually reciprocal chromosomal transloca-
vincristine or 6-thioguanine [103]. Evidence of differ- tions, leading to creation of reciprocal hybrid receptor
entiation-induction was obtained in one case of a patient proteins (X-RARα and RARα-X). APL expressing
who achieved a complete remission with cytogenetic PML-RARα, NPM-RARα or NuMA-RARα are respon-
evidence of persistence of an abnormal clone in the sive to ATRA-induced differentiation effects with
marrow [349]. the exception of PLZF-RARα type APL that is resis-
Acute promyelocytic leukemias (APL) almost uni- tant to ATRA.
formly respond to retinoic acid treatment. APL consti- ATRA induces differentiation of APL blasts into ter-
tutes approximately 5–10% of all cases of acute minally differentiated granulocytic cells that is associated
myeloid leukemia (AML) and is characterized by M3 with clinical remissions. ATRA-induced differentiation
morphology of FAB classification and chromosomal of APL blasts requires expression of PML-RARα recep-
translocations fusing retinoic acid receptor alpha (RARα) tor protein [360]. PML-RARα can heterodimerize with
gene on chromosome 17 and one of four different genes, RXR or form homodimers and subsequently bind to
including promyelocytic leukemia (PML), promyelo- retinoic acid response element (RARE), located in the
cytic zinc finger (PLZF), nucleophosmin (NPM) or promoters of the ATRA-responsive target genes. ATRA
nuclear matrix associated (NuMA) gene [300]. The can bind to PML-RARα with an affinity comparable to
most common forms of translocations are t(15,17) RARα. In the absence of ligand, RAR-RXR in normal
(q22,q21) encoding PML-RARα and t(11,17)(q23,q21) blasts and PML-RARα-RXR heterodimers in APL cells,
encoding PLZF-RARα fusion receptor proteins, found recruit nuclear co-repressor proteins, N-CoR or SMRT,
538 Growth and differentiation factors as cancer therapeutics

and Sin3A or Sin3B which in turn form complex with of chemotherapy including an anthracycline plus cyto-
histone deacetylase enzymes (HDAC1 or HDAC2), sine arabinoside in patients with white counts exceeding
resulting in transcriptional repression or silencing [300]. 10,000/m2 at initiation of therapy. The clinical studies
The transcriptional suppression occurs because deacyla- conducted over the past years have determined that the
tion of histone protein creates conformational changes, combination of ATRA and chemotherapy gives better
limiting access and binding of transcription factors and survival than chemotherapy alone in newly diagnosed
RNA polymerase to related genes [198]. At physiologic APL; the relapses were less and complete remissions
concentrations of ATRA (10−9−10−8 M), the nuclear co- are slightly higher. These studies also revealed that
repressors protein and HDAC complex are dissociated maintenance treatment with ATRA, and possibly with
from RARα in normal blasts, which in turn results in low dose of chemotherapy, can further reduce the inci-
recruitment of co-activators with histone acetyltrans- dence of relapse [92, 171].
ferase (HAT) activity, such as steroid receptor coactiva- Although ATRA therapy is usually well tolerated, two
tor-1 (SRC-1), PCAF, p300/CBP, ACTR, TIF2 or P/CIP problems are frequently encountered. The first is leuko-
(Fig. 1b). Acetylation of lysine residues in the N-terminal cytosis, an increase in peripheral leukocytes to ≥20,000
of histones by HAT activity results in transactivation of cells/μL, which occurs in about half of the APL patients
responsive genes leading to differentiation. However, treated with tRA [106]. The second, more serious prob-
the physiologic concentration of ATRA does not cause lem, retinoic acid syndrome (RAS), develops in about
dissociation of nuclear co-repressors protein and histone 20–30% of APL patients treated with tRA. RAS is char-
deacetylase complex from the PML-RARα fusion acterized by high fever, respiratory distress, weight gain,
receptors in APL blasts, leading to differentiation block. pulmonary infiltrates, hypotension, pleural effusion, and
The co-repressor complex is dissociated from PML-RARα sometimes renal failure [70, 126]; findings at autopsy
at only pharmacological concentrations (10−7−10−6 M) show extensive infiltration of mature myeloid cells into
of ATRA, resulting in removal of transcriptional repression lungs, skin, kidney, liver, and lymph nodes. Although leu-
and transcription of genes related to differentiation. kocytosis is often present in RAS, one third of APL
In addition to release of transcriptional repression, the patients who have normal leukocyte counts also develop
other possible mechanism involved in ATRA effective- this syndrome. Steroids such as dexamethasone have
ness in myeloid cell differentiation include expression ameliorated RAS [106]; possibly by affecting leukocyte
of different class of genes [207, 212] including induc- activation, cytokine production, or endothelial reactions.
tion of expression of p21WAF1/Cip1 cyclin-dependent kinase Since RAS occurs only in patients with APL or AML
inhibitor [45], upregulation of C/EBP- [270], interferon who have been treated with ATRA, this syndrome is
regulatory factor-1 (IRF-1) [308], PDCD4 [418] and considered to reflect an aberrant interaction between
regulation of the localization of PML oncogenic domains maturing granulocytes and host tissues. Because the
(PODs) [403]. More interestingly, the treatment of APL clinical signs of RAS resemble those of endotoxin shock
cells with ATRA inhibited the synthesis of vascular and adult respiratory distress syndrome it has been sug-
endothelial growth factor that in turn resulted in decrease gested that ATRA treatment may affect the expression
in bone marrow microvessel density [185]. of cytokines by myeloid cells. Induction of IL-1 and
More recent results with acute promyelocytic leukemia G-CSF secretion by APL cells under the influence of
have clarified the role of ATRA in this disorder. In multi- ATRA may contribute to hyperleukocytosis in vivo. On
ple studies [54, 91, 364, 400], it is now clear that a dose the other hand, secretion of IL-1, IL-6, TNF-α, and IL-8
of 45 mg/m2/day given by mouth in one or two divided could contribute to the pathogenesis of RAS, since they
doses until complete remission followed by anthrocycline- are involved in leukocyte activation and adherence and
based combination chemotherapy for consolidation, have been implicated in the development of the adult
will induce a high response rate and improve survival in respiratory distress syndrome.
patients with APL. On the basis of these and other studies, No evidence of synergism of 13-cis-retinoic acid with
ATRA has recently been approved by the FDA as standard chemotherapeutic agents was found in the treatment of
treatment for APL. patients with chronic myeloid leukemia. Based upon
Although trans-retinoic acid is effective in the treat- in vitro observations that retinoic acid significantly
ment of APL, resistance is common with the return of reduced the recloning capacity of bone marrow myeloid
leukemia cells shortly after treatment in the absence of progenitors in vitro in certain cases of this disease, Arlin
effective programs of consolidation. The role of chemo- et al. [12] added retinoic acid to an intensive chemo-
therapy during induction treatment of APL is still therapy protocol including daunorubicin, cytosine
unclear. European approaches have utilized high doses arabinoside, and thioguanine, and in 17 evaluable patients
Kapil Mehta et al. 539

did not observe any increase in the incidence and dura- and does not bind to retinoic acid receptor-gamma. Am80
tion of true Ph chromosome-negative remission in the has been shown to be effective in patient with APL that
chronic phase of the disease. had relapsed from CR induced by ATRA. Of 24 evaluable
The future of retinoid therapy resides in (a) develop- patients, who received Am80, 6 mg/m2, orally alone daily
ment of analogues with enhanced differentiation inducing until CR, 14 (58%) achieved CR [435]. The interval from
action with reduced toxicity, (b) developing combinations the last ATRA therapy was not different between CR and
with either conventional chemotherapeutic drugs or other failure cases. The outcome was well correlated with the
differentiation-inducing agents, applying retinoids more in vitro response to Am80 in patients examined. Adverse
extensively as a maintenance therapy, and (c) developing events observed in patient treated with Am80 included
approaches to reduce toxicity such as use of liposome- retinoic acid syndrome, hyperleukocytosis, xerosis, cheil-
encapsulated retinoic acid. In this context, a recent study itis, hypertriglyceridemia, and hypercholesterolemia, but
suggested that tyrosine kinase inhibitor ST1571 could generally milder than those of ATRA, which all patients
augment the cyto-differentiating, antiproliferative, and had received previously. Am80 is effective in APL
apoptogenic activity of ATRA [120]. Moreover, in APL relapsed from ATRA-induced CR and deserves further
cell lines made resistant to ATRA, the STI1571 could clinical trials, especially in combination with chemother-
relieve the resistance. Similarly, the use of liposomal- apy. Am580, a stable retinobenzoic derivative and a
ATRA was shown to be superior to the oral ATRA in RAR-alpha agonist, is a powerful inducer of granulocytic
terms of maintaining higher plasma levels in animal maturation in NB4 and in freshly isolated APL blasts
models and in humans [86, 88, 238, 305]. These results [437]. After treatment of APL cells with AM580 either alone
demonstrated that encapsulation of ATRA in liposomes or in combination with granulocyte colony-stimulating
and I.V. administration generate better pharmacokinetic factor (G-CSF) at concentrations that are 10- to 100-fold
profile than oral ATRA by circumventing hepatic metab- lower than those of ATRA necessary to produce similar
olism of ATRA. effects. AM580 is more powerful than ATRA in modulat-
Evaluation of liposomal ATRA in phase I trial in ing the expression of differentiation antigens only in cells
patients with refractory hematological malignancies in which PML-RAR is present.
showed that in contrast to the decline in plasma AUC In addition, Fenretinide (N-(4-hydroxyphenyl) reti-
(area under the concentration time curve) of ATRA seen namide, 4-HPR), a synthetic derivative of ATRA [438],
3 to 4 days of initiation of oral ATRA, there were no dif- and nonretinoid compounds such as, 1,25-dihydroxyvi-
ference between the AUC on day 1 and day 15 following tamin D3 [138, 438, 440] and vitamin K2 in combina-
liposomal ATRA treatment [34]. In the same study, lipo- tion with ATRA [441] have been shown to be effective
somal ATRA was shown to be safe and toxicity profiles in inducing differentiation in ATRA-resistant APL cell
were similar to oral ATRA, although liposomal ATRA lines and in cases to whom ATRA cannot be used [442–
produced much higher AUC. I.V. administration of lipo- 444]. 4-HPR inhibits cell growth through the induction
somal ATRA (90 mg/m2) monotherapy was shown to be of apoptosis in hematopoietic malignancies and variety
effective in newly diagnosed APL patients, inducing of solid tumor cells including head and neck, cervical
PCR-negative molecular CRs in a high proportion of carcinomas, neuroblastoma, lung carcinoma and breast
patients [76, 86] These studies supported the hypothesis cancer cells [445–450]. Recently, 4-HPR received a sig-
that I.V. liposomal administration may improve activity nificant amount of attention because of its promise as
of ATRA by altering its pharmacological profile and chemopreventive and therapeutic activity in breast,
remain elevated following extended treatment, providing prostate and ovarian cancers in preclinical and clinical
a basis for long-term remissions in APL patients. models with minimal toxicity [451, 453]. The ability of
New synthetic retinoid compounds: These include 4-HPR to induce apoptosis in ATRA-resistant tumor
Am-80, Am580 and 4-HPR, CD437, MX3350-1 and cell lines, such as acute myeloid leukemia (HL-60R)
LGD1069 (Targretin, Bexarotene), a RXR selective retin- and promyelocytic leukemia (NB306) [446], neuroblas-
oid. Am-80, a synthetic retinoic acid receptor (RAR)- toma [449], head and neck carcinoma [455] and breast
specific agonist, has been successful in patients with cancer cells [456] suggested that 4-HPR acts indepen-
relapsed APL previously treated with ATRA, inducing dently of retinoid receptors to mediate its biological
CR in about 60% of patients in a limited study [435, 436]. actions. However, a potential role for RARs in 4-HPR
Am80 is approximately ten times more potent than ATRA induced growth inhibition or apoptosis has been sup-
in terms of induction of differentiation in vitro, and more ported by other studies [450, 457–459]. 4-HPR inhibits
stable to light, heat, and oxidation than ATRA. Am80 has cell growth through the induction of apoptosis rather
a low affinity for cellular retinoic acid binding protein, than differentiation in HL-60 and NB4 cells [103, 111].
540 Growth and differentiation factors as cancer therapeutics

Vitamin D Metabolites has also been observed when the HL-60 cell line and the
mouse macrophage-like J774.2 cell line were exposed to
and Analogs as Leukemia 10−7−10 M 1,25-(OH)2 D3 for 24–72 h [22]. In this context,
in vivo injection of 1,25-(OH)2 D3 into patients with
Differentiation-Inducing Agents malignant osteopetrosis corrected bone binding and
The term vitamin D is generally used to describe a num- resorptive deficiencies in the patients’ monocytes [22].
ber of chemically related compounds having common Koeffler et al. [191] have shown that 10−7 to 10−9 M
antirachitic properties, but differing in the rapidity of concentrations of either the active metabolite of vitamin D
their action and the conditions under which biologic (1,25-(OH)2 D3) or certain fluorinated analogs of vitamin
activity is observed. In humans, cholecalciferol (D3) D can induce normal human myeloid progenitors
produced in the skin and the fraction obtained from the (GM-CFC) in the presence of CSF to differentiate prefer-
diet undergo sequential hydroxylation reactions, first in entially to macrophages in vitro. Marrow cultures exhib-
the liver microsomes and then in the kidney mitochon- ited an absolute, not just a proportional, increase in
dria, resulting in the formation of 25-(OH) D3 and 1,25- macrophage colonies, indicating that the action was not
(OH)2 D3, respectively. The latter is thought to be the simply inhibition of granulopoiesis. The plasma concen-
active form of D3 in enhancing bone resorption medi- tration of 1,25-(OH)2 D3 in humans is approximately 7.7 ×
ated by osteoclasts and in enhancing absorption of cal- 10−11 M [148], and the concentration of 1,25-(OH)2 D3
cium and phosphorus by the intestine. In addition, it inducing macrophage differentiation of progenitors
appears to act on the kidney in concert with parathyroid in vitro is >10−9 M, raising the question of physiologic
hormone to promote calcium resorption. It was formerly relevance of this observation. Certainly, patients receiving
believed that 1,25-(OH)2 D3 was synthesized in placenta superphysiologic doses of 1,25-(OH)2 D3 have not been
and by calvarial cell suspension containing osteoclasts, reported to have monocytosis; likewise, patients with vita-
osteoblasts, fibroblasts, and endothelial cells [366]. The min D-resistant rickets have not been reported to have low
local production of active metabolites of vitamin D by monocyte or macrophage levels. Nevertheless, the possi-
target organs such as bone raises interesting questions as bility of local production of the active form of vitamin D
to the role of these tissues in mediating vitamin D action, by cellular components of the marrow microenvironment
and may provide indications of a new dimension to local [385], possibly even by tissue macrophages, raises the
actions of vitamin D metabolites on normal or leukemic possibility of much higher local concentrations of 1,25-
marrow cell function, as well as more conventional (OH)2 D3 in the environment of the progenitor cells than
aspects of mineral metabolism. Interest in vitamin D plasma levels may suggest. Furthermore, recurrent infec-
action in hematopoiesis was prompted by the observa- tions, impaired phagocytic function, and decreased mobil-
tions that osteoclasts originate by fusion of circulating ity of leukocytes have been shown in vitamin D-deficient
mononuclear precursor cells and almost certainly repre- states. This defect in phagocytic function has been reversed
sent one of the end-stage cells of mononuclear phago- by in vitro culture of macrophages with 1,25-(OH)2 D3
cyte differentiation [43, 177, 178, 376]. Like osteoclasts, [22, 332]. The immediate biological precursor of 1,25-
other mature, nonproliferating phagocytic mononuclear (OH)2 D3, 25-(OH) D3, is without biologic effect unless
cells, such as monocytes and macrophages, possess the used at 10- to 100-fold higher concentrations than its
capacity to attach to and degrade bone matrix [178]. hydroxylated metabolite. Thus, evidence that macrophages
Monocytes and macrophages possess receptors for 1,25- themselves have 1-hydroxylase activity and can synthe-
(OH)2 D3, and the culture of human monocytes with 10−8 size 1,25-(OH)2 D3 from its precursor suggests that this
M of this metabolite results in macrophage maturation molecule may also be a monokine with important local
[320]. In cultures of normal human bone marrow, 1,25- actions in recruitment and activation of macrophages
(OH)2 D3 induces extensive macrophage differentiation [332] and of osteoclasts, with which they share a common
[236]. Long-term cultures of human cord blood myeloid derivation from a marrow hematopoietic progenitor.
cells also terminally differentiate to monocytes and
macrophages with vitamin D [344]. Direct evidence of
1,25-(OH)2 D3 induction of osteoclast development has
been reported by Abe et al. [72], who demonstrated that Action of Vitamin D Metabolites
a 1.2 nM concentration of the metabolite induced exten- on Cancer Cells
sive fusion of mouse alveolar macrophages, and that in
the presence of the lymphokine macrophage fusion fac- Besides playing a crucial role in maintaining the calcium
tor, as little as 0.012 nM was active in producing multi- homeostasis in the body, 1,25-(OH)2 D3 can also affect
nucleated giant cells. Multinucleated giant cell formation cell growth and differentiation in several cell types,
Kapil Mehta et al. 541

including cancer cells. Based on these properties of and two-dimensional gel pattern of proteins close to, but
1,25-(OH)2 D3, it has been studied for its ability to not identical with, those of peripheral blood monocytes
inhibit and prevent the cancer growth. The results from [251]. Cell division apparently is not required for expres-
various clinical studies were though encouraging but sion of these differentiation features [94, 191].
due to the calcemic adverse effects the therapeutic win- The ability of low concentrations of 1,25-(OH)2 D3 to
dow of this compound was extremely narrow [36]. Abe induce macrophage differentiation of HL-60 is similar to
et al. [46] were among the first ones to demonstrate that that reported for phorbol esters; however, this is not due
1,25-(OH)2 D3 could induce macrophage differentiation to similar binding sites, since 1,25-(OH)2 D3 did not
of myeloid leukemic cells. Induction of phagocytes, compete for phorbol diester binding sites as measured by
lysozyme production, and locomotive activity were seen (3H)phorbol dibutyrate binding on HL-60 [275]. Variants
with concentrations as low as 10−10 M of the vitamin. of HL-60 have been developed which are resistant to dif-
The potency of this differentiation inducer was such that ferentiation induction by DMSO, retinoic acid, TPA, and
a 1,000-fold higher concentration of the next most 1,25-(OH)2 D3. One variant resistant to phorbol esters
potent inducer of M1 differentiation, dexamethasone, was also resistant to 1,25-(OH)2 D3 [275], suggesting
was required to achieve a comparable level of matura- that mutants that cannot be induced to differentiate can
tion. Simultaneous treatment of M1 cells with low, involve common events following the receptor binding
physiologic concentrations of 1,25-(OH)2 D3 (0.12 nM) stage. The possibility of synergism between 1,25-(OH)2
and dexamethasone (10 nM) induced a degree of differ- D3 and phorbol esters or other differentiation inducers is
entiation equivalent to the response obtained with higher definitely indicated in the case of HL-60, and has been
concentrations of either agent alone [249]. The isolation reported with retinoic acid and with DMSO since low
of two variant clones of M1, one resistant to dexametha- concentrations of 1,25-(OH)2 D3 in combination with
sone and the other to 1,25-(OH)2 D3, strongly suggests DMSO produced cessation of proliferation of HL-60
that these differentiation-inducing agents act in different cells within 2 days, as well as a greater expression of
ways and that combination therapy with both steroids differentiation [352]. Simultaneous treatment of HL-60
may be useful in reducing leukemogenicity [249]. with suboptimal concentrations of 1,25-(OH)2 D3 (0.12–
The mouse myelomonocytic leukemic cell line 1.2 nM) showed additive effects in reducing nitroblue
WEHI-3 is also inducible to macrophage differentiation tetrazolium, a common marker for monocyte-macrophage
when exposed to 1,25-(OH)2 D3 in a clonal assay sys- and granulocyte differentiation [250].
tem. The differentiation-susceptible D+ line was both The human monocytoid cell line U937 is also induced
growth-inhibited and macrophage-differentiated to the to macrophage differentiation with loss of plating effi-
50% level with 10−8−10−10 M 1,25-(OH)2 D3. Of particu- ciency in the presence of 10−10 M 1,25-(OH)2 D3 [245,
lar interest was the observation that a subline of WEHI-3 293]. Differentiation involves development of adher-
refractory to other differentiation-inducing agents was ence, macrophage morphology, lysozyme production,
exquisitely sensitive to 1,25-(OH)2 D3, with 50% growth capacity to reduce NBT, expression of β-glucuronidase
inhibition and macrophage differentiation seen with and alkaline phosphatase, Fc receptor expression, phago-
10−13 M vitamin. cytosis, and reactivity with antimonocyte-specific mono-
The first reports of the capacity of 1,25-(OH)2 D3 to clonal antibodies [293]. As in the case of HL-60, the
induce differentiation of human leukemic cells indicated differentiation of U937 is not blocked by inhibitors of
that HL-60 was growth-suppressed, and phagocytosis DNA synthesis, but is by the calcium ionophore A23187
and C3-rosette formation were markedly induced in a [175]. The existence of synergism between 1,25-(OH)2
dose-dependent manner over a range of 10−8 to 10−10 D3 and other biological response modifiers has been
[248]. Unfortunately, these investigators concluded established with retinoic acid. U937 cells can be primed
that HL-60 was induced to form granulocytes, as had by short incubation with 1,25-(OH)2 D3 to respond by
previously been observed for retinoids and DMSO, rather maturation to agents such as cAMP, PGE, and cholera
than the uniform pattern of monocyte-macrophage toxin, which alone do not induce differentiation [293].
differentiation reported in subsequent studies [189, 228, 1,25-(OH)2 D3 or retinoic acid plus dibutyryl cAMP
234, 236, 275, 352, 356]. HL-60 cells following their is effective in inducing a variety of differentiation mark-
treatment with 1,25-(OH)2 D3 acquire several phenotypic ers in U937. Their actions on insulin receptors were the
changes that were similar to the mature monoyctes/mac- opposite, however. 1,25-(OH)2 D3 increased the bind-
rophages. These maturation features were induced in a ing, while retinoid decreased the binding. This effect
dose-dependent (10−11 to 10−7 M) and time-dependent was specific for insulin, since the transferrin receptors
(1–6 days) manner, and resulted in a functional phenotype were reduced by both methods of differentiation [335].
542 Growth and differentiation factors as cancer therapeutics

Thus, changes in insulin receptors during maturation capacity of 1,25-(OH)2 D3, it was observed that preleu-
in vitro depend on the inducing agent and are not caus- kemic marrows exhibiting colony formation analogous
ally related to the differentiation process. to normal marrow were least responsive to 1,25-(OH)2
Other differentiation-inducing agents have been com- D3, whereas preleukemic marrows with cluster-forming
pared with 1,25-(OH)2 D3, which was the most effec- acute myeloid leukemia-type clonal growth were most
tive. The polar/planar compound HMBA was most responsive, being comparable to the clinical observa-
effective in reducing cell recovery, but did not induce tions. These observations suggest the interesting, per-
cell maturation. Retinoic acid-reduced cell and total haps surprising, conclusion that the more acute the
blast cell recovery with an increase in neutrophil dif- leukemia the more it is responsive to 1,25-(OH)2 D3.
ferentiation. Protein inducers of differentiation, α- and Koeffler et al. [191] have also shown that 1,25-(OH)2
γ-interferon, showed slight activity in reducing cell and D3 and two fluorinated analogs, 24, 24-F2-1,25-(OH)2
blast recovery, whereas a murine serum source of dif- D3, inhibited colony formation of marrow from patients
ferentiation factor (GM-DF) was highly effective in with ANLL (four cases) and chronic myeloid leukemia
inducing macrophage differentiation and reduction in (four cases) at concentrations of 10−8 M, with 50–80%
recovery of immature cells. 1,25(OH)2 D3 or IFN-γ of leukemic colony-forming cells assuming a mac-
decreased blast cells and increased macrophage differ- rophage-like morphology. This result is comparable to
entiation in suspension cultures of marrow from patients the report of Moore et al. [265], with the exception that
with myelodysplastic syndrome [380, 417]. in the latter study more patients were investigated and
As an alternative to the suspension culture technique the greater sensitivity seen in acute myeloid leukemia
for monitoring differentiation induction of fresh leukemic patients (50% inhibition at 4 × 10−12 M) may be explained
marrow, an agar cloning assay has been used, in which by the use of total clonogenic units measured (i.e., colo-
colony or cluster incidence was measured in 7-day cul- nies of >40 cells and clusters of 3–40 cells) rather than
tures of 105 leukemic marrow cells. Unlike the clonal restricting inhibition analysis to colonies of >40 cells.
assay for differentiation-induction of HL-60 or WEHI-3 The cells of the majority of patients with acute myeloid
cells, primary human leukemia cultures formed small leukemia do not form colonies of 40 cells, and the leu-
clusters of 10–20 cells (microclusters), 20–40 cells (mac- kemic cells in general form small clusters.
roclusters), small colonies with an excess of clusters A variety of metabolites and analogs of vitamin D3
(microcolonies), or colonies of normal size with a normal have been developed and tested for biological activity
cluster-to-colony ratios [148, 191, 265]. In most cases, and in vivo toxicity. Recent studies have extended bio-
the clonal growth was diffuse and colonies or clusters logical screening to leukemia differentiation systems.
failed to differentiate (with the exception of most patients HL-60 is induced to differentiate upon exposure to
with chronic myeloid leukemia and some patients with 10−7−10−10 M 1,24-(OH)2 D3 or 1,24R-(OH)2 D3 in a
preleukemia) [250, 332, 336, 382]. In view of this growth fashion comparable to that with 1,25-(OH)2 D3. A differ-
pattern, clonal differentiation can only be measured by ent analog of 1,25-(OH)2 D3 has been found highly
isolation and staining of individual clones, a laborious active in stimulating intestinal calcium transport and
and inexact procedure at best. As an alternative, 1,25- bone calcium mobilization in vitamin D-deficient rats
(OH)2 D3-induced reduction of leukemic cloning capacity [374]. This 24, 24-F2-1,25-(OH)2 is highly active in
was used as an index of differentiation. The legitimacy of induction of WEHI-3 and HL-60 differentiation and
this approach has been validated by studies showing that growth inhibition, with 50% activity at 10−14 M, thus
50% growth inhibition of proliferation generally approxi- making it the most active D3 analog tested in the leuke-
mated to 50% growth inhibition when 1,25-(OH)2 D3 or mic assay system [258]. Unfortunately, this analog is
its derivatives were used to induce differentiation. It considerably more toxic in vivo than is 1,25-(OH)2 D3.
should be stressed that this linkage between proliferation These compounds showed the same relative activities,
inhibition in clonal assay and differentiation induction in that normal marrow was always least sensitive to
generally does not hold true for other types of differentia- growth inhibition and acute myeloid leukemia marrow
tion agents. For example, inhibition of the colony growth most sensitive, with preleukemia and chronic myeloid
by most chemotherapeutic agents is not associated with leukemia occupying intermediate positions.
differentiation, and protein sources of differentiation 24-Homo-, and 26-homo-1,25(OH)2 D3 and delta [42]
activity may not influence primary leukemic cloning analogues were ten-fold more potent than 1,25(OH)2 D3
capacity, but only recloning capacity. in inducing differentiation of HL-60 [298]. The
In a more extensive analysis of heterogeneity of 24-homo-analogue was significantly less active in mobi-
responsiveness to the growth-inhibitory/differentiation lizing calcium from bone.
Kapil Mehta et al. 543

It is obvious that more extensive screening must be 1,25-(OH)2 D3 receptors in leukemic patients may be
undertaken to determine if the calcium-mobilizing predictive of the ultimate response of the patient to
activity and consequent toxicity as a feature of D3 adjunct therapy with 1,25-(OH)2 D3 [228]. This possi-
metabolites are invariably related to efficacy in induc- bility is supported by the observation that the equilib-
tion of leukemic growth inhibition and differentiation. rium dissociation constant of the 1,25-(OH)2 D3 receptor
Obviously, as compared with a long in vivo half-life, on HL-60 is close to the vitamin concentration, causing
low toxicity and retention of selective leukemia-cell 50% of HL-60 cells to reduce NBT [189]. Against this
differentiating activity would be highly desirable for view, there is the fact that the human myeloblastic leu-
clinical studies. Clinical trials with vitamin D have been kemic cell line KG-1 has approximately the same num-
limited because of intolerable hypercalcemia which ber of 1,25-(OH)2 D3 receptors as HL-60, yet cannot be
often develops with this compound. Some responses induced to differentiate by the vitamin [189].
have been seen in patients with myelodysplastic There is extensive evidence for a classic steroid
syndromes when treated at 2 mg/day [192]. With new receptor-DNA interaction for the 1,25-(OH)2 D3 recep-
analogues of vitamin D3 now available, a separation of tor, with selective binding of the receptor to A + T-rich
the effects of calcium metabolism and those on cell segments of double-stranded DNA. Franceschi [101]
differentiation is now possible and clinical investiga- demonstrated that the receptor can also interact with
tions are underway [301]. RNA as well as DNA. The physiologic significance of
this observation remains obscure. However, in other ste-
Receptors for Vitamin D and its roid hormone systems, steroids can influence certain
nontranscriptional processes, such as the stability of
Metabolites hormone-dependent mRNA, as well as posttranscrip-
The role of 1,25-(OH)2 D3 as an agent responsible for tional processing of secretory proteins, the regulation of
mineral homeostasis has been extensively studied. Its 5S RNA synthesis, and the processing of heterogeneous
mechanisms of action within target cells is retinoid and nuclear RNAs.
steroid hormone-like in that the binding of 1,25-(OH)2 Interaction of 1,25-(OH)2 D3 with receptors on normal
D3 to vitamin D receptor (VDR) induces conformational or malignant target cells results in variable and complex
changes in the VDR which promote heterdimerization changes in proliferation. A biphasic effect on the growth
with retinoid X receptor (RXR) and recruitment of a of breast tumor [116, 175] and osteosarcoma cells [229]
number of nuclear receptor coactivator proteins including has been reported, with physiologic concentrations of
the SCR family members (Fig. 2) [224]. All known 1,25- 1,25-(OH)2 D3 (10−10–10−11 M) stimulating growth, and
(OH)2 D3-responsive tissues, such as the intestine, bone, higher concentration (10−7–10−8 M) inhibiting it. In con-
and kidney, contain VDRs. In addition, many other trast, a uniform pattern of growth inhibition is seen with
tissues and cultured cells have been shown to possess the leukemic cell lines HL-60, U937 [294], M-1 and
1,25-(OH)2 D3 receptors, including some tumor cells. WEHI-3, and primary myeloid leukemia at concentra-
Breast cancer, melanoma, and osteogenic sarcoma cells tions as low as 10−12 M. No inhibition of normal marrow
possess a low density (8,000–15,000 per cell) of recep- CFU-GM colony formation is seen with 1,25-(OH)2 D3
tors with high affinity (Kd 10−10−10−11 M) for 1,25-(OH)2 except at very high concentrations of 0.1–1 μg/ml; growth
D3 [84, 100, 105, 229, 186]. Receptors with similar affinity stimulation of normal mouse and human CFU-GM
and density have been reported on human monocytes, colony formation can be observed.
malignant B and T leukemic cell lines, activated T-cells, Kuribayashi et al. [199] have reported on two variant
Epstein-Barr virus-transformed B-lymphocytes, and clones of HL-60, resistant to differentiation and growth
cell lines such as K562, HL-60 and U937 [228, 229, inhibition in the presence of 1,25-(OH)2 D3. One clone
320, 352]. Identification of 1,25-(OH)2 D3 receptors by was also unresponsive to phorbol ester, actinomycin-D,
specific immunochemical reactivity and selective chem- and DMSO. The variant clones were found to possess
ical dissociation has shown that nuclear binding of the reduced amounts of cytosol receptor protein, to which
vitamin receptor is a rapid event (minutes) in HL-60, 1,25-(OH)2 D3 was specifically bound, but the hormone-
whereas the cellular differentiation response is delayed receptor complex could be transferred to the chromatin
(6–7 days) [228]. This may be reconciled if the receptor acceptor site in both the wild-type and variant clones.
must be maintained within the nucleus over the long This would indicate that 1,25-(OH)2 D3 resistance is due
term. A differentiation-noninducible variant of HL-60 to a reduction in specific cytosol receptors.
has been reported to have only 8% of receptor copy Freake et al. [105] reported that whole chronic myeloid
numbers of the parent line, suggesting that assay of leukemic cells specifically took up 1,25-(OH)2 D3 with
544 Growth and differentiation factors as cancer therapeutics

high affinity (Kd = 3.6 × 10−11 M) and low capacity. fibrous tissue is also mediated by monocytes and mac-
Subcellular fractionation of labeled cells showed that rophages, and the number and activity of these cells are
binding was restricted to cytosols and nuclei; however, increased by 1,25-(OH)2 D3. Thus, the various actions of
chronic myeloid leukemic cells appeared to contain this vitamin contribute to a reduction in the collagen
both the receptor for 1,25-(OH)2 D3 and an unknown content; conversely, a deficiency of it may allow abnor-
substance that prevents its detection following the prep- mal accumulation of collagen in the marrow. In this
aration of cytosol. Cells from patients in chronic phase context, myelofibrosis in a rachitic infant regressed fol-
specifically bound more vitamin (18 fmol/107 cells) than lowing vitamin D therapy [66], and a group of rachitic
did those in the blastic phase (7 fmol/107 cells), or cells children with anemia and a blood picture typical of
from patients with acute myeloid leukemia (2.6 fmol/107 myelofibrosis also responded to vitamin D [415].
cells). From observing that only cells from patients with A novel immunoregulatory role has been proposed for
chronic myeloid leukemia responded to 1,25-(OH)2 D3 1,25-(OH)2 D3. Intracellular receptors that bind the vita-
by differentiation along the monocyte-macrophage min and sediment at 3.35 were not detected in “resting”
pathway, it was concluded that differentiation induction T or B lymphocytes, but T cells activated by Epstein-
was most likely dependent upon adequate levels of Barr virus produced the receptor, and the amount of the
receptors, and that intact cells rather than cytosol prepa- macro-molecule induced was the same as in normal
rations should be studied before cells of a particular monocytes. Since the D3-binding macro-molecule is
tissue are designated as receptor negative. In view of seen in actively mitotic cells, it may exert an antiprolif-
the heterogeneity of the cellular composition of chronic erative-differentiative influence in the immune system.
myeloid leukemia and the heterogeneity of morpho- 1,25-(OH)2 D3 at picomolar concentrations has also
logical type in blastic chronic myeloid leukemia been shown to inhibit production of the T-lymphocyte
(30% terminal transferase positive) and ANLL, general growth-promoting lymphokine interleukin 2 (IL-2).
conclusions on receptor display with such small groups Other metabolites of vitamin D3 were less effective, and
of patients should be treated with caution. their order of potency corresponded to their respective
The HL-60 genome contains several different onco- affinity for the 1,25-(OH)2 D3 receptor, suggesting that
genes, but only one, c-myc, is significantly amplified suppression of T-cell production of IL-2 was mediated
and transcribed at high levels (×20). Westin et al. [404] by this specific receptor [384]. 1,25-(OH)2 D3 may
reported that myc on mRNA is no longer present in selectively inhibit the action of IL-1 in stimulation of
HL-60 cells induced to granulocytic differentiation by thymocyte proliferation [273]. It also modulates GM-CSF
DMSO or retinoic acid. Reitsma et al. [330] have shown production by T cells by posttranscriptional reduction in
that 1,25-(OH)2 D3 reduced myc nRNA levels in HL-60 the half-life of GM-CSF, nRNA in mitogen-activated T
within 4 h of exposure to the vitamin/hormone, and this cells and T-cell lines [377].
change preceded the onset of other measurable pheno- In view of the well-established calcium-mobilizing
typic changes by at least 8 h. It remains to be determined effect of 1,25-(OH)2 D3 in its classic target tissues [226],
whether the altered transcription is the consequence of it is possible that the suppressive effect of this hormone
the hormone interaction with the c-myc promoter or a on IL-2 is mediated by an influence on calcium translo-
spectrum of promoters, each associated with a gene or cation and again indicates a physiologic role of this
gene cluster involved in determining cell phenotype. hormone in immunoregulation.

Action of 1,25-(OH)2D3 on other Aspects In vivo Effects of Vitamin D Metabolites


of Hematopoiesis Sato et al. [347] have evaluated the effect of 1-α-(OH)
Myelofibrosis with myeloid metaplasia is considered a D3 on the growth of two solid tumors transplanted sub-
neoplastic disorder in which fibroblast proliferation and cutaneously in mice. 1-α-(OH) D3 administered orally
collagen synthesis in the marrow are increased by plate- by stomach tube at daily doses of 0.1 and 0.2 μg/kg body
let-derived growth factor, or related substances, released weight for 114 days suppressed sarcoma 180 tumor
by neoplastic megakaryocytes [48]. It has been postu- growth by 37 and 64% respectively, without signifi-
lated that 1,25-(OH)2 D3 may inhibit the formation of cantly affecting serum calcium levels. Similar oral treat-
fibrous tissue (mainly collagen) in bone marrow and ment with 0.1 and 0.2 μg/kg of 1-α-(OH) D3 for 21 days
also may increase its degradation [237]. The hormone resulted in a 75–80% decrease in pulmonary metastases
also inhibits the proliferation of megakaryocytes that in mice injected subcutaneously with Lewis lung carci-
normally promote collagen synthesis. Degradation of noma fragments. While the data illustrate a potential
Kapil Mehta et al. 545

therapeutic anticarcinogenic effect of this metabolite, demonstrated virtual identity of cells from the induced
they should be considered as preliminary, since small granulocytic sarcomas in nude mice with respective
numbers of mice (three to six) were tested in each treat- cells from the cultured lines. The validity and reliability
ment group. of the living model have thus been established. Using
1-α-(OH) D3 and 1,25-(OH)2 D3 have marked effects this in vivo model, preliminary experiments were under-
on the growth and differentiation of cultured murine taken to evaluate the effect(s) of 1,25-(OH)2 D3 on the
myeloid leukemia cells (M1 cells), established from an development of these subcutaneous leukemic tumors
SL mouse with myeloid leukemia. These results, and the induced on nude mice from cultured cells. Based on the
fact that syngeneic SL mice inoculated with M1 cells all work of Hartmann and Moore [143], a dosage of 1 μg/
die of leukemia, prompted a recent evaluation of the ml of 1,25-(OH)2 D3 was chosen as potentially the most
in vivo effects of 1-α-(OH) D3 and 1,25-(OH)2 D3 by effective, but least toxic. The method of administration
Honna et al. [166]. Thrice-weekly intraperitoneal injections involved subcutaneous implantation of an osmotic
of picomole amounts of 1-α-(OH) D3 and 1,25-(OH)2 D3 minipump (OMP) at a site remote from the concurrently
considerably prolonged the survival time of syngeneic SL inoculated tumorigenic cells. One group of mice
mice inoculated with M1 cells. A similar, marked prolon- received only washed cultured leukemic cells, another
gation of survival time was observed in athymic nude received cells and an OMP containing only solvent, and
mice with M1 leukemia and treated intraperitoneally with a third group of mice received leukemic cells and an
1-α-(OH) D3. The results with the athymic mice sug- OMP delivering 1 μg/ml of 1,25-(OH)2 D3. All mice
gested to the authors that T-lymphocyte-mediated immune were weighed on alternate days and observed for tumor
responses were not directly involved in the effects of the development.
vitamin D3 metabolite. Serum levels of calcium and phos- Of the mice receiving cultured K562 or HL-60 cells
phorus were not significantly affected in the nude mice only, almost all developed granulocytic sarcomas within
given M1 cells and 1-α-(OH) D3 for 30 days, compared an average of 4–5 weeks after inoculation. A similar
with mice given M1 cells alone. result was seen in mice inoculated with cultured cells
Attempts to duplicate these observations in BALB/c from either of these lines that also bore implants of an
mice inoculated with syngeneic WEHI-3 myelomono- OMP containing only solvent. Granulocytic sarcomas
cytic leukemia cells proved unsuccessful [143]. Neither either failed to develop, or developed infrequently and
dexamethasone nor a combination of it with 1,25-(OH)2 (on the average) later in the group similarly inoculated
D3 prolonged the survival of WEHI-3 tumor-bearing with cultured cells from these lines, and bearing OMPs
mice [143, 166]. The disparity between the in vivo containing 1,25-(OH)2 D3.
observations with M1 and WEHI-3 leukemias could Since each mouse underwent the same priming and
have a number of explanations, which require further received the same number of cultured human leukemic
testing before dismissing the therapeutic potential of cells, it appears that the absence of, or delay in the
vitamin D derivatives in leukemia. The WEHI-3B grows development of, induced granulocytic sarcomas may be
more rapidly than M1, and 105 cells generally result in attributable to the influence of the administered 1,25-
100% mortality within 21 days. However, treatment (OH)2 D3 in the system. These results suggest that 1,25-
with conventional cytotoxic agents, such as Cytoxan, (OH)2 D3 might have an inhibitory effect on the
produces an increase in life span, even producing cures proliferative capacity of human leukemic cells in vivo.
in a dose-dependent fashion [143]. In retrospect, however, Whether the mechanism involves induction of terminal
the explanation may reside in the relative resistance of differentiation on the human tumor cell inoculum
the WEHI-3D+ line to 1,25-(OH)2 D3-induced growth requires investigation. The involved mechanism(s) of
inhibition (50% inhibition in vitro with 4 × 10−4 M), in inhibition are also of interest. In those few treated mice
contrast to the much greater sensitivity of the WEHI-3D−, demonstrating proliferation of xenogenic (human) leu-
the HL-60, M1, and fresh acute myeloid leukemic cells kemic cells, the possibility of emergence of 1,25-(OH)2
(10−9–10−12 M). D3-resistant clones should not be overlooked, and the
Potter and Moore [317] have extended in vivo studies availability of receptors for 1,25-(OH)2 D3 should be
with successful growth and maintenance of the human determined on cells derived from these tumors. Recently
myeloid leukemia lines U937, KG-1, and K562, as subcu- developed analogues of 1,25-(OH)2 D3 notably 1,25
taneously induced granulocytic sarcomas in nude mice. (OH)2-16-ene-23-yne-D3, were remarkably effective in
Studies involving routine and special histochemistry, “curing” mice bearing the WEHI-3B + leukemia [32].
enzyme histochemistry, immunocytochemistry, surface Differentiation was induced at dosages that did not
markers, cytogenetics, and electron microscopy have produce hypercalcemia.
546 Growth and differentiation factors as cancer therapeutics

Translating these observations into a clinical applica- 428]. The differentiation-inducing action has been most
tion, it is clear that in vitro screening for response to extensively analyzed in murine erythroleukemic (MEL)
1,25-(OH)2 D3 can be used to identify resistant and cells. Inducer-mediated differentiation is a multistep
susceptible patients with leukemia and preleukemia. process characterized by a latent period when a number
While it is unlikely that the vitamin could prove effec- of changes occur [232, 348]. These include alterations
tive on its own, it could be combined with conventional in ion flux, increase in membrane bound PKC, appearance
chemotherapy or used as maintenance therapy in patients of CA2+ and phospholipid-independent PKC activity in
achieving remission by conventional protocols. The the cytosol and modulation of the expression of genes
toxicity of 1,25-(OH)2 D3 in mice at dosages unable to such as c-myc, c-myb, c-fos and p53. Commitment to
produce leukemic regression in vivo would require pre- differentiation is seen within 12 h and increases stochas-
cise dose-response analysis in humans. In this regard, tically over 48 h and is associated with suppression of c-myb
1,25-(OH)2 D3 has been used effectively in patients with expression and a 10- to 30-fold increase in globin gene
post-menopausal osteoporosis [64]. Short-term treat- expression [232, 331]. The levels of ornithine decarbox-
ment (6–8 months) with 0.5 mg/day restored calcium ylase are also regulated by HMBA. HMBA induces a
absorption to normal, and calcium balance improved transient, genome-wide hypomethylation of DNA achieved
and the bone resorption rate decreased. With long-term by replacement of 5-methylcytosine with cytosine resi-
therapy (2 years) both bone resorption and formation dues [327]. This may be a necessary but not sufficient
rates increased. The lack of side effects in long-term step in triggering the whole program of differentiation.
treatment with 1,25-(OH)2 D3 provides a dosage guide- Superoxide dismutase activity is induced by HMBA in
line for studies in leukemia [111]. parallel with differentiation and enzyme levels are
Oral administration of 1-α-(OH) D3 in two patients directly related to the degree of cytosolic hemoglobin-
with AML and one with MDS was reported to reduce ization [24, 304]. Introduction of superoxide dismutase
the number of leukemic cells in the marrow. In a study into MEL cells with liposomes induces differentiation
of 18 patients with MDS, 1,25-(OH)2 D3 produced a par- as do other oxidative treatments (liposome amino acid
tial or minor response in blood in eight cases but with no oxidase, xanthine oxidase, potassium superoxide). In
significant improvement in blood or marrow blasts contrast, antioxidants inhibit HMBA-induced differen-
[192]. Seven of the patients developed leukemia before tiation [24]. The induction of superoxide dismutase in
or by 12 weeks of treatment and half the patients devel- MEL cells may also be a cellular response to oxidative
oped hypercalcemia. It was not possible to sustain serum stress from hemoglobin autooxidation.
levels of 1,25-(OH)2 D3 at levels necessary to induce Potential improvements in efficacy of HMBA may be
differentiation or growth inhibition without producing accomplished by changes in the chemical structure of
unacceptable toxicity. Future clinical trials await the use the inducing agents and by increasing the sensitivity of
of recently developed analogues with reduced calcium tumor cells to inducers of differentiation. MEL cell lines
mobilizing action and enhanced differentiating activity that have acquired low levels of resistance to vincristine
[32]. A further rationale for therapy with 1,25-(OH)2 D3 display a markedly increased sensitivity to HMBA
and its analogues is provided by the observation that the [231]. A series of hybrid increased polar/apolar com-
metabolic pathway of 1,25-(OH)2 D3 is defective in pounds have been produced that in certain instances are
patients with MDS or AML [30]. Bone marrow plasma more active than HMBA in vitro and whose chemical
levels of 1,25-(OH)2 D3, but not its immediate precursor structure makes it likely that they have different phar-
25-(OH) D3, was decreased significantly in 50% of macokinetics [231].
MDS and 30% of AML patients. The first Phase I trials of HMBA involved continu-
ous infusion of escalating doses of HMBA for 5 or 10
days in patients with refractory solid tumors [11, 83,
213, 340]. Dose limiting toxicity, specifically thrombo-
Polar-Planar Compounds cytopenia, was observed at 20–40 g/m2. The MTD of
as Differentiation Inducers continuously infused HMBA was 28 g/m2/day and aci-
dosis and CNS dysfunction were toxicities, as well as
Differentiation-inducing activity has been reported hemorrhage related to thrombocytopenia. The plasma
for various polar-planar compounds, most particu- half-life was 2.5 h and plasma levels of 1.42 nM or
larly DMSO, hexamethylene bisacetamide (HMBA), higher could be achieved [419]. In Phase I trials nasogas-
N-methylformamide and, cotylenin A [62, 63, 169, 427, tric or oral administration of HMBA at 30–36 g/m2/day
Kapil Mehta et al. 547

was also associated with thrombocytopenia and neutro- patients with acute myeloid leukemia, actinomycin D
toxicity. Attempts have been made to individualize was reported effective in differentiation induction in all 14
patient dosage based on plasma levels and clearance cases studied [169]. In all three cases in another study
rates of HMBA [64]. Plasma levels of 1.5–2.0 mM cytosine arabinoside [246] and 5-aza-2-deoxycytidine
could be sustained but toxicity was seen. The cause of [314] were also effective in inducing macrophage
the thrombocytopenia is obscure but appears to be a differentiation of leukemic blast cells. In the absence of
production defect rather than a peripheral destruction an in vivo measure of leukemogenicity, the ability of
or pooling of platelets. At the dosages of HMBA these agents to eliminate the proliferative potential of the
required to produce differentiation, significant inhibi- leukemic clone must be measured indirectly. In reclon-
tion of cloning of normal myeloid, erythroid, and mega- ing studies of primary human leukemias cultured in
karyocyte progenitors is seen in vitro in murine bone methylcellulose, a decrease self-renewal potential of clo-
marrow culture. Fifty percent inhibition of cloning, and nogenic cells (plating efficiency 2, PE2) can be observed
suppression myelopoiesis in long-term bone marrow with various chemotherapeutic agents, independent of
cultures, are seen with doses of HMBA of 1.2 mM with their chemosuppressive action on primary leukemic
an unusually steep dose response. This observation cloning efficiency [41]. This action on PE2 is a likely
does not fully explain the observed clinical toxicity, differentiation index, since more primitive leukemic
since the thrombocytopenia was not usually associated stem cells with extensive proliferative potential are
with a neutropenia. The development of analogs of “differentiated” into a non-self-renewing compartment.
HMBA with differentiation-inducing capacity and Furthermore, this PE2 parameter correlated with clinical
reduced suppressive activity against myeloid progeni- response [42]. The combination of low doses of the
tors is a like direction in development of an effective above-mentioned chemotherapeutic agents with other
clinical differentiation protocol [231] that could be differentiation-inducing agents may prove effective
applied in leukemia and MDS as well as in a variety of when neither type of agent can induce differentiation
solid tumor systems (colon, bladder, breast). directly. In this regard, differentiation-resistant clones of
M1 (D−) that failed to respond to protein differentiation
factor or chemotherapeutic agents could be ‘sensitized’
by as little as 0.25 μg/ml of actinomycin D, daunomycin,
Chemotherapeutic Agents mitomycin C, hydroxyurea, bleomycin, 5-fluorouracil,
as Differentiation Inducers prednisone, or dexamethasone to terminal differentiation
when combined with protein factor [149, 169].
It is becoming apparent that of the variety of agents uti- Lotem et al. [219] report that in vitro screening for
lized in cancer chemotherapy, some may be effective differentiation-inducing compounds and compounds that
because of their capacity to induce, selectively, tumor show toxicity to blast cells may be useful in selecting
cell differentiation, and this property may be more appropriate treatments. However, their results were
important than cytotoxic potential. After extensive ambiguous, in that with some patients studied both before
screening (for reviews, see [169, 189, 343]) a number of and after in vivo chemotherapy there was a similar dif-
agents have been found to have differentiation capacity ferentiation response, or an apparent loss, or a gain of
[63, 65, 169, 214, 246]: mitomycin, doxorubicin, bleo- response in vitro of the remaining leukemic cells tested.
mycin, daunomycin [169, 189, 214, 246], cytosine arabi- In further studies, using five compounds known to induce
noside [130, 167, 169, 189, 214, 274, 369], hypoxanthine HL-60 differentiation (DMSO, hexamethylene bisacet-
[63], 3-deazauridine, 5-azacytidine, methotrexate [33, amide (HMBA), hypoxanthine, actinomycin D, and
351], 5-aza-2-deoxycytodine [51, 57, 254, 314], aphidi- 6-thioguanine), differentiation of fresh myelogenous leu-
colin [125], and adenine arabinoside [274]. kemic cells was tested in vitro [192]. Of 12 patients studied,
In all the preceding cases the differentiation action the blast cells in most cases showed little morphological,
was observed at concentrations either below cytotoxic histological, or functional maturation after exposure to
levels or well below maximum growth inhibition in sus- the various compounds, as compared with the blast cells
pension or clonal assays of leukemic cells. Most com- cultured without the compound. Actinomycin D was the
pounds have been tested against murine (M1) and human only agent capable of causing significant maturation. This
(HL-60) myeloid leukemic cell lines, in which differen- study suggests that many compounds shown to differenti-
tiation into macrophages and granulocytes is reported. ate HL-60 may not trigger differentiation of less mature
When tested in vitro against leukemic blast cells from myeloid leukemic cells.
548 Growth and differentiation factors as cancer therapeutics

Molecular Mechanism Implicated The mechanism by which 5-aza-deoxycytidine induces


leukemic cell differentiation most likely involves DNA
in Leukemia Cell Differentiation
hypomethylating ability [254, 314]. Synthesis of hypom-
Cytosine arabinoside is one of the most effective single ethylated DNA takes place after incorporation of the
agents for the treatment of myeloid leukemia and is con- drug into DNA and is due partially to the chemical
ventionally considered to act by incorporation into DNA structure of the compound and mainly to the trapping of
and by inhibiting DNA replication through production DNA methyltransferases, thus blocking their action.
poor primer termini. Cytosine arabinoside induces non- The known differences regarding the molecular targets
specific esterase activity in HL-60 cells [107] and increases of the two drugs could account for the greater differen-
surface expression of the monocyte surface antigen MY-4. tiation-inducing ability and lower toxicity of the 2-deoxy
Aphidicolin, an analogue of deoxycytodine, also induces derivative. It is well known that 5-azacytidine is actively
HL-60 differentiation and slows DNA synthesis, but, incorporated into mRNA and tRNA, thus producing its
unlike cytosine arabinoside, it is not incorporated into major toxic effects.
DNA and acts as an inhibitor of DNA polymerase [130]. A cautionary note should be introduced in considering
Using a purine rather than a pyrimidine antimetabolite- the potential therapeutic role of 5-azacytodine. Motojo
adenosinearabinoside, Monroe et al. [274] also observed et al. [271] exposed blast cells from patients with acute
differentiation of HL-60 that correlated with slowing of myeloid leukemia to 5-azacytidine, 6-azacytidine, and
DNA synthesis by an agent known to act at the level the 2-deoxy derivative. Simple negative exponential
of the DNA polymerase template complex. The relation- colony survival curves were obtained for the three drugs,
ship of differentiation to DNA synthesis is complicated with the 5-aza-2-deoxy compound being most toxic and
by observations that terminal differentiation of HL-60 the 6-aza least toxic. Although confirming other reports
to macrophages induced by the tumor promoter TPA can of increased expression of certain antigenically defined
occur in the absence of DNA synthesis [339] and that phenotypic markers of leukemic blast cell differentiation,
terminal differentiation to granulocytes without cell colonies surviving drug exposure to 5-aza and 5-aza-2-
division is observed following treatment with actinomy- deoxy compounds had increased secondary replating
cin D, DMSO, and butyric acid [236]. efficiency. This suggests that hypomethylation of DNA
Agents can, however, induce HL-60 differentiation may promote leukemic cell self-renewal.
without inhibiting cell proliferation [33], or inhibit pro-
liferation without inducing differentiation [351]. Thus,
it remains to be determined whether inhibition of DNA
In vivo Induction of Differentiation with
synthesis is causally or indirectly related to differentia- low-dose Cytosine Arabinoside or
tion. Other mechanisms of action of chemotherapeutic Azocytidine in Patients with acute
agents directed toward differentiation induction may Myeloid Leukemia and Myelodysplastic
involve cell membrane effects. Anthracyclines may play
Syndrome (MDS)
a role in alteration of leukemic cell glycoproteins, and
there is evidence that these drugs bind extensively to The preceding in vitro observations provide a strong
membrane-lipid domains, altering membrane fluidity case for the efficacy of low doses of ara-C in induction
and directly modifying the synthesis or expression of of terminal differentiation of myeloid leukemic cell lines
glycoproteins at the cell surface [350]. and fresh leukemic blast cells, this action being either
A more specific action has been suggested involving direct or by synergy with endogenous differentiation-
DNA methylation changes: for example, cytosine arabi- inducing factors. It was thus of interest to investigate the
noside may have a direct influence on methylation of actions of low-dose ara-C in patients with acute myeloid
the c-myc oncogene [130], the expression of which is leukemia and myelodysplastic refractory anemia with
considerably amplified in myeloid leukemic cells and excess of blasts. Baccarani and Tura [17] reported the
rapidly reduced once the cells are exposed to differenti- first remission in a patient with RAB and subsequently
ation-inducing agents [330, 404]. extended the study to 20 patients with myelodysplastic
5-Azacytidine and the less toxic 2-deoxy derivatives are syndrome (MDS), observing one complete and two par-
also interesting candidates for in vivo use in leukemia tial remissions. Complete or partial remissions in MDS
differentiation therapy. These compounds have been have been reported by other groups [167, 253, 318, 402,
shown to trigger gene expression in several systems, 409], but no response to low-dose-ara-C was observed in
including globin gene expression, when given in vivo a large cooperative study of refractory anemia with
to thalassemic and sickle-cell anemia patients [206]. excess of blasts [280]. In acute myeloid leukemia,
Kapil Mehta et al. 549

Housset et al. [168] reported remission in all three patients [71]. In other recent studies of low-dose ara-C
patients, and Weh et al. [402] had seven complete and in 73 patients with ANLL and MDS [10] and 40 patients
two partial remissions in 12 cases. Encouraging results with ANLL and MDS, complete remission rates of 24–31%
have been obtained by others [253], but no response was were reported with 35–45% responders. In these studies
reported by Hagenbeek et al. [136] in four acute myel- evidence for leukemic cell differentiation was obtained.
oid leukemia patients treated with the same protocol that Cytogenetic and morphologic studies suggested that
produced responses in other studies [168, 402]. The rea- cytotoxicity rather than differentiation was responsible
sons for variable results are unclear, but the patient pop- for remissions observed in two other studies with low-dose
ulation, particularly the MDS cases, was heterogeneous, ara-C in poor-risk ANLL [19, 49].
and the dosage and timing of drug administration in dif- Low-dose 5-azacytodine has also proved effective in
ferent studies varied between 10 and 30 mg/m2, every 12 or MDS, and in one study of 44 patients, 48% responded
24 h, for 7–28 days. Indeed, the more encouraging results with 11% complete and 25% partial remissions [357].
obtained by Housset et al. [168], compared with those The median duration of remission was 53 weeks. 5-Aza-
of Baccarani et al. [16], may be due to the more frequent 2′-deoxycytidine in 134 patients with AML, CML in
and longer duration of ara-C administration in the former blastic crisis, and MDS produced two complete and four
study, thereby probably achieving a rather constant in partial remissions. The drug appeared to modify the leu-
vivo concentration of the drug. kemic phenotype in vivo as well as producing a direct
In one series of 21 patient (five with refractory anemias cytolytic effect [423].
with an excess of blasts and 16 with acute leukemias) Tiazofurin (2-beta-D-ribofuranosylthiazole-4-carbox-
treated with small doses of ara-C (10 mg/m2/12 h for amide) is an inhibitor of inosine5-monophosphate (IMP)
15–21 days), improvement was noted in 15 cases (71%), dehydrogenase and is the enzyme in the rate-limiting
and complete remission was observed in 12 (57%) [47]. synthesis of guanylate [401]. Tiazofurin has both cyto-
Complete remission was obtained after one course of toxic and cytodifferentiation activities and has been
treatment in eight cases. The fact that these patients used to induce responses in patients with blast crisis in
entered remission relatively slowly and did not suffer CML [381]. Toxicity has been severe with nausea, rash,
marrow aplasia suggests that low-dose ara-C was func- myalgia and serositis. Given the high toxicity, further
tioning by inducing differentiation. Generally, when clin- clinical trials to this approach must be conducted to
ical response has been obtained, the evidence points to a determine optimal dose and schedule. Combinations of
differentiating role for the drug rather than an antitumor tiazofurin with other differentiations may be of interest
effect, with progressive evolution of recovery, absence of to investigate in the near future.
aplasia, and the simultaneous presence of normal islets of
promyelocytes and leukemic myeloblasts [168].
The availability of the in vitro assay for detection of
leukemic cell responses to differentiation agents should Cytokines and Hematopoietic
mandate prescreening of patients for in vitro sensitivity
before enrollment in a differentiation protocol such as
Growth Factors active in Regulating
low-dose ara-C. Toward this goal, leukemic marrow Proliferation and Differentiation
cultures were exposed to low doses of ara-C, and a
reduction in blast cells and an increase in more mature
of Leukemic Hematopoietic Cells
granulocyte and macrophage elements were noted in Recognition that physiologic inducers of differentiation of
two or three patients. Subsequent low-dose ara-C treat- normal hematopoietic cells could also influence prolifera-
ment in vivo resulted in complete remission in the two tion and differentiation of leukemic cell lines led to a series
patients that showed a strong in vitro response [246]. of studies over the last 2 decades involving characterization
In a large study of low-dose (10 mg/m2) ara-C in of hematopoietic growth factors, in many cases using leu-
poor-risk ANLL, overall survival was comparable to kemic cell lines as sources of growth factor and/or as target
that observed following conventional therapy with high- for growth factors in various bioassays. As discussed in
dose (200 mg/m2) ara-C and anthrocyclin [71]. While the other chapters, the genes for these factors have been cloned
high-dose regimen produced more complete remissions and recombinant factors have been tested in vitro and in
(55%) than the low dose (33%), there were more early vivo for biological activity. This large family of cytokines
deaths in the intensive therapy group. Very-low-dose and polypeptides includes factors with pleotropic and over-
(3 mg/m2) ara-C produced hematologic improvement in lapping activities, and additive or synergistic interactions
the majority of patients in a study trial of 73 MDS are frequently observed [139, 209, 321, 426].
550 Growth and differentiation factors as cancer therapeutics

CSF-Dependence of Myeloid Leukemic producer cells through functional external receptors.


Support for the concept has been obtained by studies on
Progenitors
oncogene action, since oncogenes may confer growth-
The cloning of normal or leukemic human CFU-GM in factor autonomy on cancer cells by coding directly for
either agar or methylcellulose has permitted analysis of autocrine polypeptide growth factors or their receptors,
both quantitative and qualitative changes in this cell or by amplifying the mitogenic signals generated as a
compartment in leukemia and other myeloproliferative consequence of growth factor-receptor interaction.
disorders. Changes observed include abnormalities in Strong evidence for an autocrine role for GM-CSF
the maturation of leukemic cells in vitro, defective pro- has been provided in a study of 22 cases of primary
liferation as measured by colon size or cluster-to-colony human acute myeloid leukemia in which Northern blot
ratio, abnormalities in biophysical characteristics of leu- analysis revealed expression of the GM-CSF gene in 11
kemic GM-CFC, the existence of cytogenetic abnor- cases [420]. GM-CSF expression was not found in nor-
malities in vitro, and regulatory defects in responsiveness mal hematopoietic tissue, with the exception of acti-
to positive and negative feedback control mechanisms vated T-cells, strongly indicating that the GM-CSF gene
(for reviews, see refs. [255–258, 261, 263–265]). activation was intimately associated with the transfor-
Detection of this spectrum of abnormalities has proved mation event. Furthermore, in six cases, GM-CSF was
to be of clinical use in diagnosis of leukemia and preleu- secreted by leukemic cells of both early and late stages
kemic states, in classification of leukemias, and in pre- of differentiation, and activity was specifically neutral-
dicting remission in acute myeloid leukemia. Variation ized by antiserum to GM-CSF [129].
has been reported among different groups investigating The paradox of CSF dependence of primary human
the characteristics of human acute myeloid leukemia myeloid leukemias in clonal assay, and the autocrine
cells in culture. These differences reflect, in part, the production of GM-CSF, can be answered in part by the
heterogeneity of the disease as well as variation in concentration at which the cells are plated: at high con-
the culture criteria, the source and activity of CSF, and the centrations, ‘spontaneous’ leukemic colony or cluster
timing of the culture. The preceding studies indicated formation is the norm. In the study of Young et al. [420],
that leukemic cells from patients with acute or chronic 9 of 22 cases showed autonomous growth of leukemic
myeloid leukemia or preleukemic states were absolutely clusters when cells were plated at 5 × 104/ml, yet exog-
dependent upon a source of stimulatory factors for their enous GM-CSF increased cloning number and clone
clonal proliferation in culture at low plating densities. size in 15 of 22 cases. It is possible that in some cases
Analysis of the dose response of myeloid leukemia GM-CSF is not actually secreted by the leukemic cells,
CFU-GM indicates that in the majority of cases they do but is present in active form as a membrane-bound moi-
not differ markedly from normal in their responsiveness ety. In this regard, Nara and McCulloch [281] showed
to various sources of CSF, although occasionally, hyper- that purified cell membranes from cells of some acute
responsiveness of leukemic cells is seen [255, 257, 258, myeloid leukemia patients, but not normal bone marrow
261, 263, 265]. of ALL cells, could promote proliferation of other acute
In an analysis of growth-factor responsiveness in sus- myeloid leukemic cells in short-term suspension cul-
pension cultures of leukemic marrow from 25 patients, ture, enhancing self-renewal.
Lowenberg et al. [220] showed that 17/25 cases exhib- A direct link between constitutive GM-CSF expres-
ited spontaneous “autocrine” proliferation, [362] and sion and leukemic transformation was provided by Lang
this was enhanced in 21 cases by IL-3, in 17 cases by et al. [202], who transfected the murine GM-CSF gene
GM-CSF of G-CSF, and in five cases by M-CSF. In four into CSF-dependent, nonleukemic, myeloid cell lines
cases the cells responded to all factors and the remain- and produced leukemic cell lines that constitutively
der responded to three, two, or only one source of stimu- secreted GM-CSF. To date, no example of human myel-
lus. As with normal progenitor assays, “spontaneous” oid leukemia constitutively producing and responding
leukemic cloning is observed as the marrow cell or to IL-3 has been reported, but in the mouse system, the
peripheral blood leukocyte plating density increases, well-characterized spontaneous murine myelomono-
owing to endogenous production of CSFs by accessory cytic leukemia WEHI-3 constitutively secretes IL-3,
populations, which may be residual normal cells of apparently because of insertion of retroviral sequences
T-cell lineage or leukemic cell subpopulations. The adjacent to the IL-3 gene [416].
autocrine concept of malignant transformation proposes Retroviral insertion of the IL-3 gene with a viral long
that cells become malignant by the endogenous produc- terminal repeat promoter produces leukemic transfor-
tion of polypeptide growth factors that act upon their mation [140]. The fact that insertion of G-CSF, GM-CSF,
Kapil Mehta et al. 551

or IL-3 genes into normal hematopoietic stem cells or in establishment of continuous lines of AML cells that
transgenic mice did not lead to leukemia [240] suggests grew for >2 months [67].
that autocrine induction of factor-independence is nec- A more complex autocrine loop is suggested by the
essary, but not sufficient, for converting normal cells to studies of Bradbury et al. [36]. AML cells at low cell density
a transformed state. Genetic alterations may predispose were stimulated independently by exogenous GM-CSF
them to undergo leukemic transformation by an auto- or IL-1, and the response to both factors was inhibited by
crine mechanism. In this context the murine factor- antibody to GM-CSF. Antibody to IL-1 inhibited sponta-
dependent cell line FDC-P1 develops into fully leukemic neous proliferation of these cells, and endogenous
cells when transplanted into whole-body-irradiated mice GM-CSF could be detected. Thus, IL-1 is produced by
associated with development of autocrine GM-CSF or the leukemic cells, particularly from the more differenti-
IL-3 production [80, 81]. ated cells, and this in turn induces GM-CSF production,
Expression of mRNA for GM-CSF, G-CSF and which directly mediates autocrine proliferation. Autocrine
M-CSF is ubiquitous in the blast cells of AML but not IL-1 induction of GM-CSF may account for the appear-
CML [55, 285, 397, 420, 421]. In a minority of these ance of GM-CSF mRNA in cultured rather than fresh
cases significant quantities of bioactive CSF were pro- AML blasts [180]. Therapeutic strategy in the face of
duced, sufficient to stimulate autocrine blast cell prolif- IL-1, GM-CSF-, or IL-3-dependent leukemogenesis,
eration [329, 420]. The significance of these observations either autocrine or paracrine, would require intervention
was questioned by studies showing that enhanced expres- to block CSF or IL-1 production or action. High-affinity
sion of the GM-CSF gene in ABL blast cells was a con- antibodies to the factor or its receptor, in the latter case
sequence of manipulations used to enrich blast cell coupled to some form of toxin, may be one possible strat-
populations [180, 367]. egy which is under active consideration as a therapy in
Constitutive expression of IL-1 on mRNA and bioac- the case of IL-2-dependent T-cell lymphoma (anti-IL-2
tive protein production is a characteristic of the majority receptor antibody and IL-2 toxin conjugates). A second
of AMLs and is not a consequence of manipulation of strategy envisages the use of GM-CSF to recruit resting
the cells. IN 10/17 cases of AML, IL-1β mRNA was leukemic stem cells into cell cycle and synchronize the
identified [131], and this autocrine IL-1 can induce population in S phase in conjunction with cycle-specific
AML proliferation [247]. The proliferative effect of chemotherapy [87, 217, 322].
IL-1 on leukemic blast cells, like its action on normal
cells, is mediated in synergy with growth factors such as Colony-Stimulating Factors as Leukemia
IL-3, GM-CSF, and G-CSF [161, 162]. Since IL-1
induces G-CSF, GM-CSF, and M-CSF production by
Differentiating Agents
bone marrow stromal cells (endothelium, fibroblasts, The G-CSF and GM-CSF induce granulocytic differen-
macrophages; see Moore [260] for review), and these tiation of HL-60 and WEHI-3B leukemic cell lines
factors stimulate leukemic cell proliferation, a paracrine [24, 25, 108, 315], but leukemic blast-cell proliferation
mode of stimulation of leukemic cell proliferation has is the more general response in primary cultures of AML
been proposed for AML [131], and juvenile CML [18]. bone marrow. GM-CSF is a universal proliferative stim-
An IL-1-mediated autocrine growth stimulation was ulus for AML cells, generally without differentiation,
proposed in a study of 13 cases of AML [67]. In all whereas G-CSF stimulates proliferation and in a
cases, immunofluorescence showed that up to 80% of proportion of cases granulocytic differentiation [73].
all fresh leukemic blast cells in all patients contained This area is reviewed elsewhere in this volume and the
either the 33-kDa IL-1β propeptide or both the 33- and actions of G-CSF and GM-CSF in clinical trials in
17-dDa mature form. The bioactive IL-1α propeptide myelodysplastic syndrome and AML are discussed.
was also detected in all cases but was less frequently M-CSF is also a proliferative stimulus for some AML
released. In six cases studied, anti-IL-1β and to a lesser cells, but in many instances it induces differentiation to
extent anti-IL-1α inhibited spontaneous proliferation, adherent macrophages [397]. Most leukemic blast pop-
and in 10/12 cases sufficient exogenous IL-1 was pro- ulations express the M-CSF receptor, fms, and M-CSF
duced to stimulate significant proliferation. AML cells mRNA is found in about half the blast population.
constitutively released as much IL-1 as did endotoxin- Marked heterogeneity of M-CSF response is seen with
stimulated normal monocytes, and 2/12 patients that did different patients, ranging from cases where blast popu-
not respond to exogenous IL-2 were both high endoge- lations were unresponsive to M-CSF, to examples where
nous producers and presumably were maximally stimu- blast-cell self-renewal was stimulated, to instances
lated. In most instances, exogenous IL-1 supported the where the major effect of M-CSF was the generation of
552 Growth and differentiation factors as cancer therapeutics

terminally differentiated cells with monocyte-mac- chamber. A more likely mechanism implicates IL-1
rophage characteristics. This heterogeneity in prolifera- induction of autocrine production of IL-6 by leukemic
tive versus differentiation responses of leukemic blasts cells, and this was observed within 6 h in IL-1-treated
to various CSF species in different patients indicates the M1 cells, with production increasing by 2–3 days [215,
necessity of prescreening for factor response on an indi- 216]. The indirect differentiation induction mechanism
vidual patient basis. was blocked using antibodies to IL-6. As with the auto-
crine IFNβ route for differentiation, the endogenous
IL-6 may require a synergistic interaction with IL-1 to
Interleukin 1 and Cell Proliferation and produce optimal differentiation. These results point to
Differentiation the importance of considering combinations of cytokines,
Interleukin 1 has been implicated in an autocrine or para- both exogenous and endogenous, in designing optimal
crine mode of stimulation of myeloid leukemic cells in differentiation strategies.
synergy with GM-CSF [36, 67, 131, 161, 162, 247]. The ability of IL-1 to induce certain leukemic cells to
It has also been reported to inhibit proliferation of a human differentiate in association with growth inhibition
myelomonocytic leukemic cell in the presence of low to involves synergistic interactions with other cytokines,
intermediate, but not high, levels of GM-CSF [346]. some of which are produced by leukemic cells in response
The IL-1 inhibition of tritiated thymidine incorpora- to IL-1. In normal hematopoiesis, IL-1 also induces a
tion into murine M1 cells was first reported by Onozaki spectrum of hematopoietic growth factors (G-CSF,
et al. [295]. On its own, IL-1 did not induce differentia- GM-CSF, M-CSF) by an action on endothelial cells,
tion, but both growth inhibition and macrophage differ- fibroblasts, and macrophages and acts synergistically
entiation were induced synergistically by IL-1 and LPS with these and other (e.g., IL-3) hematopoietic growth
coadministration. In further studies these investigators factors to stimulate proliferation of early hematopoietic
reported that while IL-1α, TNFα, and IFNβ1 all had stem cells [65, 79, 262, 266]. In vivo IL-1 alone or in
antiproliferative, but not differentiation-inducing, action combination with G-CSF was particularly effective in
on M1 cells, as little as 1 unit of IL-1, in conjunction accelerating myeloid regeneration following high-dose
with TNF or IFNβ1, induced FcR, phagocytic activity, chemotherapy or radiation [259, 262].
and morphological charge [297]. The differentiation
induced by IL-1 plus TNF was inhibited by antibodies Interleukin-6 and Leukemic Cell
to IFNβ1, as was the antiproliferative effect of TNF (but
not IL-1), indicating autocrine IFNβ1 is induced and
Proliferation and Differentiation
mediates direct differentiation and antiproliferative Interleukin 6 promotes the terminal maturation of B
effects. Thus IFNβ1 is one of two signals required for cells to antibody-producing cells, augmenting IgM, IgG,
M1 differentiation, the other being IL-1, and the anti- and IgA production in stimulated B cells [184]. It was
proliferative effect of IL-1 appeared to be a direct one. originally identified as novel fibroblast-type interferon
The differentiation-inducing factor for M1 cells in LPS- (IFNβ2) and was found to be identical to a B-cell dif-
stimulated peritoneal macrophage conditioned medium ferentiation factor, BSF2 [158], a hybridoma/plasmacy-
was identified as TNF, synergistically active with IL-1 toma growth factor [390], and a macrophage-granulocyte
[372]. Interleukin 1 induction of another cytokine, inducer of leukemic cell differentiation (MGI-2) [354].
MG1-2 or IL-6, has also been implicated in IL-1 induc- Its action on normal hematopoiesis includes stimulation
tion of M1 differentiation. Lotem and Sachs [215, 216], of multilineage blast cell colonies in synergy with IL-6
in contrast to the results of Onozaki et al. [297], showed [172]. It may act to induce the entry of Go stem cells
that IL-1 on its own induced M1 differentiation as mea- into cell cycle [197]. Interleukin 6 potentiates thrombo-
sured by FcR and C3R induction, lysozyme secretion, cytosis [155, 174]. Chronic stimulation of B cells was
and morphology. In vitro, and in vivo in diffusion cham- achieved in transgenic mice carrying the human IL-6
bers implanted in mice, IL-1 induced granulocytic dif- gene conjugated with the Ig enhancer [184]. These mice
ferentiation of M1 cells. IL-1 also acted synergistically developed polyclonal nontransplantable plasmocy-
with GM-CSF to induce differentiation of a GM-CSF- tomas. Freshly isolated human myeloma cells also pro-
responsive, IL-1-unresponsive clone of leukemic cells. duce and respond to IL-6, indicating an autocrine role of
The in vivo action of IL-1 was associated with rapid this molecule in the proliferation, but not differentia-
induction of elevated serum levels of MGI-2/IL-6, but tion, of certain malignant B cells [13, 424]. Dependence
serum levels were not sufficiently elevated to account of myelomas on IL-6 may also involve a paracrine
for the differentiation observed within the diffusion mechanism involving marrow stromal cell production
Kapil Mehta et al. 553

of IL-6 in response to myeloma cells [187]. This could LIF treatment [240, 241]; however, antibody to IL-6
involve malignant B-cell production of IL-1, which is a does not block LIF induction of M1 cells. In U937 cells,
potent inducer of IL-6 gene transcription and translation IL-6 did not induce IL-1, nor did IL-1 induce IL-6, and
[260]. In contrast to its action on normal B cells, IL-6 both factors appear to provide distinct signals for differ-
does not augment Ig secretion in myeloma cells [373]. entiation of this neoplastic macrophage cell line [296].
The IL-6-induced proliferation and autocrine IL-6 The action of IL-6 on human acute myeloid leukemic
action have also been reported for certain human B-cell blast cells is observed only when it is combined with
lymphomas, and elevated serum levels of IL-6 were GM-CSF or IL-3. A heterogeneous response is seen
found in 50% of patients with active lymphoma [413]. involving synergistic blast cell proliferation [131, 161,
Interleukin 6 was found to be identical to a mac- 162]. Like IL-1, IL-6 is secreted by most leukemic cells
rophage-granulocyte inducer (MGI-2A) that caused dif- where there is evidence of monocytic differentiation
ferentiation of myeloid leukemic cells but lacked the (12/15 patients with ANLL: of FAB type M4 or M5)
ability to stimulate colony formation [218, 354]. It was [389]. The significance of this potential autocrine look
also identical to a differentiation factor (DIF or D is uncertain.
Factor) produced by Krebs ascites tumor cells, although
it should be noted that this source contains a second
D-factor, which is leukemia inhibitory factor (LIF, see
Leukemia Inhibitory Factor
following section). IL-6 produces complete growth A spectrum of factors have been described that induce
arrest of the murine myeloid leukemia M1 within 48 h, differentiation of the murine M1 myeloblastic leukemic
and this is associated with morphologic maturation of line. This includes G-CSF, IL-1, IL-6, and TNF as well
the blast cells to macrophages with upregulation of as a unique factor termed leukemia inhibitory factor
cfms, FcR, C3R, lysozyme secretion, and development (LIF) on the basis of its potent and selective inhibition
of phagocytic capacity [241, 251, 252]. In clonogenic of M1 cell proliferation in association with induction of
assay of M1 cells diffuse differentiated colonies develop macrophage differentiation [127, 118]. This factor had
within 48 h of addition of IL-6 and the number of colo- earlier been characterized in the conditioned media of
nies is reduced [241]. The murine myelomonocytic leu- L929 cells and termed D-factor, inducing morphologi-
kemic WEHI-3 (D+ variant but not D−) was also induced cal maturation in M1 cells with the development of
by IL-6 to macrophage differentiation with up-regula- locomotor activity, phagocytic capacity, and upregula-
tion of FcR and cfms [50, 240]. Interleukin 6 did not tion of FcR, C3R, and prostaglandin E [381]. The factor
reduce the W3 leukemic clonogenic capacity. was purified from Krebs II ascites conditioned medium
Synergistic or additive interactions between IL-6 and as a 58-kDa glycoprotein [118] and from mouse Ehrlich
other cytokines influence both proliferation and differ- ascites cells as a 40-kDa activity [221]. The native mol-
entiation. The IL-6 and LIF or G-CSF have additive or ecule of 179 amino acids [118] or 180 amino acids [221]
supraadditive effects on M1 differentiation [240, 241, has a molecular weight of 20 kDa with seven N-linked
251, 252]. The IL-1 and IL-6 were additive or synergis- glycosylation sites. The human gene is located on human
tic in growth inhibition of U937, HL-60, and M1 [296, chromosome 22q11–q 12.2 [366]. The murine and
316]. Interleukin 1 alone induced lysozyme production human molecules are active across species and are 78%
by M1 cells but had no direct effect on the expression of homologous at the amino acid level.
FcR or on morphology. Interleukin 6 independently LIF was found to be identical to a variety of other
induced FcR and lysozyme, but the combination trig- cytokines that had been reported to possess diverse
gered the entire sequence of differentiation markers functions. Alloreactive human T cell clones obtained
[316]. The M-CSF also synergized with IL-6 to enhance from rejected kidney, when stimulated with a specific
differentiation of M1 cells but it counteracted the growth antigen and IL-2, produced a factor that triggered the
inhibition, resulting in increased size and number of proliferation of a subline of the IL-3-sensitive murine
macrophage colonies [240, 241]. The possibility existed factor-dependent cell line DA-1 [268, 269]. This factor,
that these interacting cytokines were in fact members of called HILDA (human interleukin DA), was a 38- to
a cascade involving autocrine production of factors by 41-kDa glycoprotein that was also reported to have
leukemic cells. Interleukin 6, IL-1, LIF, and G-CSF are eosinophil chemotactic and activating properties and
all differentiation-inducing factors, they are all endo- erythroid burst-promoting activity [122]. In retrospect
toxin-inducible macrophage products, and they can be these latter activities were probably due to a minor con-
produced by myelomonocytic leukemic cells. The M1 tamination of the protein with GM-CSF and/or other
cells produce IL-6, and levels are increased following lymphokines.
554 Growth and differentiation factors as cancer therapeutics

HILDA cDNA cloned from human lectin-stimulated B myelomonocytic leukemic cells. Abe et al. [3] reported
T cells was shown to be identical to LIF [269]. A human a direct differentiation-inducing, and proliferation-
macrophage differentiation-inducing factor (DIF) for inhibiting, action on HL-60 and U937 cells. Maekawa
M1 cells isolated from a human monocytic leukemia and Metcalf [225] did not observe a direct action of LIF
line, THP-1, was also shown by amino acid sequencing on HL-60 or U937 cells but reported clonal suppres-
to be identical to LIF [3]. The growth of totipotential sion of these leukemic cells by LIF in combination with
embryonic stem cells in vitro is sustained by a differen- GM-CSF or C-CSF. Sequential recloning of these cells
tiation inhibitory activity (DIA) produced by a number was also suppressed by LIF-CSF combinations.
of sources, including the 5,637 human bladder carci- Despite the relatively weak action of LIF on human
noma cell line. Purified DIA and LIF were very similar leukemic cells, LIF clearly is able to exhibit suppressive
in biochemical features, and purified recombinant LIF actions. This, coupled with its lack of suppressive effects
can substitute for DIA in the maintenance of totipotent on normal hematopoietic cells, suggests a role in sup-
embryonic stem cells that retain the potential to form pressing human leukemias, possibly in combination
chimeric mice [406]. A factor constitutively produced with other hematopoietic factors.
by human squamous carcinoma cells stimulated hepato-
cytes to produce the same spectrum of acute phase pro- Tumor Necrosis Factor (TNFa)
teins as IL-6 [23]. This hepatocyte stimulating factor III
was distinct from IL-6 but was identical to LIF. Rat
and Lymphotoxin (TNFb)
heart tissue produces a cholinergic differentiation factor Tumor necrosis factor was identified as a protein with
that controls the phenotypic choice in neurons without antitumor activity in serum of mice infected with Bacillus
affecting their survival or growth [411]. This factor Calmette-Guerin and treated with endotoxin [335].
was also identical to LIF and acted on postmitotic rat TNFα has significant sequence homology to human
sympathetic neurons to specifically induce expression lymphotoxin β and binds to the same receptor. The bio-
of acetylcholine synthesis and cholinergic function and logical actions of TNF are multifaceted, involving both
suppress adrenergic function. stimulatory and inhibitory actions depending upon the
Receptor analysis with 125LIF indicated that M1 cells target cell population.
possess a single class of high-affinity (Kd 100–200 pM) In normal human hematopoiesis TNFα was reported
receptors of relatively low frequency (300–500 per cell) to inhibit CFU-GM day 7 (ED50 = 10 U) and CFU-GM
[156]. In bone marrow, spleen, and peritoneum, mono- on day 14, BFU-E, CFU-E, and CFU-GEMM (ED50 =
cytes, macrophages, and their precursors were the most 50 U) [39, 223, 306]. Even greater inhibition of BFU-E,
obvious cells binding LIF. CFU-GEMM and CFU-E was reported with both TNF
The in vivo action of LIF was revealed by studies in alpha or β in other studies [276]. TNF inhibition was
which mice were engrafted with cells of the murine enhanced in a synergistic manner by interferon γ [39,
hematopoietic cell line FDC-P1 multiply infected with a 276, 306, 307]. TNF inhibition was reported to be nonre-
retroviral construct containing cDNA encoding LIF versible within 60 min, suggesting a cytotoxic mecha-
[242, 243]. The mice developed within 12–70 days a nism [39], but others have reported either full reversibility
fatal syndrome characterized by cachexia, excess osteo- following a 24 h preincubation [276] or partial revers-
blasts with new bone formation, calcification in heart ibility [279]. The variable results can be attributed to the
and skeletal muscle, pancreatitis, thymus atrophy, and use of different sources of CSF and the variable presence
abnormalities in the adrenal cortex and ovarian corpora of accessory cells in the preparation. With G-CSF as a
lutea. The development of this osteosclerotic syndrome, stimulus, the ED50 with TNFα or -β was 10 U whereas
with marked increases in osteoblasts, indicates a role for with a GM-CSF stimulus only a 20% inhibition was seen
LIF in osteoblast production and function and in regulation with 1,000 U TNF [20]. Thus in normal marrow one cell
of bone formation and calcium metabolism. The presence in 500 will form a colony with G-CSF or GM-CSF, but
of LIF receptors on osteoblasts further indicates the direct with TNF only one cell in 100,000 will respond to
nature of the stimulation [242, 243]. G-CSF, yet the frequency of cells responding to GM-CSF
The action of LIF on leukemic cells appears to be remains essentially unchanged [21, 194]. With highly
highly restricted, inducing M1 cell differentiation purified marrow progenitors, the ED50 for TNF was 5 U
alone, or synergistically with G-CSF, IL-6, or M-CSF with G-CSF-stimulated colonies, and 500 U with
[244]. The growth inhibition and differentiation effect GM-CSF stimulation [267]. Using CD34-positive selec-
was rapid, being evident at 24 h and marked at 48 h. tion for CFU-GM, G-CSF-stimulated colonies were
LIF was without effect on normal CFU-GM or WEHI-3 strongly inhibited by TNF, but an actual enhancement of
Kapil Mehta et al. 555

GM-CSF or IL-3-stimulated colonies was recently 90–100% with 100–1,000 U TNF; (b) PE1 and PE2 were
reported [345]. In murine serum-free bone marrow cul- not inhibited and possibly potentiated at low doses of
tures the ED50 for CFU-GM was 20–200 U or murine TNF but suspension cells were markedly inhibited; and
TNF and for BFU-E and CFU-GEMM it was 2,000 U (c) marked inhibition of PE1 and PE2 but not of suspen-
[85]. Using human TNF, a lower degree of inhibition is sion cells. In no case was there evidence of TNF induc-
generally found. In most studies, human TNF does not tion of differentiation. The nature of the stimulus may
inhibit at doses below 10,000 U/ml, and indeed some determine whether the TNF response is inhibitory or
slight potentiation of M-CSF stimulated colonies was stimulatory. Proliferation of AML blast cells (ten
reported [407]. patients), induced by G-CSF, was further stimulated by
In long-term murine or human bone marrow culture, TNF, and clonogenic assays were potentiated 50–250%
TNF exhibits a dose-dependent (10–1,000 U/ml) inhibi- of maximum [163]. In contrast, IL-3- or G-CSF-
tion of CFU-GM production and neutrophil generation stimulated cultures were inhibited 100% and 5,637
[85, 267]. The inhibition induced by TNF appears to be CM-stimulated cultures by 30% with 600 pM of TNF.
selective for neutrophil differentiation and is not manifest Autocrine stimulation of AML cell proliferation seen
in reduction of eosinophils or monocyte/macrophages. at high cell densities was also enhanced by TNF, sug-
The interaction of TNF with leukemic cells is a com- gesting synergism with leukemia cell-derived GM-CSF.
plex one, variously resulting in differentiation, and The mechanism of TNF potentiation is probably indi-
growth inhibition or growth potentiation. A differentiat- rect and was inhibited by antibodies to IL-1. Furthermore,
ing factor for HL-60 and ML-1 leukemic cells, produced IL-1 synergises with GM-CSF in stimulating leukemic
by PHA-stimulated lymphocytes or the HUT 102 T cell blast cells [162], and TNF synergism was not seen in
line was shown to be TNF [370]. As little as 1, 10 U of cultures in the presence of exogenous IL-1. IL-1 and
TNF alone or in synergy with IFNγ promoted mono- TNF have been shown to be constitutive products of
cytic differentiation of HL-60, U937 and ML-1 [135, AML cells and production of bioactive IL-1 can be
194, 306, 346]. increased by TNF treatment [131, 163, 398]. The prolif-
The action of TNF on fresh AML marrow cells has erative response of leukemic blast progenitors to TNF
been investigated in clonogenic (PE1) and recloning (PE2) under conditions that favor autocrine stimulation may
assays, and suspension culture. Early reports uniformly represent one property that allows leukemic cells to
reported inhibition. In one study of ten patients 50% inhi- escape from negative regulation.
bition was seen with 15 pM TNF and there was synergy In the chronic phase of CML, TNF substantially
with IFNγ [306]. Inhibition to a degree comparable to inhibits DNA synthesis in cultures of purified progeni-
normal was reported in seven patients [39], and in nine of tors and inhibits day 7 and day 14 CFU-GM at doses of
ten patients 75% inhibition of leukemic cell cloning was 10–500 U [82, 194]. Some patient-to-patient variation is
seen with 100 U TNF compared to 44–48% inhibition in seen with inhibition in the normal to less-than-normal
remission marrow. Enhanced differentiation to monocyte range. Antibodies to TNFα blocked the TNF inhibition
macrophages with enhanced NBT-reducing activity was and enhanced GM-CSF-stimulated colony formation
seen with TNF or TNF plus IFNγ in suspension cultures twofold in cultures of accessory cell depleted progeni-
of marrow from 1 AML and 5 CML in blastic crisis [119]. tors [82]. Autocrine production of TNF by immature
Differentiated macrophages were observed with Auer CML cells was confirmed by Northern analysis and
rods, indicating their leukemic origin. ELISA assays. The quantities of TNF produced by leu-
In other, more recent studies, a more complex picture kemic cells were sufficient to inhibit normal hematopoi-
emerges. Clonogenic assays stimulated with G-CSF or esis and may provide a selective advantage for leukemic
GM-CSF and exposed to high (1,000 U) or low (100 U) clones if CML progenitors are less sensitive to TNF
doses of TNFβ showed four types of response in AML inhibition than normal. In this context TNF production
and preleukemic myelodysplastic syndrome: inhibition by hairy-cell leukemic cells has been identified as the
greater than normal or remission inhibition equivalent possible cause of myelosuppression and neutropenia
to normal, no response, and significant enhancement. seen in this cancer [211].
This latter pattern, seen in 4/13 cases, involved a syner-
gism with either G-CSF or GM-CSF and clonogeneic
capacity increased up to 60-fold. Assessment of plating
Transforming Growth Factor b
efficiency (PE1, PE2) using 5637 CM as a source of Transforming growth factor β (TGFβ) is a member of a
stimulus revealed three patterns of response with TNF group of polypeptide growth factors that regulate cell
[279]; (a) PE1, PE2, and suspension cell were inhibited growth and differentiation. TGFβ exists as a 25-dK
556 Growth and differentiation factors as cancer therapeutics

disulfide-linked dimer, and subtypes include TGFβ1, 24 h with reversal of inhibition at later times. This obser-
TGFβ2, TGFβ1, 2, existing in homodimer and heterodi- vation suggests that TGFβ, by reversibly inhibiting the
mer forms, and TGFβ3. The factor is produced by most cycling of early stem cells, may be effective in protecting
normal cells and its pleiotropic actions include growth such cells from damage inflicted by cell cycle-specific
stimulation of fibroblasts and growth inhibition of epi- chemotherapy.
thelial cells, endothelial cells, and various malignant The action of TGFβ on myeloid leukemic cells gener-
cells. It also affects differentiation of cells as varied as ally involves potent growth inhibition; however, an
adipocytes, myoblasts, chondrocytes, and epithelium influence on differentiation is seen in some systems.
and is involved in production of extracellular matrix, TGFβ alone induced monocyte-macrophage differentia-
bone remodeling, and repair. TGFβ acts as a modifier of tion of the U937 and THP-1 cell lines but was only a weak
the immune response and may play an important role in inducer of HL-60 [179]. Synergism between TGFβ and
hematopoiesis. It is a potent monocyte chemoattractant TNFα, IFNγ, dexamethasone and phorbol esters is seen
and induces these cells to produce IL-1 and TNF [394]. in differentiation induction of U937, THP-1, and ML-1 in
In vitro, TGFβ1 and -β2 (2–4 pM) inhibit megakaryo- human monocytic or myeloblastic leukemias [133, 179].
cytopoiesis and CFU-MK [40, 173]. In both murine and Low doses (0.5 ng) of TGFβ induced hemoglobinization
human systems, pluripotential progenitors (high prolif- of K562 with growth inhibition [53]. Inhibition of prolif-
erative potential CFU, CFU-GEMM) and BFU-E are eration of M1 murine myeloid leukemic cells is produced
strongly inhibited [7, 137, 182, 299, 340, 358]. by low doses of TGFβ with enhanced adherence, but
Erythropoietin-stimulated CFU-E are not inhibited differentiation-associated properties such as phagocytic
[181, 182, 210, 341]. The action of TGFβ on in vitro activity, lysozyme secretion, and morphologic change
myelopoiesis is determined by the stage of differentia- were not induced [290]. Indeed, TGFβ inhibited the
tion of the progenitor the type of CSF used, and the spe- dexamethasone-induced differentiation of this cell line.
cies. In the murine and human systems, G-CSF and Potent inhibition of proliferation was seen when TGFβ
M-CSF stimulated colonies are not inhibited [137, 181, was added to a variety of factor-dependent cell lines
182, 358]. The GM-CSF-stimulated murine colonies are (NFS-60, 32D, FDCP-1, B6 S4t A, DA-3) and to the
variously reported to be resistant to TGFβ [181, 182] or IL-3-producing WEHI-3 myelomonocytic leukemic cell
inhibited, but to a lesser extent than earlier progenitors, line [181]. Inhibition was also seen with the human leu-
at doses of 0.2–0.5 ng/ml [75, 137, 157, 288, 341]. kemic cell lines U937, THP-1, and KG-1 but not with
Human day 7 CFU-GM (predominantly G-CSF respon- various other lines (e.g., HL-60, various T and B leukemias
sive granulocyte-committed progenitors) were enhanced and lymphomas) [182]. The response of the cell lines to
150–175% by TGFβ1 or -β2 even in the presence of TGFβ appeared to correlate with the extent of display of
plateau concentrations of G-CSF, GM-CSF, or IL-3 [56, TGFβ receptors.
299]. Day 14 CFU-GM were variously reported to be In primary cultures of bone marrow from 15 patients
unaffected by TGFβ [7, 157], or inhibited to a lesser with chronic myeloid leukemia, all showed TGFβ inhi-
degree than earlier progenitors [182, 358]. Synergistic bition of D14 CFU = GM but only 4/15 showed the
or additive interactions occur between TGFβ and other stimulation of D7 CFU-GM, which is the response seen
cytokines. BFU-E were synergistically inhibited by with normal marrow, and in 11/15 TGFβ inhibited D7
TGFβ plus TNFα, and these cytokines additively inhib- CFU-GM [7]. Both TGFβ1 and -β2 inhibited 45% of
ited CFU-GEMM and CFU-GM [359]. Interferon CFU-GM stimulated by GM-CSF in marrow cultures
gamma and TGFβ synergistically inhibited CFU-GM from five CML patients but, in contrast to normal,
and additively inhibited BFU-E and CFU-GEMM. G-CSF-stimulated colonies were also inhibited [358]. In
In long-term bone marrow culture, TGFβ serves as a studies of acute myeloid leukemia (18 patients), TGFβ
potent inhibitor of myelopoiesis, probably by inhibiting suppressed both primary and secondary leukemic clo-
proliferation of early stem cells in the adherent stromal nogenic capacity [282, 375]. These results showed that
layer [44, 150]. Indeed, TGFβ production by marrow 1–10 ng/ml of TGFβ delayed progression of leukemic
stromal cells may be an important negative regulator of blast-cell progenitors from G1 to S phase in a cytostatic
steady-state hematopoiesis. Further evidence for a phys- manner with no induction of differentiation.
iological role for TGFβ as negative regulator was provided It is now apparent that a large number of factors
in studies in which TGFβ1 (1–5 μg/mouse) was injected responsible for growth and differentiation have been
via the femoral artery into normal mice [123, 124]. The identified. In addition, their receptors on normal and
CFU-GEMM stimulated by IL-3 were completely inhib- neoplastic cells are also being identified and investi-
ited in the femur, and CFU-GM were inhibited 50% by gated. It is clear that some of these factors are produced
Kapil Mehta et al. 557

by tumor cells and represent autocrine growth factors. adjacent tissues. This approach, when employed in con-
Obviously, the blockage of these growth factors by cert with the use of differentiation agents, may allow for
reducing their production or by blocking their attach- the regulation and control of tumor growth in a manner
ment to receptors could be an effective strategy for cancer very different than chemotherapy and radiation therapy.
treatment. Likewise, the production of stimulatory factors Perhaps one can regulate cancer in a manner similar to
for angiogenesis by tumor cells has now been well char- the use of insulin to regulate diabetes rather than
acterized and the blockage of their paracrine effect on attempting to destroy every last cancer cell with all the
adjacent blood vessels in normal tissue is another poten- toxicity inherent in these historical approaches.
tial method of therapy. Some tumors produce a consider-
able quantity of growth factor proteins and peptides and
others produced smaller amounts or a wider diversity of References
these factors. In some, factors which promote the growth 1. Abe E, Miyaura C, Sakagami H, et al. Differentiation of mouse
of tumor cells clearly play a role in cancer growth and myeloid leukemia cells induced by 1alpha, 25-dihydroxy-vitamin D3.
metastasis and the autonomy that cancer seems to enjoy. Proc Natl Acad Sci USA 1981; 78:4990.
An interruption of this process will clearly lead to further 2. Abe E, Miyaura C, Tanaka H, et al. 1α25-Dihydroxyvitamin D3
promotes fusion of mouse alveolar macrophages both by a direct
approaches in cancer treatment.
mechanism and by a spleen cell-mediated indirect mechanism.
A good example of the clinical significance of growth Proc Natl Acad Sci USA 1983; 80:5583.
factors has been demonstrated in studies showing that 3. Abe T, Murakami M, Sato T, et al. Macrophage differentiation
approximately 50% of tumor tissue from breast cancer inducing factor from human monocytic cells is equivalent to
patients expresses epidermal growth factor receptor. murine leukemia inhibitory factor. J Biol Chem 1989; 264:8941.
4. Abita J, Gauville C, Saal F. Characterization of insulin receptors in
These receptors appear to be inversely related to steroid
human promyelocytic leukemia cell HL-60. Biochem Biophys Res
receptors and therefore bring a poor prognosis to patients Commun 1982; 106:574.
who are steroid-receptor negative and epidermal growth 5. Abita JP, Gauville C, Baltrand N. Loss of insulin receptor during
factor receptor positive. Since thousands of tumors have differentiation of U937 cells. IRCS Med Sci 1983; 11:390.
been studied with regard to this particular characteristic, 6. Aggarwal PP, Kohr WJ, Hass PE, et al. Human tumor necrosis
factor: production, purification and characterization. J Biol Chem
it is abundantly clear that epidermal growth factor recep- 1985; 260:2345.
tor should be explored as a target for cancer treatment. 7. Aglietta M, Stacchini A, Severino A, et al. Interaction of trans-
Monoclonal antibodies have been produced by various forming growth factor-beta 1 with hemopoietic growth factors in
investigators which are being administered to patients the regulation of human normal and leukemic myelopoiesis. Exp
with breast cancer in an attempt to block the epidermal Hematol 1989; 17:296.
8. Altucci L, Rossin A, Raffelsberger W, et al. Retinoic acid induced
growth factor receptor. Many of these studies are being apoptosis in leukemia cells is mediated by paracrine action of
done in association with chemotherapy to try to get an tumor-selective death ligand TRAIL Nat Med 2001; 7:680.
effective one–two punch in inhibiting tumor growth. 9. Amento EP, Kurnick JT, Epstein A, et al. Modulation of synovial
Oncogenes such as erb and v-cis are known to encode cell products by a factor from a human cell line: T lymphocyte
induction of a mononuclear cell factor. Proc Natl Acad Sci USA
for growth factor receptors. Soluble receptors with high
1982; 79:5307.
affinity binding might be useful in therapy to bind with 10. Andreeff M, Kreis W, Miller W, et al. Low-dose cytosine-arabinoside
growth factors and reduce their activities on membrane- (ARA-C) in acute non-lymphocytic leukemia (ANLL) and myelo-
bound receptors present in the tumor cell or adjacent dysplastic syndromes (MDS): analysis of response and response
blood vessel. Factors which are structurally similar to mechanisms. Proc Annu Meet Am Soc Clin Oncol 1988; 7:A741.
11. Andreeff M, Stone R, Young C, et al. Treatment of myelodysplastic
growth factors but are inactive in inducing receptor
syndromes and acute myeloid leukemia with hexamethylene
function (blockers) are also being explored as treatment bisacetamide. Blood 1990; 76(Suppl.):251a.
strategies. Growth factor interaction can also be blocked 12. Arlin ZA, Mertelsman R, Berman E, et al. 13-cis-Retinoic acid
by certain drugs such as Suramin. Suramin and its ana- does not increase the true remission (induced by cytotoxic chemo-
logues are currently in clinical trials for these effects. therapy) in patients with chronic phase chronic myelogenous
leukemia. J Clin Oncol 1985; 3:473.
Anti-angiogenesis factors have also been identified and 13. Asaoku H, Kawano M, Kwato K, et al. Decrease in BSF-2/IL-6
these are also in clinical trials. In summary, the applica- response in advanced cases of multiple myeloma. Blood 1988;
tion of techniques to block growth factors and/or the 72:429.
transduction of biochemical effects from growth factor 14. Ascencao J, Kay NE, Earenfight-Engler T, et al. Production of eryth-
receptor stimulation represent advent technologies for roid potentiating factor(s) by a human monocytic cell line. Blood
1981; 57:170.
the millennium. As more is known about autocrine and 15. Aul C, Runde, Gattermann N. All-trans retinoic acid in patients
paracrine effects of growth factors, more can be done with myelodysplastic syndromes: results of a pilot study. Blood
with respect to altering their effects on tumor cells and 1993; 82:2967.
558 Growth and differentiation factors as cancer therapeutics

16. Baccarani M, Zaccaria A, Bandini G, et al. Low dose arbinosyl 37. Breitman T, Collins S, Keene B. Terminal differentiation of human
cytosine for treatment of myelodysplastic syndromes and subacute promyelocytic leukemia cells in primary culture in response to
myeloid leukemia. Leuk Res 1983; 7:539. retinoic acid. Blood 1981; 57:1000.
17. Baccarin M, Tura S. Differentiation of myeloid leukaemic cells: 38. Broxmeyer H, Gentile G, Bognacki J, et al. Lactoferrin, transferrin
new possibilities for therapy. Br J Haematol 1979; 42:485. and acidic isoferritins: regulatory molecules with potential thera-
18. Bagby GC, Dinarello CA, Neerhout RC, et al. Interleukin 1-dependent peutic value in leukemia. Blood Cells 1983; 9:83.
paracrine granulopoiesis in chronic granulocytic leukemia of the juve- 39. Broxmeyer HE, Williams DE, Lu L, et al. The suppressive influence
nile type. J Clin Invest 1988; 82:1430. of human tumor necrosis factors on bone marrow hematopoietic
19. Balaban EP, Cox JV, Schneider NR, et al. Treatment of “poor risk” progenitor cells from normal donors and patients with leukemia:
acute non-lymphocytic leukemia with continuously infused low-dose synergism of tumor necrosis factor and interferon-γ. J Immunol
cytosine arabinoside. Am J Hematol 1988; 29:79. 1986; 136:4487.
20. Barbar KE, Crosier PS, Gillis S, et al. Human granulocyte-mac- 40. Bruno E, Hoffman R. Interacting cytokines regulate human mega-
rophage progenitors and their sensitivity to cytotoxins: analysis by karyocytopoiesis. Exp Hematol 1988; 16:505.
limiting dilution. Blood 1987; 70:1773. 41. Buick RN, Till JE, McCulloch EA. Colony assay for proliferative
21. Barbar KE, Crosier PS, Watson JD. The differential inhibition of blast cells circulating in myeloblastic leukemia. Lancet 1977;
hemopoietic growth factor activity by cytotoxins and interferon-γ. 1:862.
J Immunol 1987; 139:1108. 42. Buick RN, Chang LJ-A, Curtis JE, et al. Self-renewal capacity of
22. Bar-Shavit Z, Teitelbaum SL, Reitsma P, et al. Induction of mono- leukemic blast progenitor cells. Cancer Res 1981; 4:4849.
cyte differentiation and bone resorption by 1,25-dihydroxy-vitamin 43. Burger EH, Van Der Meer JWM, Van De Geuell JS, et al. In vitro
D3. Proc Natl Acad Sci USA 1983; 80:5907. formation of osteoclasts from long-term cultures of bone marrow
23. Baumann H, Wong GG. Hepatocyte-stimulating factor III shares mononuclear phagocytes. J Exp Med 1982; 156:1604.
structural and functional identity with leukemia-inhibitory factor. 44. Cashman JD, Eaves AC, Raines EW, et al. Mechanisms that regu-
J Immunol 1989; 143:1163. late the cell cycle status of very primitive hematopoietic cells in
24. Beckman BS, Balin AK, Allen RG. Superoxide dismutase induces long-term marrow cultures. I. Stimulatory role of a variety of mes-
differentiation of Friend erythroleukemia cells. J Cell Physiol enchymal cell activators and inhibitory role of TGF-β. 1990;
1989; 139:370. 75:96.
25. Begley C, Metcalf D, Nicola N. Purified colony stimulating factors 45. Casini T, Pelicci PG. A function of p21 during promyelocytic leu-
(G-CSF and GM-CSF) induce differentiation in human HL-60 leu- kemia cell differentiation independent of CDK inhibition and cell
kemic cells with suppression of clonogenicity. Int J Cancer 1987; cycle arrest. Oncogene 1999;18:3235.
39:99. 46. Castaigne S, Chomienne C, Daniel MT et al. All-trans retinoic
26. Ben-Bassat I, Gale RP. New and evolving concepts in leukemia. acid as differentiation therapy for acute promyelocytic leukemia.
Leukemia 1988; 2:704. I. Clinical results. Blood 1990; 76:1704.
27. Bernal S, Chen L. Induction of cytoskeleton-associated proteins 47. Castaigne S, Daniel MT, Tilly H, et al. Does treatment with Ara-C
during differentiation of human myeloid leukemic cell lines. in low dosage cause differentiation of leukemic cells? Blood 1983;
Cancer Res 1982; 421:5106. 62:85.
28. Besa EC, Abrham JL, Bartolomew MJ, Hizinski M, Nowell, P.C. 48. Castro-Malaspina H, Rabellino EM, Yen A, et al. Human mega-
Treatment with 13-cis-retinoic acid in transfusion dependent karyocyte stimulation of proliferation of bone marrow fibroblasts.
patients with myelodysplastic syndrome and decreased toxicity Blood 1981; 57:781.
with addition of alpha-tocopherol. Am J Med 1988; 89:739. 49. Chan CSP, Schechter GP. In vitro evidence for dose-dependent
29. Besa EC, Abraham JL, Nowell PC. Combinations of 13-cis-retin- cytotoxicity as the predominant effect of low dose Ara-C on human
oic acid (RA) and alpha-tocopherol (AT) for the chronic phase and leukemic and normal marrow cells. Cancer Chemother Pharmacol
addition of alpha-interferon (IFN) for the transformed phase of 1988; 23:87.
myelodysplasia. Third Int Conf Prev Hum Cancer 1988:S4–13. 50. Chen CP, Lee F. IL-6 is a differentiation factor for M1 and
30. Blazsek I, Labat ML, Boule D, et al. Endocrine switch in myeloid WEHI-3B myeloid leukemic cells. J Immunol 1989; 142:1909.
neoplasia? Proc Annu Meet Am Assoc Cancer Res 1989; 30:A928. 51. Chen JD, Evans RM. A transcriptional co-repressor that interacts
31. Bleiberg I, Fabian I, Kantor S, et al. The effect of 13-cis-retinoic with nuclear hormone receptors. Nature 1995; 377, 454.
acid on hematopoiesis in human long-term bone marrow culture. 52. Chen JD, Umesono K, Evans RM. SMRT isoforms mediate repres-
Leuk Res 1988; 12:545. sion and anti-repression of nuclear receptors heterodimers. Proc
32. Bloch A, Koeffler HP, Pierce GB, et al. Meeting Report: Ninth Natl Acad Sci, USA 1996; 93:7567.
Annual Sapporo Cancer Seminar on Cell Differentiation and 53. Chen LL, Dean A, Jenkinson T, et al. Effect of transforming
Cancer Control. Cancer Res 1990; 50:1346. growth factor-β1 on proliferation and induction of hemoglobin
33. Bodner AJ, Ting RC, Gallo RC. Induction of differentiation of accumulation in K-562 cells. Blood 1989; 74:2368.
human promyelocytic leukemia cells (HL-60) by nucleosides and 54. Chen ZX, Xue YQ, Zhang RI, et al. A clinical and experimental
methotrexate. J Natl Cancer Inst 1981; 67:1025. study on all-trans retinoic acid-treated acute promyelocytic leukemia
34. Bougnoux P, Bovini E, Chang Z, et al. Effect of interferon on patients. Blood 1991; 78:1413–1419.
phospholipid methylation by peripheral blood mononuclear cells. 55. Cheng GYM, Kelleher CA, Miyauchi J, et al. Structure and expres-
J Cell Biochem 1983; 20:215. sion of genes of GM-CSF and G-CSF in blast cells from patients
35. Bourantas KL, Tsiara S, Christou L Treatment of 34 patients with with acute myeloblastic leukemia. Blood 1988; 71:204.
myelodysplastic syndromes with 13-cis retinoic acid. Eur J 56. Chenu C, Pfeilschifter J, Mundy GR, et al. Transforming growth
Haematol 1995; 55:235. factor beta inhibits formation of osteoclast-like cells in long-term
36. Bradbury D, Bowen G, Kozlowski R, et al. Endogenous human marrow cultures. Proc Natl Acad Sci USA 1988; 85:5683.
interleukin-1 can regulate the autonomous growth of the blast cells 57. Christman JK, Mendelsohn N, Herzog D, et al. Effect of 5-azacy-
of acute myeloblastic leukemia by inducing autocrine secretion of tidine on differentiation and DNA methylation in human promy-
GM-CSF. Leukemia 1990; 4:44. elocytic leukemia cells (HL-60). Cancer Res 1983; 43:763.
Kapil Mehta et al. 559

58. Clark RE, Ismail SA, Jacobs A et al. A randomized trial of 13-cis- 76. Douer D, Estey E, Santillana S, et al. Treatment of newly diag-
retinoic acid with or without cytosine arabinoside in patients with nosed and relapsed acute promyelocytic leukemia with intrave-
myelodysplastic syndromes. Br J Haematol 1987; 66:77. nous liposomal all-trans retinoic acid. Blood 2001; 97:73.
59. Clark RE, Smith SA, Jacobs A Myeloid surface antigen abnormali- 77. Douer D, Koeffler HP. Retinoic acid inhibition of the clonal growth
ties in myelodysplasia: relation to prognosis and modification by of human myeloid leukemia cells. J Clin Invest 1982; 69:277.
13-cis retinoic acid. J Clin Path 1987; 40:652. 78. Douer D, Koeffler HP. Retinoic acid enhances growth of human
60. Cobb MA, Hseuh W, Pachman L, et al. Prostaglandin biosynthesis early erythroid progenitor cells in vitro. J Clin Invest 1982;
by a human macrophage-like cell line, U937. J Reticuloendothel 60:1039.
Soc 1983; 33:197. 79. Doyle M, Brindley L, Kawasaki E, et al. High level human inter-
61. Colbert D, Fontana J, Bode U, et al. Changes in the translational leukin 1 production by a hepatoma cell line. Biochem Biophys Res
activity of polyadenylated messenger RNA of HL-60 promyelo- Commun 1985; 130:768.
cytic leukemia cells associated with myeloid or macrophage dif- 80. Duhrsen U. In vitro growth patterns and autocrine production of
ferentiation. Cancer Res 1983; 43:229. hemopoietic colony stimulating factors: analysis of leukemic pop-
62. Collins SJ, Ruscetti FW, Gallagher RE, et al. normal functional ulations arising in irradiated mice from cells of an injected factor-
characteristics of cultured human promyelocytic leukemia cells dependent continuous cell line. Leukemia 1988; 2:334.
(HL-60) after induction of differentiation by DMSO. J Exp Med 81. Duhrsen U, Metcalf D. A model system for leukemic transforma-
1979; 149:969. tion of immortalized hemopoietic cells in irradiated recipient mice.
63. Collins S, Bodner A, et al. Induction of morphological and func- Leukemia 1988; 2:329.
tional differentiation of human promyelocytic leukemia cells (HL- 82. Duncombe AS, Heslop HE, Turner M, et al. Tumor necrosis
60) by compounds which induce differentiation of murine leukemia factor mediates autocrine growth inhibition in a chronic leukemia.
cells. Int J Cancer 1980; 25:213. J Immunol 1989; 143:3828.
64. Conley BA, Forrest A, Egorin MJ, et al. Phase I trial using adap- 83. Egorin MJ, Sigman LM, Van-Echo DA, et al. Phase I clinical and
tive control dosing of hexamethylene bisacetamide (NSC 95580). pharmacokinetic study of hexamethylene bisacetamide (NSC
Cancer Res 1989; 49:3436. 95580) administered as a five-day continuous infusion. Cancer Res
65. Cooper PC, Metcalf D, Burgess AW. Biochemical and functional 1987; 47:617.
characterization of mature progeny purified from a myelomono- 84. Eisman JA, Martin TJ, MacIntyre I, et al. 1,25-Dihydroxyvitamin D3
cytic leukemia. Leuk Res 1982; 6:313. receptor in a cultured human breast cancer cell line (MCF 7 cells).
66. Cooperberg AA, Singer OP. Reversible myelofibrosis due to vita- Biochem Biophys Res Commun 1980; 93:9.
min D deficiency rickets Can Med Assoc J 1980; 94:392. 85. Eliason JF, Vassalli P. Inhibition of hemopoiesis in murine marrow
67. Cozzolino F, Rubartelli A, Aldinucci D, et al. Interleukin 1 as an cell cultures by recombinant murine tumor necrosis factor γ: evi-
autocrine growth factor for acute myeloid leukemia cells. Proc dence for long-term effects on stromal cells. Blood Cells 1988;
Natl Acad Sci USA 1989; 86:2369. 14:339.
68. Curtis JE, Messner HA, Minden MD, et al. Improved maintenance 86. Estey EH, Giles FJ, Kantarjian H, O’Brien S et al.. Molecular
therapy for acute myelogenous leukemia (AML) using 13-cis- remissions induced by liposomal-encapsulated all-trans retinoic
retinoic acid-containing regimen. Proc Annu Meet Am Assoc acid in newly diagnosed acute promyelocytic leukemia. Blood
Cancer Res 1989; 30:A1066. 1999; 94:2230.
69. Dayton ET, Perussia G, Trinchiere G. Correlation between differ- 87. Estey E, Thall PF, Kantarijian H, et al. Treatment of newly diag-
entiation, expression of monocyte-specific antigens, and cytotoxic nosed acute myelogenous leukemia with granulocyte-macrophage
functions in human promyelocytic cell lines treated with leukocyte colony-stimulating factor (GM-CSF) before and during continu-
conditioned media. J Immunol 1983; 3:1120. ous infusion high-dose ara-C + daunorubicin: comparison to
70. De Botton S, Dombret H, Sanz M, Miguel JS, Caillot D, Zittoun R, patients treated without GM-CSF. Blood 1992; 79:2246.
Gardembas M, Stamtoulas, Conde E, Fenauz P. Incidence, clinical 88. Estey E, Thall PF, Mehta K et al. Alterations in tretinoin pharma-
features, and outcome of all trans retinoic acid syndrome in 413 cokinetics following administration of liposomal all-trans retinoic
cases of newly diagnosed acute promyelocytic leukemia. Blood acid. Blood 1996; 87:3650.
1998; 92:2712. 89. Fearon ER, Burke PJ, Schiffer CA, et al. Differentiation of leuke-
71. Degos L, Castaigne S, Chomienne C, et al. Therapeutic trials of mic cells to polymorphonuclear leukocytes in patients with acute
acute myeloid leukemia and of myelodysplastic syndromes by nonlymphocytic leukemia. N Engl J Med 315:15.
LD-ARA C and treatment of promyelocytic leukemia by retinoic 90. Fenaux P, Chomienne C Biology and treatment of acute promyelo-
acid. In: Waxman S, Ross GB, Takaku F, eds. The status of differ- cytic leukemia. Curr Opin Oncol 1996; 8:3.
entiation therapy of cancer. Serono Symposia, Publications from 91.Fenaux P, Le Dely MC, Castaigne S, et al. Effect of all-trans retinoic
Raven Press, 1988; 45:361. acid in newly diagnosed acute promyelocytic leukemia: results of
72. Degos L, Dombret H, Chomienne C. et al. All-trans-retinoic acid a multicenter randomized trial. Blood 1993; 82:3241–3249.
as a differentiating agent in the treatment of acute promyelocytic 92. Fenaux P, Chomienne C, Degos L. All trans-retinoic acid and
leukemia. Blood 1995; 85:2643 chemotherapy in the treatment of acute promyelocytic leukemia.
73. Delwel R, Salem M, Pellens C, et al. Growth regulation of human Sem Hematol. 2001; 38:13.
acute myeloid leukemia: effects of five recombinant hematopoietic 93. Ferrero D, Bruno B, Pregno P et al. Combined differentiating ther-
factors in a serum-free culture system. Blood 1988; 72:1944. apy for myelodysplastic syndromes: a phase II study. Leuk Res
74. DiGiovanna MP, Sartorelli AC. Myeloid differentiation-associated 1996; 20:867.
protein tyrosine kinase activity in a murine monomyelocytic leukemia 94. Ferrero D, Tarella C, Gallo E, et al. Terminal differentiation of the
cell line. Proc Annu Meet Am Assoc Cancer Res 1989; 30:A212. human promyelocyte leukemia cell line, HL-60, in the absence of
75. Dooley DC, Law P. Influence of transforming growth factor-beta cell proliferation. Cancer Res 1982; 42:4421.
one and T cells on human hematopoietic progenitor cells. Exp 95. Fialkow PJ, Singer JW, Adamson JW, et al. Acute nonlymphocytic
Hematol 1988; 16:438. leukemia: heterogeneity of stem cell origin. Blood 1981; 57:1068.
560 Growth and differentiation factors as cancer therapeutics

96. Fibach E, Peled T, Rachmilewitz EA. Self-renewal and commit- colony stimulating factor in patients with myelodysplastic syn-
ment to differentiation of human leukemic promyelocytic cells dromes. Leukemia 1994; 8:369.
(HL-60). J Cell Physiol 1982; 113:152. 115. Garewal HS, List A, Meyskens F, et al. Phase II trial of fenretinide
97. Filmus J, Buick RN. Relationship of c-myc expression to differ- [N-(4-hydroxyphenyl) retinamide] in myelodysplasia: possible
entiation and proliferation of HL-60 cells. Cancer Res 1985; retinoid-induced disease acceleration. Leuk Res 1989; 13:339.
45:822. 116. Gasson J, Weisbart R, Kaufman S, et al. Purified human granulo-
98. Fitchen J, LeFevre C, Ferrone S, et al. Expression of Ia-like and cyte-macrophage colony-stimulating factor: direct action on neu-
HLA-A,B, antigens on human multipotential hematopoietic pro- trophils. Science 1984; 226:1339.
genitor cells. Blood 1982; 59:188. 117. Gately CL, Wahl SM, Oppenheim J. Characterization of a hydrogen
99. Fontana JA, Wright DG, Schifman E, et al. Development of peroxide-potentiating factor, a lymphokine that increases the
chemotactic responsiveness in myeloid precursor cells: studies capacity of human monocytes and monocyte-like cell lines to
with a human leukemia cell line. Proc Natl Acad Sci USA 1980; produce hydrogen peroxide. J Immunol 1983; 131:2853.
77:3664. 118. Gearing DP, Gough NM, King JA. Molecular cloning and expres-
100. Franpton RJ, Suva LJ, Eisman JA, et al. Presence of 1,25-dihy- sion of cDNA encoding a murine myeloid leukaemia inhibitory
droxyvitamin D3 receptors in established human cancer cell lines factor (LIF). EMBO J 1987; 6:3995.
in culture. Cancer Res 1982; 42:1116. 119. Geissler K, Tricot G, Leemhus T, et al. Differentiation-inducing
101. Franceschi RT. Interaction of the 1-alpha, 25-dihydroxyvitamin D3 effect of recombinant human tumor necrosis factor α and
receptor with RNA and synthetic polyribonucleotides. Proc Natl γ-interferon in vitro on blast cells from patients with acute myel-
Acad Sci USA 1984; 81:2337. oid leukemia and myeloid blast crisis of chronic myeloid leuke-
102. Francis GE, Berney JJ, North PS, et al. Evidence for the involvement mia. Cancer Res 1989; 49:3057.
of DNA topoisomerase II in neutrophil-granulocyte differentia- 120. Gianni M, Kalac Y, Ponzanelli I, et al. Tyrosine kinase inhibitor
tion. Leukemia 1987; 1:653. ST1571 potentiates the pharmacological activity of retinoic acid
103. Francis GE, Mufti GJ, Knowles SM, et al. Differentiation induc- in acute promyelocytic leukemia cells: effect on the downregula-
tion in myelodysplasia and acute myeloid leukemia: use of syner- tion of RARα and PML-RARα. Blood 2001; 97:3234.
gistic drug combinations. Leuk Res 1987; 11:971. 121. Gitter BD, Finn OJ, Metzgar RS. Cytofluorometric isolation of
104. Francis GE, Gray DA, Berney JJ, et al. Role of ADP-ribosyl trans- I937, an Ia antigen-bearing variant of the Ia-negative human
ferase in the differentiation of human granulocyte-macrophage monocytic cell line U937. J Immunol 1985; 134:280.
progenitors to the macrophage lineage. Blood 1983; 62:1055. 122. Godard A, Gascan H, Naulet J, et al. Biochemical characteriza-
105. Freake HC, Marcocci C, Iwasaki J, et al. 1,25-Dihydroxyvitamin D3 tion and purification of HILDA, a human lymphokine active on
specifically binds to a human breast cancer cell line (T47D) and eosinophils and bone marrow cells. Blood 1988; 71:1618.
stimulates growth. Biochem Biophy Res Commun 1981; 101:1131. 123. Goey H, Keller JR, Jansen R, et al. Antiproliferative effects of
106. Frankel SR, Eardley A, Lauwers G et al. The ‘retinoic acid syn- transforming growth factor beta 1 for murine tumors and
drome’ in acute promyelocytic leukemia. Ann Intern Med 1992; hematopoietic progenitor cells. Proc Annu Meet Am Assoc Cancer
117:292. Res 1989; 30:A1295.
107. Gabrilove JL, Welte K, Li L, et al. Constitutive production of a 124. Goey H, Keller JR, Back T, et al. Inhibition of early murine
leukemia differentiation, colony stimulating, erythroid burst pro- hemopoietic progenitor cell proliferation after in vivo locoregional
moting and pluripoietic factor by a human hepatoma cell line: administration of transforming growth factor-β1. J Immunol
characterization of the leukemia differentiation factor. Blood 1989; 143:877.
1985; 66:407. 125. Gold EJ, Mertelsmann R, Itri LM, et al. Phase I clinical trial of
108. Gabrilove JL, Welte K, Harris P, et al. Pluripoietin alpha: a sec- 13-cis-retinoic acid in myelodysplastic syndrome. Cancer Treat
ond human hematopoietic colony-stimulating factor produced by Rep 1983; 11:981.
the human bladder carcinoma cell line 5637. Proc Natl Acad Sci 126. Goodman RB, Strieter RM, Martin DP et al. Inflammatory cytok-
USA 1986; 83:2478. ines in patients with persistence of the acute respiratory distress
109. Gahmberg CG, Nilsson K, Anderson LC. Specific changes in the syndrome. Am J Respir Crit Care Med 1996;1 54:602.
surface of glycoprotein pattern of human promyelocytic leuke- 127. Gough NM, Gearing DP, King JA, et al. Molecular cloning and
mia cell line HL-60 during morphologic and functional differen- expression of the human homologue of the murine gene encoding
tiation. Proc Natl Acad Sci USA 1979; 76:4087. myeloid leukemia-inhibitory factor. Proc Natl Acad Sci USA
110. Gallagher R, Collins S, Trujillo J, et al. Characterization of the 1988; 85:2623.
continuous, differentiating myeloid cell line (HL-60) from a 128. Greenberg BR, Durie BG, Barnett TC, Meyskens F. Phase I/II
patient with acute promyelocytic leukemia. Blood 1979; 54:713. study of13-cis-retinoicacid in myelodysplastic syndrome. Cancer
111. Gallagher JC, Jerpbak CM, Jee WSS, et al. 1,25-Dihydroxyvitamin Treat Rep 1985; 69:1369.
D3: short- and long-term effects on bone and calcium metabolism 129. Griffin JD. Role of colony stimulating factors in the biology of
in patients with postmenopausal osteoporosis. Proc Natl Acad Sci leukemia. Exp Hematol 1988; 16:417.
USA 1982; 79:3325. 130. Griffin J, Munroe D, Major P, et al. Induction of differentiation of
112. Gallagher RE, Bilello PA, Ferrari AC, et al. Characterization of human myeloid leukemia cells by inhibitors of DNA synthesis.
differentiation-inducer-resistant HL-60 cells. Leuk Res 1985; Exp Hematol 1982; 9:774.
9:967. 131. Griffin JD, Rambaldi A, Vellenga E, et al. Secretion of interleu-
113. Gallo RC, Breitman TR, Ruscelli FW. Proliferation and differen- kin-1 by acute myeloblastic leukemia cells in vitro induces
tiation of human myeloid leukemia cell lines in vitro. In: Moore endothelial cells to secrete colony stimulating factors. Blood
MAS, ed. Maturation factors in cancer. New York: Raven Press, 1987; 70:1218.
1987:255. 132. Grosso LE, Pitot HC. Transcriptional regulation of c-myc during
114. Ganser A, Seipelts G, Verbeek W et al. Effects of combination ther- chemically induced differentiation of HL-60 cultures. Cancer
apy with all-trans-retinoic acid and recombinant human granulocyte Res 1985; 45:847.
Kapil Mehta et al. 561

133. Guan XP, Fujii Y, Hromchak RA, et al. Synergistic induction of D∞ in combination with low dose ara-C is not superior to ara-C alone.
ML-1 human myeloblastic leukemia cell differentiation by com- Results from a randomized study. The Scandinavian Myelodysplasia
binations of 12-O-tetradecanoyl-phorbol 13-acetate (TPA) with Group (SMG). Eur J Haematol 1990; 45:255.
transforming growth factor (TGF-beta) or with human tumor 153. Hemmi H, Breitman TR. Combinations of recombinant human
necrosis factor (TNF-alpha). Proc Annu Meet Am Assoc Cancer interferons and retinoic acid synergistically induce differentiation
Res 1989; 30:A175. of the human promyelocytic leukemia cell line HL-60. Blood
134. Guernsey DL, Yen A. Retinoic-acid-induced modulation of 1987; 69:501.
c-myc not dependent on its continued presence: possible role in 154. Hemmi H, Nakamura T, Shimizu Y, et al. Identification of com-
precommitment for HL-60 cells. Int J Cancer 1988; 42:576. ponents of differentiation-inducing activity of human T-cell lym-
135. Guyre PM, Morganelli PM, Miller R. Recombinant immune phoma cells by induction of differentiation in human myeloid
interferon increases IgG FcR receptors on cultured human mono- leukemia cells. J Natl Cancer Inst 1989; 81:952.
nuclear phagocytes. J Clin Invest 1983; 72:393. 155. Hill R, Warren K, Stenberg P, et al. Purified human recombinant
136. Hagenbeek A, Sizoo W, Lowenberg B. Treatment of acute myelo- IL-6 stimulates murine megakaryocytopoiesis and increases
cytic leukemia with low dose cytosine arabinoside: results of a platelet levels. Blood 1989; 74(suppl 1):207 (abstr).
pilot study in four patients. Leuk Res 1983; 7:443. 156. Hilton DJ, Nicola NA, Metcalf D. Specific binding of murine
137. Hampson J, Ponting ILO, Cook N, et al. The effects of TGF beta leukemia inhibitory factor to normal and leukemic monocytic
on haemopoietic cells. Growth Factors 1988; 1:193. cells. Proc Natl Acad Sci USA 1988; 85:5971.
138. Hansen CM, Binderup L, Hamberg KJ, Carlberg C. Vitamin D 157. Hino M, Tojo A, Miyazono K, et al. Effects of type β transform-
and Cancer: effects of 1,25-(OH)2 D3 and its analogs on growth ing growth factors on haematopoietic progenitor cells. Br J
control and tumorigenesis. Front Biosci 2001; 6:820. Haematol 1988; 70:143.
139. Hansen F. Hemopoietic growth and inhibitory factors in treat- 158. Hirano T, Yasukawa K, Harada H, et al. Complementary DNA for
ment of malignancies. Acta Oncologica 1995; 34(4):453–468. a novel human interleukin (BSF-2) that induces B lymphocytes
140. Hapel AJ, Vande Woude G, Campbell HD, et al. Generation of an to produce immunoglobulin. Nature 1986; 324:73.
autocrine leukemia using a retroviral expression vector carrying 159. Hittelman WN, Agbor P, Petkovic I, et al. Detection of leukemic
the interleukin-3 gene. Lymphokine Res 1986; 5:249. clone maturation in vivo by premature chromosome condensa-
141. Harris PE, Ralph P, Litcofsky P, et al. Distinct activities of inter- tion. Blood 1988; 72:1950.
feron-gamma, lymphokine and cytokine differentiation inducing 160. Ho AD, Martin H, Knauf W, et al. Combination of low dose
factors acting on the human monoblastic leukemia cell line U937. cytarabine and 13-cis retinoic acid in the treatment of myelodys-
Cancer Res 1985; 45:9. plastic syndromes. Leuk Res 1987; 11:1041.
142. Harris PE, Ralph P, Gabrilove J, et al. Broad spectrum induction 161. Hoang T, Haman A, Goncalves O, et al. Interleukin-6 enhances
by cytokine factors and limited induction by gamma interferon of growth factor-dependent proliferation of the blast cells of acute
differentiation in the human promyelocytic leukemia cell line myeloblastic leukemia. Blood 1988; 72:823.
HL-60. Cancer Res 1985; 45:3090. 162. Hoang T, Haman A, Goncalves O, et al. Interleukin 1 enhances
143. Hartman D, Sheridan AP, Moore MAS. Effects of 1,25(OH)2 D3, growth factor dependent proliferation of the clonogenic cells in
dexamethasone and cytoxan on the survival of leukemic (WEHI- acute myeloblastic leukemia and of normal human primitive
3B(D+) Balb/c mice. Unpublished observation, 1987. hemopoietic precursors. J Exp Med 1988; 168:463.
144. Hashimoto Y, Kagechika H, Shudo K Expression of retinoic acid 163. Hoang T, Levy B, Onetto N, et al. Tumor necrosis factor a stimulates
receptor genes and ligand-binding selectivity of retinoic acid the growth of the clonogenic cells of acute myeloblastic leukemia
recptors. Biochem Biophys Res Commun 1990; 166:1300. in synergy with granulocyte-macrophage colony-stimulating
145. Hassan HT, Rees JKH. Triple combination of retinoic acid + factor. J Exp Med 1989; 170:15.
naclacinomycin A + dimethylformamide induces differentiation 164. Hoffman SJ, Robinson WA. Use of differentiation-inducing agents
of human acute myeloid leukaemic blasts in primary culture. in the myelodysplastic syndrome and acute non-lymphocytic
Anticancer Res 1989; 9:647. leukemia. Am J Hematol 1988; 28:124.
146. Hast R, Axdorph S, Lauren L, et al. Absent clinical effects of 165. Hofmann WK, Ganser A, Ottmann OG et al. All-trans-retinoic
retinoic acid and isorethinoin treatment in the myelodysplastic acid, interferon-a, and granulocyte colony-stimulating factor as
syndrome. Hematol Oncol 1989; 7:297. combination therapy in patients with low risk myelodysplastic
147. Hattori T, Pack M, Bougnoux P, et al. Interferon-induced differ- syndromes. Blood 1997; 90 (Suppl. 1); abst. 366.
entiation of U937 cells. J Clin Invest 1983; 72:237. 166. Homma Y, Hozumi M, Abe E, et al. 1,25-Dihydroxyvitamin D3
148. Haussler MR, Baylink DJ, Hughes MR, et al. Assay of 1-alpha prolong survival time of mice inoculated with myeloid leukemia
25-dihydroxyvitamin D3: physiological and pathological modu- cells. Proc Natl Acad Sci USA 1983; 80:201.
lation of circulating hormone levels. Clin Endocrinol 1979; 167. Hossfeld DK, Weh H-J, Kleeberg UR. Low dose cytarabine:
5:157. chromosomal findings suggesting its cytostatic as well as differ-
149. Hayashi M, Okabe J, Hozumi M. Sensitization of resistant myeloid entiating effect. Leuk Res 1985; 9:329.
leukemia clone cells by anti-cancer drugs to factor-stimulating 168. Housset M, Daniel MT, Degos L. Small doses of ARA-C in the
differentiation. Gann 1979; 70:235. treatment of acute myeloid leukaemia: differentiation of myeloid
150. Hayashi SI, Gimble JM, Henley A, et al. Differential effects of leukaemia cells? Br J Haematol 1982; 51:125.
TGFβ1 on lympho-hemopoiesis in long-term bone marrow cultures. 169. Hozumi M. Fundamentals of chemotherapy of myeloid leukemia
Blood 1989; 74:1711. by induction of leukemia cell differentiation. Adv Cancer Res
151. Hellstrom E, Robert KH, Gahrton G, et al. Therapeutic effects of 1983; 38:121.
low-dose cytosine arabinoside, alpha-interferon, alpha-hydroxyvi- 170. Huang M, Chen S, Chi J-R, Lu J-X et al. Use of all trans-retinoic
tamin D3 and retinoic acid in acute leukemia and myelodysplastic acid in treatment of APL. Blood 1988; 72:567.
syndromes. Eur J Haematol 1988; 40:449. 171. Huang ME, Ye YC, Chen SR. All-trans retinoic acid with or
152. Hellstrom E, Robert KH, Samuelsson J et al. Treatment of myelodys- without low dose cytosine arabinoside in acute promyelocytic
plastic syndromes with retinoic acid and 1 alpha-hydroxy-vitamin leukemia. Report of 6 cases. Clin Med J 1987; 100:949.
562 Growth and differentiation factors as cancer therapeutics

172. Ikebuchi K, Ihle JN, Hirai Y, et al. Synergistic factors for stem stem cells by 1,25-dihydroxyvitamin D3 and its fluorinated ana-
cell proliferation: further studies of the target stem cells and the logues. Cancer Res 1984; 44:5624.
mechanism of stimulation by interleukin-1, interleukin-6, and 192. Koeffler HP, Kirfi K, Itri L. 1,25-dihydroxyvitamin D-3: in vivo
granulocyte colony-stimulating factor. Blood 1988; 72:2007. and in vitro effects on human preleukemic and leukemic cells.
173. Ishibashi T, Miller SL, Burstein SA. Type β transforming growth Cancer Treat Rep 1985; 69:1399.
factor is a potent inhibitor of murine megakaryocytopoiesis 193. Koeffler HP, Yelton L, Prokocimer L, et al. Study of the differen-
in vitro. Blood 1987; 69:1737. tiation of fresh myelogenous leukemic cells by compounds that
174. Ishibashi T, Kimura H, Shkiama Y, et al. Thrombopoietic effect induce a human promyelocytic leukemic cell line (HL-60) to dif-
of interleukin 6 (IL-6) in vivo in mice: comparison of the action ferentiate. Leuk Res 1985; 9:73.
of erythropoietin (Epo) and granulocyte colony-stimulating factor 194. Koeffler HP, Tobler A, Munker R. Tumor necrosis factor: role in
(G-CSF) in combination with IL-6 on thrombocytopoiesis. Blood normal and abnormal hematopoiesis. In: Gale RP, Golde DW, eds.
1989; 74(suppl 1):18 (abstr). Recent advances in leukemia and lymphoma. UCLA Symposia on
175. Iwasaki Y, Iwasaki J, Feake HC. Growth inhibition of human Molecular and Cellular Biology, New Series, 1987; 61:433.
breast cancer cells induced by calcitonin. Biochem Biophys Res 195. Koeffler HP, Heitjan D, Mertelsmann R, et al. Randomized study
Commun 1983; 110:235. of 13-cis-retinoic acid v placebo in the myelodysplastic disorders.
176. Jetten AM. Action of retinoids and phorbol esters on cell growth Blood 1988; 71:703.
and the binding of epidermal growth factor. Ann NY Acad Sci 196. Kohlhepp EA, Condon ME, Hamburger AW. Recombinant
1981; 119:200. human interferon alpha enhancement of retinoic-acid-induced
177. Kahn AJ, Simmons DJ. Monocyte origin of osteoclasts. Nature differentiation of HL-60 cells. Exp Hematol 1987; 15:414.
1975; 258:325. 197. Koike K, Nakahata T, Takagi M, et al. Synergism of BSF-2/inter-
178. Kahn AJ, Stewart CC, Teitelbaum SL. Contact mediated bone leukin 6 and interleukin 3 on development of multipotential
resorption by human monocytes in vitro. Science 1978; 199:988. hemopoietic progenitors in serum-free culture. J Exp Med 1988;
179. Kamijo R, Takeda K, Nagumo M, et al. Effects of combinations 168:879.
of transforming growth factor-β1 and tumor necrosis factor on 198. Kouzarides T. Histone acetylases and deacetylases in cell prolif-
induction of differentiation of human myelogenous leukemic cell eration. Curr Opin Genet Dev 1999; 9:40.
lines. J Immunol 1990; 144:1311. 199. Kuribayashi T, Tanaka H, Abe E, et al. Functional defect of variant
180. Kaufman DC, Baer MR, Gao X, et al. Enhanced expression of the clones of a human myeloid leukemia cell line (HL-60) resistant to
granulocyte-macrophage colony stimulating factor gene in acute 1,25-dihydroxyvitamin D3. Endocrinology 1983; 113:1992.
myeloblastic leukemia cells following in vitro blast cell enrich- 200. Kurland JI, Pelus LM, Ralph P, et al. Induction of prostaglandin
ment. Blood 1988; 72:1329. E synthesis in normal and neoplastic macrophages: role for
181. Keller JR, Mantel C, Sing GK, et al. Transforming growth factor colony-stimulating factor(s) distinct from myeloid progenitor
beta 1 selectively regulates early murine hematopoietic progeni- cell proliferation. Proc Natl Acad Sci USA 1979; 76:2326.
tors and inhibits the growth of IL-3-dependent myeloid leukemia 201. Kurzrock R, Estey E, Talpaz. M. All-trans retinoic acid: toler-
cell lines. J Exp Med 1988; 168:737. ance and biological effect in myelodysplastic syndrome. J Clin
182. Keller JR, Sing GK, Ellingsworth LR, et al. Transforming growth Oncol 1993; 11:1489.
factor beta: possible roles in the regulation of normal and leukemic 202. Lang R, Metcalf N, Gough N, et al. Expression of a hemopoietic
hematopoietic cell growth. J Cell Biochem 1989; 39:175. growth factor cDNA in a factor-dependent cell line results in
183. Kendrup G, Benedix-Hansen K, Pedersen B, Ellegaaard J. 13-cis- autonomous growth and tumorigenicity. Cell 1985; 43:531.
retinoic acid treatment of myelodysplastic syndromes. Leuk Res 203. Largman C, Detmer K, Corral JC, et al. Expression of retinoic
1987; 11:7. acid receptor alpha MRNA in human leukemia cells. Blood 1989;
184. Kishimoto T. The biology of interleukin-6. J Am Soc Hematol 74:99.
1989; 74:1. 204. Larrick JW, Ficher DG, Anderson SJ, et al. Characterization of a
185. Kini AR, Peterson LC, Tallman MS, Lingen MW. Angiogenesis human macrophage-like cell line stimulated in vitro: a model of
in acute promyelocytic leukemia: inhibition by vascular endothe- macrophage functions. J Immunol 1980; 125:6.
lial growth factor and inhibition by all trans retinoic acid. Blood 205. Leoni F, Ciolli S, Longo G et al. 13-cis-retinoic acid treatment of
2001; 97:3919. myelodysplastic syndromes. Acta Haematol.1988; 80:8.
186. Kizaki M, Norman AW, Bishop JE, et al. 1,25-dihydroxyvitamin D3 206. Ley TJ, DeSimone J, Noguchi CT, et al. 5-Azacytidine increases
receptor RNA: expression in hematopoietic cells. Blood 1991; γ globin synthesis and reduces the proportion of dense cells in
71:1238–1247. patients with sickle-cell anemia. Blood 1983; 62:370.
187. Klien B, Zhang XG, Jourdan M, et al. Paracrine rather than auto- 207. Lian Z, Wang L, Yamaga S et al. Genomic and proteomic analy-
crine regulation of myeloma-cell growth and differentiation by sis of the myeloid differentiation program. Blood 2001; 98:513.
interleukin-6. Blood 1989; 73:517. 208. Lin BC, Hong SH, Krig S, et al. A conformational switch in nuclear
188. Klijn JGM, Berns PMJJ, Schmitz PIM, Foekens JA. The clinical hormone receptors is involved in coupling hormone binding to
significance of epidermal growth factor receptor (EGF-R) in human corepressor release. Molecular & Cellular Biology 1997; 17:6131.
breast cancer: a review on 5232 patients. Endocrine Reviews 1992; 209. Lin J, Sartorelli AC. Stimulation by interferon of the differentia-
13:3–17. tion of human promyelocytic leukemia (HL-60) cells produced by
189. Koeffler HP. Induction of differentiation of human acute myeloge- retinoid acid and actinomycin D. J Interferon Res 1987; 7:379.
nous leukemia cells: therapeutic implications. Blood 1983; 62:709. 210. Lin FK, Suggs S, Lin C-H, et al. Cloning and expression of the
190. Koeffler H, Bar-Eli M, Territo M. Phorbol ester effect on differ- human erythropoietin gene. Proc Natl Acad Sci USA 1985;
entiation of human myeloid leukemia cell lines blocked at different 82:7580–7584.
stages of maturation. Cancer Res 1981; 41:919. 211. Lindemann W-D, Ludwig W, Oster R. High-level secretion of
191. Koeffler HP, Amatruda T, Ikekawa N, et al. Induction of mac- tumor necrosis factor-alpha contributes to hematopoietic failure
rophage differentiation of human normal and leukemic myeloid in hairy cell leukemia. Blood 1989; 73:880.
Kapil Mehta et al. 563

212. Liu T-X, Zhang J-W, Tao J et al. Gene expression network under- 232. Marks PA, Rifkind RA. Hexamethylene bisacetamide-induced
lying retinoic acid-induced differentiation of acute promyelocytic differentiation of transformed cells: molecular and cellular effects
leukemia cells. Blood 2000; 96:1496. and therapeutic application. Int J Cell Cloning 988; 6:230.
213. Lombardo FA, Ward FT, Chun HG, et al. Phase I trial of oral 233. Martin SJ, Bradley JG, Cotter TG. HL-60 cells induced to dif-
hemamethylene bisacetamide (HMBA). Proc Annu Meet Am Soc ferentiate towards neutrophils subsequently die via apoptosis.
Clin Oncol 1989; 8:A297. Clin Exp Immunol 1990; 79:448.
214. Lotem J, Sachs L. Different blocks in the differentiation of myeloid 234. Matsui T, Nakao U, Kobayashi N, et al. Phenotypic differentia-
leukemic cells. Proc Natl Acad Sci USA 1974; 71:3507. tion-linked growth inhibition in human leukemia cells by active
215. Lotem J, Sachs L. In vivo control of differentiation of myeloid vitamin D3 analogues. Int J Cancer 1984; 33:193.
leukemic cells by cyclosporine A and recombinant interleukin-1α. 235. Maurer AB, Ganser A, Seipelt G et al. Changes in erythroid pro-
Blood 1988; 72:1595. genitor cell and accessory cell compartments in patients with
216. Lotem J, Sachs L. Indirect induction of differentiation of normal myelodysplastic syndromes during treatment with all-trans retin-
and leukemic myeloid cells by recombinant interleukin 1. Leuk oic acid and haemopoietic growth factors. Br J Haematol 1995;
Res 1989; 13:13. 89:449.
217. Lotem J, Sachs L. Hematopoietic cytokines inhibit apoptosis 236. McCarthy DM, San Miguel JF, Freake HC, et al. 1,25-
induced by transforming growth factor β1 and cancer chemother- Dihydroxyvitamin D3 inhibits proliferation of human promyelo-
apy compounds in myeloid leukemic cells. Blood 1992; 80:1750. cytic leukaemia (HL-60) cells and induces monocyte-macrophage
218. Lotem J, Lipton J, Sachs L. Separation of different molecular differentiation in HL-60 and normal human bone marrow cells.
forms of macrophage- and granulocyte-inducing proteins for nor- Leuk Res 1983; 7:51.
mal and leukemic myeloid cells. Int J Cancer 1980; 25:763. 237. McCarthy DM, Hibbins JA, Goldman JM. A role for 1,25-dihy-
219. Lotem J, Berrebi A, Sachs L. Screening for induction of differen- droxyvitamin D3 in control of bone-marrow collagen deposition?
tiation and toxicity to blast cells by chemotherapeutic compounds Lancet 1984; 1:78.
in human myeloid leukemia. Leuk Res 1985; 9:249. 238. Mehta K, Sadeghi T, McQueen T, Lopez-Berestein G. Liposome
220. Lowenberg B, Salem M, Delwel R. Effects of recombinant multi- encapsulation circumvents the hepatic clearance mechanisms of
CSF, GM-CSF, G-CSF and M-CSF on the proliferation and mat- all-trans-retinoic acid. Leuk Res 1994;18:587.
uration of human AML in vitro. Blood Cells 1988; 14:539. 239. Mehta K, McQueen T, Neamati N et al. Activation of retinoid
221. Lowe DG, Wylla N. Bombara M, et al. Genomic cloning and receptors RARα and RXRα induces differentiation and apopto-
heterologous expression of human differentiation-stimulating sis, respectively, in HL-60 cells. Cell Growth Diff 1996; 7:179.
factor. DNA 1989; 8:351. 240. Metcalf D. The roles of stem cell-renewal and autocrine growth
222. Lu L, Broxmeyer HE, Pelus LM, et al. Detection of Luxol-fast- factor production in the biology of myeloid leukemia. Cancer Res
blue positive cells in human promyelocytic leukemia cell line 1989; 49:2305.
HL-60. Exp Hematol 1982; 9:887. 241. Metcalf D. Actions and interactions of G-CSF, LIF, and IL-6 on
223. Lu L, Welte K, Gabrilove JL, et al. Effects of recombinant human normal and leukemic murine cells. Leukemia 1989; 3:349.
tumor necrosis factor a, recombinant human γ-interferon, and 242. Metcalf D, Gearing DP. A myelosclerotic syndrome in mice
prostaglandin E on colony formation of human hematopoietic engrafted with cells producing high levels of leukemia inhibitory
progenitor cells stimulated by natural human pluripotent colony- factor (LIF). Leukemia 1989; 3:847.
stimulating factor, pluripoietin a, and recombinant erythropoietin 243. Metcalf D, Gearing DP. Fatal syndrome in mice engrafted with
in serum-free cultures. Cancer Resp 1986; 46:4357. cells producing high levels of the leukemia inhibitory factor. Proc
224. MacDonald PN, Baudino TA, Tokumaru H, et al. Vitamin D Natl Acad Sci USA 1989; 86:5948.
receptor and nuclear receptor coactivators: crucial interactions in 244. Metcalf D, Hilton DJ, Nicola NA. Clonal analysis of the actions
vitamin D-mediated transcription. Steroids 2001:66:171. of the murine leukemia inhibitory factor on leukemic and normal
225. Maekawa T, Metcalf D. Clonal suppression of HL-60 and U937 murine hemopoietic cells. Leukemia 1988; 2:216.
cells by recombinant human leukemia inhibitory factor in combi- 245. Mezzetti G, Bagnara G, Monti MG, et al. 1,25-Dihydroxy-
nation with GM-CSF or G-CSF. Leukemia 1989; 3:270. cholecalciferol and human histocytic lymphoma cell (U937): the
226. Majeska RJ, Rodan GA. The effect of 1,25(OH)2 D3 on alkaline presence of receptor and inhibition of proliferation. Life Sci
phosphatase in osteoblastic osteosarcoma cells. J Biol Chem 1984; 34:2185.
1982; 257:3362. 246. Michaelewicz R, Lotem J, Sachs L. Cell differentiation and ther-
227. Mangelsdorf DJ, Kliewer SA, Kakizuka A et al. Retinoid receptors. apeutic effect of low doses of cytosine arabinoside in human
Recent Progress in Hormone Research, 1993; 48:99. myeloid leukemia. Leuk Res 1984; 8:783.
228. Mangelsdorf DJ, Koeffler HP, Donaldson CA, et al. 1,25- 247. Miyauchi J, Wang C, Kelleher CA, et al. The effects of recombi-
Dihydroxyvitamin D3-induced differentiation in a human promy- nant CSF-1 on the blast cells of acute myeloblastic leukemia in
elocytic leukemia cell line (HL-60): receptor-mediated maturation suspension culture. J Cell Physiol 1988; 135:55.
to macrophage-like cells. J Cell Biol 1984; 98:391. 248. Miyaura C, Abe E, Kuribayashi T, et al. 1-α25-Dihydroxyvitamin D3
229. Manloagas SC, Haussler MR, Deftos LJ. 1,25-Dihydroxyvitamin induces differentiation of human myeloid leukemia cells. Biochem
D3 receptor-like macromolecule in rat osteogenic sarcoma cell Biophys Res Commun 1981; 102:937.
lines. J Biol Chem 1980; 255:4414. 249. Miyaura C, Abe E, Honma Y, et al. Cooperative effect of
230. March C, Mosley B, Larsen A, et al. Cloning, sequence and 1,25-dihydroxyvitamin D3 and dexamethasone in inducing dif-
expression of two distinct human interleukin-1 complementary ferentiation of mouse myeloid leukemia cells. Arch Biochem
DNA. Nature 1985; 315:641. Biophys 1983; 227:397.
231. Marks PA, Breslow R, Rifkind RA, et al. Polar/apolar chemical 250. Miyaura C, Abe E, Suda T, et al. Alternative differentiation of
inducers of differentiation of transformed cells: strategies to human promyelocytic leukemia cells (HL-60) induced selec-
improve therapeutic potential. Proc Natl Acad Sci USA 1989; tively by retinoic acid and 1,25-dihydroxyvitamin D3. Cancer
86:6358. Res 1985; 45;4244.
564 Growth and differentiation factors as cancer therapeutics

251. Miyaura C, Onozaki K, Akiyama Y, et al. Recombinant human 269. Moreau J-F, Donaldson DD, Bennett F, et al. Leukaemia inhibi-
interleukin-6 (B-cell stimulatory factor 2) is a potent inducer of tory factor is identical to the myeloid growth factor human inter-
differentiation of mouse myeloid leukemia cells (M1). FEBS Lett leukin for DA cells. Nature 1988; 336:690.
1988; 234:17. 270. Morosetti R, Park DJ, Chumakov AM et al. A novel, myeloid
252. Miyaura C, Jin CH, Yamaguchi Y, et al. Production of interleukin 6 transcription factor, C/EBP epsilon, is upregulated during granu-
and its relation to the macrophage differentiation of mouse locytic, but not monocytic, differentiation. Blood 1997;90:2591.
myeloid leukemia cells (M1) treated with differentiation-inducing 271. Motoji T, Hoang T, Tritchler D, McCulloch EA. The effect of
factor and 1α, 25-dihydroxyvitamin D3. Biochem Biophys Res 5-aza-cytidine and its analogues on blast cell renewal in acute
Commun 1989; 158:660. myeloblastic leukemia. Blood 1985; 65:894.
253. Moloney WC, Rosenthal DS. Treatment of early acute non-lymphatic 272. Muller R, Muller D, Guilbert L. Differential expression of c-fos
leukemia with low dose cytosine arabinoside. Haematol Blood in hematopoietic cells: correlation with differentiation of myelo-
Transfus 1981; 26:59. monocytic cells in vitro. EMBO J 1984; 3:1887.
254. Momparler RL, Bouchard J, Onetto N, et al. 5-Aza-2′-deoxycytidine 273. Muller K, Svenson M, Bendtzen K. 1-Alpha, 25-dihydroxyvita-
therapy in patients with acute leukemia inhibits DNA methylation. min D3 and a novel vitamin D analogue MC 903 are potent inhibi-
Leuk Res 1984; 8:181. tors of human interleukin 1 in vitro. Immunol Lett 1988; 17:361.
255. Moore M. Prediction of relapse and remission in AML by marrow 274. Munroe E, Sugiura M, Griffin J, Kufe D. Effect of ara-A on dif-
culture criteria. Blood Cells 976; 2:109. ferentiation and proliferation of HL-60 cells. Leuk Res 1984;
256. Moore M. Agar culture in CML and blastic transformation. Ser 8:355.
Haematol 1977; 8:11. 275. Murao S, Gemmell MA, Callaham MF, et al. Control of mac-
257. Moore M. Proliferation and differentiation control mechanisms in rophage cell differentiation in human promyelocytic HL-60 leuke-
myeloid leukemia. In: Mehich H, ed. Biological responses in cancer mia cells by 1,25-dihydroxyvitamin D3 and phorbol-12-myristate-13
– progress toward potential applications, vol. 2. New York: Plenum, acetate. Cancer Res 1983; 43:4989.
1984:93. 276. Murphy M, Perussia B, Trinchieri G. Effects of recombinant
258. Moore MAS. Role of interleukin-1 in hematopoiesis. Immunol tumor necrosis factor, lymphotoxin, and immune interferon on
Res 1989; 8:165. proliferation and differentiation of enriched hematopoietic pre-
259. Moore MAS. Interactions between hematopoietic growth factors cursor cells. Exp Hematol 1988; 16:131.
in normal and leukemic stem cell differentiation. In: Diamond L, 277. Nagy L, Kao H-Y, Chakravarati D et al. Nuclear receptors repres-
Wolman SR, eds. Viral oncogenesis and cell differentiation. Ann sion mediated by a complex containing SMRT, mSin3A and histone
NY Acad Sci 1989; 567:171. deacetylase. Cell, 1997; 89:373.
260. Moore MAS. Coordinate actions of hematopoietic growth factors 278. Nagy L, Thomazy VA, Shipley GL et al. Activation of retinoid X
in stimulation of bone marrow function. In: Sporn MB, Roberts receptors induces apoptosis in HL-60 cell lines. Mol Cell
AB, eds. Handbook of experimental pharmacology – peptide Biol1995;15:3540.
growth factors and their receptors. 1990: Chap. 32 95 II:299. 279. Nagata K, Tohda S, Suzuki T, et al. Effects of recombinant human
261. Moore M, Sheridan A. The role of proliferation and maturation tumor necrosis factor on the self-renewal capacity of leukemic
factors in myeloid leukemia. In: Moore MAS, ed. Maturation blast progenitors in acute myeloblastic leukemia. Leukemia
factors in cancer. New York: Raven Press, 1982:361. 1989; 3:626.
262. Moore MAS, Warren DJ. Interleukin-1 and G-CSF synergism. 280. Najean Y, Pecking A. Refractory anemia with excess of blast
in vivo stimulation of stem cell recovery and hematopoietic cells: prognostic factors and effect of treatment with androgens or
regeneration following 5-fluorouracil treatment in mice. Proc cytosine arabinoside. Cancer 1976; 44:345.
Natl Acad Sci USA 1987; 84:7134. 281. Nara N, McCulloch E. Membranes replace irradiated blast cells
263. Moore M, Williams N, Metcalf D. In vitro colony formation by as growth requirement for leukemic blast progenitors in suspen-
normal and leukemic human hematopoietic cells: characterization sion culture. J Exp Med 1985; 162:1435.
of the colony-forming cells. J Natl Cancer Inst 1973; 50:603. 282. Nara N, Tohda S, Nagata K, et al. Inhibition of the in vitro growth
264. Moore M, Spitzer G, Williams N, et al. Agar culture studies in of blast progenitors from acute myeloblastic leukemia patients by
127 cases of untreated acute leukemia. The prognostic value of transforming growth factor-beta (TGF-beta). Leukemia 1989;
reclassification of leukemia according to in vitro growth charac- 3:572.
teristics. Blood 1974; 44:1. 283. Nervi C, Grippo JF, Sherman MI, et al. Identification and charac-
265. Moore MAS, Gabrilove JL, Welte K, Platzer E. Maturational factors terization of nuclear retinoic acid-binding activity in human
in leukemia. In: Reif AE, Mitchell MS, ed. Immunity to cancer. myeloblastic leukemia HL-60 cells. Proc Natl Acad Sci USA
New York: Academic 1985:513. 1989; 86:5854.
266. Moore MAS, Warren DJ, Souza L. Synergistic interaction 284. Newburger PE, Chovaniec ME, Greenburger JS, Cohen H.
between interleukin-1 and CSFs in hematopoiesis. In: Gale RP, Functional changes in human leukemic cell line HL-60. J Cell
Golde DW, eds. UCLA symposium on leukemia, recent advances Biol 1979; 82:315.
in leukemia and lymphoma. New York: Alan R. Liss, 1987:445. 285. Nicola N, Begley C, Metcalf D. Identification of the human ana-
267. Moore M, Welte K, Gabrilove J, et al. Biological activities of logue of a regulator that induces differentiation in murine leukaemia
recombinant human granulocyte-colony stimulating factor (rhG- cells. Nature 1985; 316:625.
CSF) and tumor necrosis factor: in vivo and in vitro analysis. 286. Niedel J, Kahane I, Lachman L, Cuartrecassas P. A subpopulation
Haematol Blood Transfus 1987; 31:210. of cultured human promyelocytic leukemia cells (HL-60) dis-
268. Moreau J-F, Bonneville M, Godard A, et al. Characterization of a plays the formyl peptide chemotactic receptor. Proc Natl Acad
factor produced by human T cell clones exhibiting eosinophil- Sci USA 1980; 77:1000.
activating and burst-promoting activities. J Immunol 1987; 287. Nilsson K, Forsbeck K, Gidlund M, et al. Surface characteristics of
138:3844. the U937 histiocytic lymphoma cell line: specific changes during
Kapil Mehta et al. 565

inducible morphologic and functional differentiation in vitro. In: 306. Peetre C, Gullberg U, Nilsson E, et al. Effects of recombinant
Net R, Gallo R, Graf T, Mannweiler K, Winkler H, eds. Haematology tumor necrosis factor on proliferation and differentiation of leu-
and blood transfusion, vol. 26. Berlin: Springer, 1981:215. kemic and normal hemopoietic cells in vitro. J Clin Invest 1986;
288. Ohta M, Massague J, Anklesaria P, et al. Two forms of transforming 78:1694.
growth factor-β distinguished by multipotential haematopoietic 307. Pelus LM, Ottmann OG, Nocka KH. Synergistic inhibition of
progenitor cells. Nature 1987; 329:539. human marrow granulocyte-macrophage progenitor cells by
289. Ohno R. Differentiation therapy of myelodysplastic syndromes prostaglandin E and recombinant interferon-α,-β and -γ and an
with retinoic acid. Leuk Lymph. 1992; 14:401. effect mediated by tumor necrosis factor. J Immunol 1988;
290. Okabe-Kado J, Honma Y, Hayashi M, et al. Inhibitory action of 140:479.
transforming growth factor-β on induction of differentiation of 308. Pelicano L, Li F, Schindler C, Chelbi-Alix MK. Retinoic acid
myeloid leukemia cells. Jpn J Cancer Res 1989; 80:228. enhances the expression of interferon-induced proteins. Evidence
291. Olsson IL, Breitman T. Induction of differentiation of the human for multiple mechanisms of action. Oncogene 1997;15:2349.
histiocytic lymphoma cell line U937 by retinoic acid and cyclic 309. Pennica D, Nedwin GE, Hayflick JS, et al. Human tumor necrosis
adenosine 3′:5′-monophosphate-inducing agents. Cancer Res factor: precursor structure, expression and homology to lympho-
1982; 42:3924. toxin. Nature 1985; 312:724.
292. Olsson IL, Breitman TR, Gallo RC. Priming of human myeloid 310. Perussia B, Lebman D, Ip S, et al. Terminal differentiation sur-
leukemic cell lines HL-60 and U937 with retinoic acid for dif- face antigens of myelomonocytic cells are expressed in human
ferentiation effects of cyclic adenosine 3′:5′-monophosphate- promyelocytic leukemia cells (HL-60) treated with chemical
inducing agents and a T-lymphocyte-derived differentiation inducers. Blood 1981; 58:836.
factor. Cancer Res 1982; 42:3928. 311. Piacibello W, Broxmeyer HE. Modulation of expression of
293. Olsson I. Gullberg U, Ivhed I, et al. Induction of differentiation of Ia(HLA-DR)-antigens on human monocyte cell line U937 by
the human histiocytic lymphoma cell line U937 by 1,25-dihy- gamma interferon and prostaglandin E and responsiveness of U937
droxycholicalciferol. Cancer Res 1983; 43:5862. colony forming cells to inhibition of lactoferrin, transferrin, acidic
294. Olsson I, Sarngadharan MG, Breitman TR, Gallo R. Isolation and isoferritins and prostaglandin E. Blood 1983; (suppl. 1) 62:86.
characterization of a T-lymphocyte-derived differentiation inducing 312. Picozzi VJ, Swanson GF, Morgan R, et al. 13-cis-retinoic acid
factor for the myeloid leukemic cell line HL-60. Blood 1984; treatment of myelodysplastic syndromes. J Clin Oncol 1986;
63:510. 4:589.
295. Onozaki K, Tamatani T, Hashimoto T, et al. Growth inhibition 313. Pike MC, Fischer DG, Koren H, Snyderman R. Development of
and augmentation of mouse myeloid cell line differentiation by specific receptors for N-formylated chemotactic peptides in a
interleukin 1. Cancer Res 1987; 47:2397. human monocytic cell line stimulated with lymphokines. J Exp
296. Onozaki K, Akiyama Y, Okano A, et al. Synergistic regulatory Med 1980; 152:31.
effects of interleukin 6 and interleukin 1 on the growth and dif- 314. Pinto A, Attadia V, Fusco A, et al. 5-Aza-2′-deoxycytidine
ferentiation of human and mouse myeloid leukemic cell lines. induces terminal differentiation of leukemic blasts from patients
Cancer Res 1989; 49:3602. with acute myeloid leukemias. Blood 1984; 64:922.
297. Onozaki K, Urawa H, Tamatani T, et al. Synergistic interactions of 315. Platzer E, Welte K, Gabrilove J, et al. Biological activities of
interleukin 1, interferon-β and tumor necrosis factor in terminally human pluripotent hemopoietic colony stimulating factor on normal
differentiating mouse myeloid leukemic cell line (M1). J Immunol and leukemic cells. J Exp Med 1985; 162:1788.
1988; 140:112. 316. Pluznik DH. Synergistic activity of interleukin-1 alpha and inter-
298. Ossenkoppele GJ, Wijermans PW, Nauta JJP, et al. Maturation leukin-6 induces differentiation of myeloid cells into mature
induction in freshly isolated human myeloid leukemic cells, macrophages. Exp Hematol 1988; 16:504.
1.25(OH)2 vitamin D3 being the most potent inducer. Leuk Res 317. Potter GK, Mohamed AN, Dracapoli NC, et al. The action of
1989; 13:609. 1,25(OH)2 D3 in nude mice bearing transplantable human myel-
299. Ottmann OG, Pelus LM. Differential proliferative effects of ogenous leukemic cell lines. Exp Hematol 1985; 13:722.
transforming growth factor-β on human hematopoietic progeni- 318. Powell BL, Capizzi RL, Jackson DV, et al. Low dose ara-C for
tor cells. J Immunol 1988; 140:2661. patients with myelodysplastic syndromes. Leukemia 1988;
300. Ozpolat B, Berestein GL, Mehta K. ATRA(ouble) in the treat- 2:153.
ment of acutepromyelocytic leukemia. J Biol Regul Homeost 319. Prigent SA, Lemoine NR. The type 1 (EGFR-related) family of
Agents 2001; 15:107. growth factor receptors and their ligands. Progress in Growth
301. Pakkala S, de Vos S, Elstner E, et al. Antileukemic activities and Factor Research 1992; 4:1–24.
effects on serum calcium of three novel vitamin D3 analogs. 320. Provendini DM, Tsoulas CD, Deftos LJ, Manolagas SC.
Blood (Abstract) (Suppl.) 1993; 82:255a. 1,25-Dihydroxyvitamin D3 receptors in human leukocytes. Science
302. Palacios R, Ivhed I, Sideras P, et al. Accessory function of human 1983; 221:1181.
tumor cell lines I., production of interleukin 1 by the human his- 321. Pusztai L, Lewis CE, Lorenzen J, McGee JO’D. Growth factors:
tocytic lymphoma cell line U937. Eur J Immunol 1982; 12:895. regulation of normal and neoplastic growth. Journal of Pathology
303. Pantazis P, Lazarou S, Papadopoulos N. Isoenzymes of lactate 1993; 169:191–201.
dehydrogenase in human leukemic cells in culture treated with 322. Raff MC. Social controls on cell survival and cell death. Nature
inducers of differentiation. J Cell Biol 1981; 90:396. 1992; 356:397.
304. Paoletti F, Mocali A. Changes in CuZn-superoxide dismutase 323. Ralph P, Moore MAS, Nilsson K. Lysozyme synthesis by estab-
during induced differentiation of murine erythroleukemia cells. lished human and murine histiocytic cell lines. J Exp Med 1976;
Cancer Res 1988; 48:6674. 143:1528.
305. Parthasarathy R, Mehta K. Altered metabolism of all-trans-retinoic 324. Ralph P, Williams N, Moore MAS, Litcofsky P. Induction of
acid in liposome-encapsulated form. Cancer Lett 1998; 134:121. antibody-dependent and nonspecific tumor killing in human
566 Growth and differentiation factors as cancer therapeutics

monocyte leukemic cells by nonlymphocyte factors and phorbol 345. Salem M, Delwel R, Touw I, et al. Modulation of colony stimu-
ester. Cell Immunol 1982; 71:215. lating factor-(CSF) dependent growth of acute myeloid leukemia
325. Ralph P, Harris P, Punjabi CJ, et al. Lymphokine inducing “termi- by tumor necrosis factor. Leukemia 1990; 4:37.
nal differentiation” of the human monoblast leukemia line U937: 346. Santoli D, Yang Y-C, Steven CC, et al. Synergistic and antagonis-
a role for gamma interferon. Blood 1983; 62:1169. tic effects of recombinant human interleukin (IL) 3, IL-1a, granu-
326. Raza A, Preisler H. Evidence of in vivo differentiation in myelo- locyte and macrophage colony-stimulating factors (G-CSF and
blasts labeled with bromodeoxyuridine. Cancer J 1986; 1:15. M-CSF) on the growth of GM-CSF-dependent leukemic cell
327. Razin A, Levine A, Kafri T, et al. Relationship between tran- lines. J Immunol 1987; 139:3348.
sient DNA hypomethylation and erythroid differentiation of 347. Sato T, Takusagawa K, Asso N, Konno K. Antitumor effect of 1
murine erythroleukemia cells. Proc Natl Acad Sci USA 1988; alpha-hydroxyvitamin D3. Tohoku J Exp Med 1982; 138:445.
85:9003. 348. Sawyer S, Krantz S, Luna J. Identification of the receptor for
328. Rees J. The molecular biology of retinoic acid receptors: orphan erythropoietin by cross-linking to Friend virus-infected erythroid
from good family seeks home. Brit J Dermatol 1992;126:97. cells. Proc Natl Acad Sci USA 1987; 84:3690.
329. Reily IAG, Kozlowski R, Russell NH. Heterogenous mechanisms 349. Schif RD, Stuart RK. Treatment of myelodysplastic syndromes
of autocrine growth of AML blasts. Br J Haematol 1989; 72:363. (MDS) and poor-prognosis acute myeloid leukemias (PP-AML)
330. Reitsma PH, Rothberg PG, Astrin SM, et al. Regulation of myc with low-dose cytarabine (LDS) plus 13-cis-retinoic acid (RA).
gene expression in HL-60 leukemia cells by a vitamin D metabo- Proc Annu Meet Am Soc Clin Oncol 1989; 8:A801.
lite. Nature 1983; 306:492. 350. Schwartz EL, Brown BJ, Nierenburg M, et al. Evaluation of marcel-
331. Richon VM, Ramsay RG, Rifkind RA, et al. Modulation of the lomycin using an in vivo model for studying drug-induced leukemia
c-myb, c-myc and p42 on MRNA and protein levels during cell differentiation. Proc Am Assoc Cancer Res 1982; 23:173.
induced murine erythroleukemia cell differentiation. Oncogene 351. Schwartz EL, Sartorelli AC. Structure-activity relationships for the
1989; 4:165. induction of differentiation of HL-60 human acute promyelocytic
332. Rigby WFC, Shen L, Ball ED, et al. Differentiation of a human leukemia cells by anthracyclines. Cancer Res 1982; 42:2651.
monocytic cell line by 1,25-dihydroxyvitamin D3 (calcitriol): a 352. Schwartz EL, Snoddy JR, Kreutter D, et al. Synergistic induction
morphological, phenotypic and functional analysis. Blood 1984; of HL-60 differentiation by 1alpha, 25-dihydroxyvitamin D3 and
64:1110. dimethylsulfoxide (DMSO) (meeting abstract). Proc Am Assoc
333. Robertson KA, Emami B, Collins SJ. Retinoic acid-resistant Cancer Res 1983; 23:71.
HL-60R cells harbor a point mutation in the retinoic acid receptor 353. Senior RM, Cambell EJ, Landis JA, et al. Elastase of U937
ligand-binding domain that confers dominant negative acitivity. monocyte-like cells. J Clin Invest 1982; 69:384.
Blood 1992; 80:1885. 354. Shabo Y, Lotem J, Rubinstein M, et al. The myeloid blood cell
334. Robertson KA, Emami B, Lueller L, Collins SJ. Multiple mem- differentiation-inducing protein MGI-2A is interleukin-6. Blood
bers of the retinoic acid receptor family are capable of mediating 1988; 72:2070.
the granulocytic differentiation of HL-60 cells. Mol Cell Biol 355. Shen L, Guyre PM, Fanger MW. Direct stimulation of ADCC by
1992; 12:3743. cloned gamma interferon is not ablated by glucocorticoids: stud-
335. Rosenblum MG, Donato NJ. Tumor necrosis factor alpha: a mul- ies using a human monocyte-like cell line (U937). Mol Immunol
tifaceted peptide hormone. Crit Rev Immunol 1989; 9:21. 1983; 21:167.
336. Rouis M, Thomopoulos P, Louache F, et al. Differentiation of 356. Shiina Y, Abe E, Miyaura C, et al. Biological activity of 24,
U-937 human monocyte-like cell line by 1,25-dihydroxyvitamin 24-difluoro-1alpha, 25-dihydroxyvitamin D3 and 1-alpha,
D3 or by retinoic acid. Exp Cell Res 1985; 157:539. 25-dihydroxyvitamin D3–26, 23-lactone in inducing differentia-
337. Rovera G, O’Brien T, Diamond L. Induction of differentiation in tion of human myeloid leukemia cells. Arch Biochem Biophys
human promyelocytic leukemia by tumor promoters. Science 1983; 220:90.
1977; 204:868. 357. Silverman LR, Davis RB, Holland JF, et al. 5-Azacytidine (AZ)
338. Rovera G, Santoli D, Damsky C. Human promyelocytic leukemic as a low dose continuous infusion is an effective therapy for
cells in culture differentiate into macrophage-like cells when patients with myelodysplastic syndromes (MDS). Proc Annu
treated with a phorbol diester. Proc Natl Acad Sci USA 1979; Meet Soc Clin Oncol 1989; 8:A768.
76:2779. 358. Sing GK, Keller JR, Ellingsworth LR, et al. Transforming growth
339. Rovera G, Olashaw N, Meo P. Terminal differentiation in human factor beta selectively inhibits normal and leukemic human bone
promyelocytic leukemic cells in vitro by 6-thioguanine. Cancer marrow cell growth in vitro. Blood 1988; 72:1504.
Lett 1980; 10:33. 359. Sing GK, Keller JR, Ellingsworth LR, et al. Transforming growth
340. Rowinsky EK, Ettinger DS, McGuire WP, et al. Prolonged infu- factor-β1 enhances the suppression of human hematopoiesis by
sion of hemaxamethylene bisacetamide: a phase I and pharmaco- tumor necrosis factor-α or recombinant interferon-α. J Cell
logical study. Cancer Res 1987; 47:5788. Biochem 1989; 39:107.
341. Ruscetti FW, Sing G, Ruscetti SK, et al. The role of transforming 360. Slack JL. Biology and treatment of acute progranulocytic leuke-
growth factor-beta in the regulation of normal and leukemic mia. Curr Opin Hematol 1999; 6:236.
hematopoiesis. Exp Hematol 1988; 16:417. 361. Sporn MB, Roberts AB, Goodman DS. (Eds) The Retinoids:
342. Ruutu P, Ruutu T, Vuopio P, et al. Defective chemotaxis in mono- Biology, Chemistry, and Medicine 1994; Raven Press, NY.
somy-7. Nature 1977; 265:146. 362. Sporn M, Todaro G. Autocrine secretion and malignant transfor-
343. Sachs L. Control of normal cell differentiation and the pheno- mation of cells. N Engl J Med 1980; 303:878.
topic reversion of malignancy in myeloid leukemia. Nature 1978; 363. Stevens VL, Owens NE, Winton EF, et al. Modulation of retinoic
274:535. acid-induced differentiation of human leukemia (HL-60) cells by
344. Salahuddin SZ, Markham PD, Ruscetti FW, Gallo RC. Long- serum factors and sphingamine. Cancer Res 1990; 50:222.
term suspension cultures of human cord blood myeloid cells. 364. Sun GL, Huang YG, Chang XF, Jiang GS, Zhang T. Clinical
Blood 1981; 58:931. study on all-trans retinoic acid in treatment of 544 cass of acute
Kapil Mehta et al. 567

promyelocytic leukemia treated. Clin J Hematol 1992; 383. Trinchieri G, Rosen M, Perussia B. Retinoic acid cooperates with
13:135–137. tumor necrosis factor and immune interferon in inducing differ-
365. Sundstrom C, Nilsson K. Establishment and characterization of a entiation and growth inhibition of the human promyelocytic
human histiocytic lymphoma cell line (U937). Int J Cancer 1976; leukemic cell line HL-60. Blood 1987; 69:1218.
17:565. 384. Tsoukas C, Provvedine DM, Manolagas SV. 1,25-Dihydroxy-
366. Sutherland GR, Baker E, Hyland VJ, et al. The gene for human leu- vitamin D3: a novel immunoregulatory hormone. Science 1984;
kemia inhibitory factor (LIF) maps to 22q12. Leukemia 1989; 3:9. 224:1438.
367. Svet-Moldavskaya I, Arlin Z, Svet-Moldavskaya G. Induction by 385. Turner RT, Puzas JE, Forte MD, et al. In vitro synthesis of
tumor-promoting phorbol diester of colony-stimulating activity 1,25(OH)2 D3 by isolated calvarial cells. Proc Natl Acad Sci USA
in human myeloid leukemia cells transformed to macrophage- 1980; 7i7:5720.
mimicking cells. Cancer Res 1981; 41:4335. 386. Ueno H, Kizaki M, Matsushita H et al. A novel retinoic acid
368. Swanson GF, Picozzi VJ, Morgan R et al. Response of hematopo- receptor-selective antagonist inhibits differentiation and apopto-
etic percursors to 13-cis-retinoic acid and 1.25 dihydroxyvitamin D3 sis of HL-60 cells: implications of RAR-mediated signals in
in myelodysplastic syndromes. Blood 1986; 67:1154. myeloid leukemia cells. Leuk Res 1998; 22:517.
369. Takeda K, Minowada J, Bloch A. Kinetics of appearance of 387. Van Beek W, Tulp A, Egbers-Bogards M, et al. Continuous
differentiation-associated characteristics in ML-1, a line of expression of cancer-related fucosyl glycopeptides on surface of
human myeloblastic leukemia cells, after treatment with human promyelocytic leukemic cells (HL-60) following terminal
12-O-tetradecanoylphorbol-13-acetate, dimethyl sulfoxide, or differentiation in vitro. Cancer Res 1982; 42:5222.
1-β-D-arabinofuranosylcytosine. Cancer Res 1982; 42:5152. 388. Van Beek W, Tulp A, Bolscher J, et al. Transient versus perma-
370. Takeda K, Iwamoto S, Sugimoto H, et al. Identity of differentiation nent expression of cancer-related glycopeptides on normal versus
inducing factor and tumour necrosis factor. Nature 1986; 323:338. leukemic myeloid cells coinciding with marrow egress. Blood
371. Takenaga K, Hozumi M, Sakagami Y. Effect of retinoids on 1984; 63:170.
induction of differentiation of cultured mouse myeloid leukemia 389. Van der Schoot CE, Jansen P, Porter M, et al. Interleukin-6 and
cells. Cancer Res 1980; 40:914. interleukin-1 production in acute leukemia with monocytoid
372. Tamatani T, Urawa H, Hashimoto T, et al. Tumor necrosis factor differentiation. Blood 1989; 74:2081.
as an interleukin 1-dependent differentiation inducing factor 390. Van Snick J, Cayphas S, Szikora J-P, et al. cDNA cloning of
(D-Factor) for mouse myeloid leukemic cells. Biochem Biophys murine interleukin-HP1: homology with human interleukin-6.
Res Commun 1987; 143:390. Eur J Immunol 1988; 18:193–197.
373. Tanabe O, Kawano M, Tanaka H, et al. BSF-2/IL-6 does not aug- 391. Venditti A, Scimo MT, Del Poeta G et al. Recombinant inter-
ment Ig secretion but stimulates proliferation in myeloma cells. feron-apha 2a. thymopentin and low doses of cytosine arabinoside
Am J Hematol 1989; 31:258. for the treatment of myelodysplastic syndromes: a pilot study.
374. Tanaka Y, Deluca HF, Schnoes HK, et al. 24, 24-Difluoro-1.25- Leuk Lympho. 1995; 16:335.
dihydroxyvitamin D3: In vitro production, isolation, and biologi- 392. Visani G, Cenacchi A, Tosi P, et al. All-trans retinoic acid
cal activity. Arch Biochem Biophys 1980; 199:473. improves erythropoiesis in myelodysplastic syndromes. Br J
375. Tessier N, Hoang T. Transforming growth factor beta inhibits the Haematol 1992; 81:444.
proliferation of the blast cells of acute myeloblastic leukemia. 393. Visani G, Tosi P, Manfroi S et al. All-trans retinoic acid in the treat-
Blood 1988; 72:159. ment of myelodysplastic syndromes. Leuk Lympho 1995; 19:277.
376. Thesingh CW, Burger EH. Origin of osteoclast progenitor cells 394. Wahl SM, Hunt DA, Wakefield LM, et al. Transforming growth
from central blood cell forming organs. Calcif Tissue Int 1981; factor type beta induces monocyte chemotaxis and growth factor
335:108. production. Proc Natl Sci USA 1987; 84:5788.
377. Tobler A, Gasson J, Reichel H, et al. Granulocyte-macrophage 395. Walsh JH, Karnes WE, Cuttitta F, Walker A. Autocrine growth
colony-stimulating factor. Sensitive and receptor-mediated regu- factors and solid tumor malignancy: conferences and Reviews.
lation by 1,25-dihydroxyvitamin D3 in normal human peripheral Western J Med 1991;152–163.
blood lymphocytes. J Clin Invest 1987; 79:1700. 396. Wang C, Curtis JE, Minden MD, et al. Expression of a retinoic acid
378. Tobler A, Munker R, Heitjan D, et al. In vitro interaction of recom- receptor gene in myeloid leukemia cells. Leukemia 1989; 3:264.
binant tumor necrosis factor alpha and all-trans-retinoic acid with 397. Wang C, Kelleher CA, Cheng GY, et al. Expression of the CSF-1
normal and leukemic hematopoietic cells. Blood 1987; 70:1940. gene in the blast cells of acute myeloblastic leukemia: association
379. Todd R III, Griffin J, Ritz J, et al. Expression of normal mono- with reduced growth capacity. J Cell Physiol 1988; 135:133.
cyte-macrophage differentiation antigens on HL-60 promyelo- 398. Wakamiya N, Stone R, Takeyama H, et al. Detection of tumor
cytes undergoing differentiation induced by leukocyte-conditioned necrosis factor gene expression at a cellular level in human acute
medium or phorbol ester. Leuk Res 1981; 5:491. myeloid leukemias. Leukemia 1989; 3:51.
380. Tohyama K, Ohmori S, Ueda T, et al. Cooperative effects of 399. Warrell RP Jr, Frankel SP, Miller WH Jr et al. Differentiation
gamma-interferon and 1 alpha, 25-dihydroxyvitamin D-3 on therapy of acute promyelocytic leukemia with tretinoin (all-trans
in vitro differentiation of the blast cells of RAEB and RAEB-T. retinoic acid). New Eng J Med 1991; 324:1385.
Blut 1989; 58:181. 400. Warrell RP Jr, Maslak P, Eardley A, et al. All-trans retinoic acid
381. Tomida M, Yamamoto-Yamaguchi Y, Hozumi M. Purification of for treatment of acute promyelocytic leukemia: an update of the
a factor inducing differentiation of mouse myeloid leukemic M1 New York experience. Leukemia 1994; 8:929–933.
cells from conditioned medium of mouse fibroblast L929 cells. 401. Weber G. Biochemical strategy of cancer cells and the design of
J Biol Chem 1984; 259:10978. chemotherapy: G.H.A. Clowes Memorial Lecture. Cancer Res
382. Tricot G, Jayaram HN, Zhen W, et al. Biochemically directed 1983; 43:3466–3492.
therapy with tiazofurin of refractory leukemia and myeloid blast 402. Weh HZ, Zschaber R, Hossfeld DK. Low-dose cytosine-arabinoside
crisis of chronic granulocytic leukemia. Proc Am Assoc Cancer in the treatment of acute myeloid leukemia (AML) and myelo-
Res 1991; 32:184. dysplastic syndrome (MDS). Blut 1984; 48:239.
568 Growth and differentiation factors as cancer therapeutics

403. Weis K, Rambaud S, Lavau C et al. Retinoic acid regulates aberrant Protein is Involved in Granulocytic differentitation of myeloid
nuclear localization of PML-RAR alpha in acute promyelocytic Leukemia Cells: Mol cancer Res. 2007 5(1):95–108.
leukemia cells. Cell 1994; 76:345. 419. Young CW, Fanucci MP, Declan-Walsh T, et al. Phase I trial and
404. Westin E, Wong-Staal F, Gelmann E, et al. Expressing cellular clinical pharmacological evaluation of hexamethylene bisacetamide
homologues of retroviral onc genes in human hematopoietic cells. administration by ten-day continuous intravenous infusion at
Proc Natl Acad Sci USA 1982; 74:2490. twenty-eight-day intervals. Cancer Res 1988; 48:7304.
405. Wilkins JA, Sigurdson SL, Rutherford W. The production of 420. Young D, Wagner K, Griffin J. Constitutive expression of the
immunoregulatory factors by a human macrophage-like cell line, granulocyte-macrophage colony stimulating factor gene in acute
I. Characterization of an inhibitor of lymphocyte DNA synthesis. myeloblastic leukemia. J Clin Invest 1987; 79:100.
Cell Immunol 1983; 75:328. 421. Young DC, Demetri GD, Ernst TJ, et al. In vitro expression of
406. Williams RL, Hilton DJ, Pease S, et al. Myeloid leukaemia inhib- colony-stimulating factor genes by human acute myeloblastic
itory factor maintains the developmental potential of embryonic leukemia cells. Exp Hematol 1988; 16:378.
stem cells. Nature 1988; 336:684. 422. Yourno J, Walsh J, Kornatowski G, et al. Nonspecific esterases of
407. Williams DE, Cooper S, Broxmeyer HE. Effects of hematopoi- leukemia cell lines: evidence for activation of myeloid-associated
etic suppressor molecules on the in vitro proliferation of purified zymogens in HL-60 by phorbol esters. Blood 1983; 63:238.
murine granulocyte-macrophage progenitor cells. Cancer Res 423. Zagonel V, Pinto A, Attadia V, et al. Phase I-II clinical-biological
1988; 48:1548. study of 5-aza-2′-deoxycytidine (5AZACDR) as a differentiation
408. Wing E, Koren H, Fischer D, Kelly V. Stimulation of a human inducer in acute myeloid leukemia (AML) and myelodysplastic
macrophage-like cell line (U937) to inhibit the multiplication of syndromes (MDS) of elderly. Proc Annu Meet Am Soc Clin
an intracellular pathogen. J Reticuloendothel Soc 1981; Oncol 1989; 8:A767.
29:321. 424. Zhang XG, Klein B, Bataille R. Interleukin-6 is a potent
409. Wisch JS, Griffin JD, Dufe DW. Response of preleukemic syn- myeloma-cell growth factor in patients with aggressive multiple
dromes to continuous infusion of low dose cytovabine. N Engl J myeloma. Blood 1989; 74:11.
Med 1985; 309. 425. Zinzar S, Ohnuma T, Holland JF. Effects of simultaneous and
410. Yam LT, Li CY, Crosby WH. Cytochemical identification of sequential exposure to granulocytic and monocytic inducers on
monocytes and granulocytes. Am J Clin Pathol 1971; 55:283. the choice of differentiation pathway in HL-60 promyelocytic
411. Yamamori T, Fukada K, Aebersold R, et al. The cholinergic neu- leukemia cells. Leuk Res 1989; 13:23.
ronal differentiation factor from heart cells is identical to leuke- 426. Zumkeller W, Schofield PN. Growth factors, cytokines and soluble
mia inhibitory factor. Science 1989; 246:1412. forms of receptor molecules in cancer patients. Anticancer Res
412. Yao TP, Ku, G, Zhou N et al. The nuclear hormone receptor 1995; 15:343–348.
coactivator SRC-1 is a specific target of p300. Proc Natl Acad 427. Asahi K, Honma Y, Hazeki K, Sassa T, Kubohara Y, Sakurai A,
Sci, USA 1996; 93, 10626. Takahashi N. Cotylenin A, a plant-growth regulator, induces the
413. Yee C, Biondi A, Wang XH, et al. A possible autocrine for inter- differentiation in murine and human myeloid leukemia cells.
leukin-6 in two lymphoma cell lines. Blood 1989; 74:798. Biochem Biophys Res Commun 1997; 238:758–763.
414. Yen A, Forbes M, DeGala G, et al. Control of HL-60 cell differ- 428. Yamada K, Honma Y, Asahi K, Sassa T, Hino K, Tomoyasu S.
entiation lineage specificity, a late event occurring after precom- Differentiation of human acute myeloid leukaemia cells in pri-
mitment. Cancer Res 1987; 47:129. mary culture in response to cotylenin A, a plant growth regulator.
415. Yetgin S, Ozsoylu S. Myeloid metaplasia in vitamin D deficiency Br J Haematol 2001; 114:814–821.
rickets. Scand J Haematol 1982; 28:180. 429. Hiçsönmez G. The effect of steroid on myeloid leukemic cells:
416. Ymer S, Tucker Q, Sanderson C, et al. Constitutive synthesis of the potential of short-course high-dose methylprednisolone treat-
interleukin-3 by leukemia cell line WEHI-3B is due to retroviral ment in inducing differentiation, apoptosis and in stimulating
insertion near the gene. Nature 1985; 317:255. myelopoiesis. Leuk Res 2006; 30(1):60–68.
417. Yoffe G, Spitzer G, Boggs BA. Determination of clonality in 430. Lanotte M, Martin-Thouvenin V, Najman S, Ballerini P, Valensi
acute nonlymphocytic leukemia by restriction fragment length F, Berger R. NB4, a maturation inducible cell line with t(15;17)
polymorphism and methylation analysis. Leukemia 1987; marker isolated from a human acute promyelocytic leukemia
1:226. (M3). Blood 1991; 77:1080–1086.
418. Ozpolat B, Akar U, Steiner M, Zorrila-Calancha J, Sanguino A, 431. Khanna-Gupta A, Kolibaba K, Zibello TA, Berliner, N. NB4 cells
Tirado-Gomez M, Nancy Colbum Danilenko M, and Lopez show bilineage potential and an aberrant pattern of neutrophil sec-
Berestcin G. Programmed cell Death 4 (PDCD4) Tumor suppressor ondary granule protein gene expression. Blood 1994; 84:294–302
17 Granulocyte colony-stimulating factor:
biology and clinical potential
MARYANN FOOTE and GEORGE MORSTYN

Introduction bone marrow cells, so it was concluded that this factor


was the human counterpart to mouse G-CSF. The protein
The study of hematopoiesis was greatly facilitated in the initially identified as G-CSF also was called CSF-13
mid-1960s when techniques for studying hematopoietic and pluripoietin (pCSF).
cells in clonal culture were developed. Initially, serum The study of G-CSF progressed to the purification
or conditioned medium was added to cultures as a source and molecular cloning of both murine and human forms
of growth factors, i.e., the colony-stimulating factors and then to the first clinical trials of recombinant human
(CSFs) [56, 57, 85]. One of the factors that was isolated, (rHu) G-CSF in cancer patients [7–9, 26, 27, 60–62].
purified, cloned, and produced in commercial quantities Because of its unique biologic activities, rHuG-CSF is
was granulocyte colony-stimulating factor (G-CSF), a used for reversing or ameliorating neutropenias of vari-
protein that acts on the neutrophil lineage to selectively ous causes, for allowing increased cancer chemotherapy
stimulate the proliferation and differentiation of com- dose intensity, and for mobilizing hematopoietic stem
mitted progenitor cells and activation of mature neutro- cells for transplantation.
phils (Fig. 1). A property that distinguished G-CSF from Filgrastim, the nonglycosylated rHuG-CSF, was the
other CSF and facilitated its purification, molecular first hematopoietic growth factor approved for commer-
cloning, and large-scale production in prokaryotic cells cial use. A second form of rHuG-CSF, a glycosylated
was its ability to induce terminal differentiation of a version, lenograstim, was developed and approved for
murine leukemic cell line (WFHI-3B). After observing commercial use in Europe. Nartograstim (also known as
that serum from endotoxin-treated mice was capable of marograstim, KW-2228, or NTG), a mutein rHuG-CSF,
causing the differentiation of a WFHI-3B myelomono- was developed, but it had limited clinical development
cytic leukemic cell line, Metcalf [55] named the activity [89]. A polyethylene glycol-modified rHuG-CSF (peg-
GM-DF (granulocyte-macrophage differentiating fac- filgrastim) has been approved for commercial use [28].
tor). Further analysis showed that this serum contained Another pegylated rHuG-CSF is RO 258315; it is nar-
G-CSF as well as granulocyte-macrophage colony- tograstim with polyethylene glycol molecules at the
stimulating factor (GM-CSF). Nicola et al. [66, 68] fur- amino terminus and the four lysine residues. The final
ther purified G-CSF from medium conditioned by lung product of pegylation of RO 258315 is a mixture of one,
tissue of endotoxin-treated mice. This G-CSF could two, three, or four polyethylene glycol molecules
stimulate WFHI-3B cells as well as normal cells, sup- attached to each molecule of KW-2228, with the dipe-
porting the formation of numerous small, neutrophil- gylated protein most common (60%) [94]. Table 1 sum-
containing colonies at a concentration similar to that marizes what is known about endogenous G-CSF and
needed for WFHI-3B differentiation [67]. Subsequently, Table 2 summarizes some characteristics of the rHuG-
murine G-CSF was identified as a protein and was CSFs. The molecule will be specified in this review
shown to have both differentiation-inducing activity for when the term ‘rHuG-CSF’ is insufficient. G-CSF’ will
WFHI-3B and granulocyte colony-stimulating activity be used to discuss the endogenous protein.
in bone marrow cells [68]. Other researchers, notably
Asano et al. [2] and Welte et al. [102], found several
human carcinoma cells that constitutively produce colony-
stimulating factors. One of these factors was purified to
Biochemistry and Structure
apparent homogeneity from the conditioned medium of Native human G-CSF appears to exist as a mixture of
bladder carcinoma 5637 cells [102] or a squamous car- two forms, one having 177 amino acids and the other
cinoma cell line [71]. The purified CSF selectively stim- having 174 amino acids [1, 76, 86]. The gene for human
ulated neutrophilic granulocyte colony formation from G-CSF is positioned at 17q11–22 and is approximately

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 569
© Springer Science + Business Media B.V. 2009
570 Granulocyte colony-stimulating factor: biology and clinical potential

Pluripotential Stem Cell

CFU-Blast

flk-2/flt-3 ligand flk-2/flt-3 ligand flk-2/flt-3 ligand flk-2/flt-3 ligand


SCF SCF SCF SCF

Lymphoid NK
CFU-GEMM
Stem Cell Precursor

SCF MGDF/TPO SCF SCF SCF SCF SCF SCF


IL-3 SCF IL-3 IL-3 IL-3 IL-3 IL-7 IL-7
IL-3 flk-2/flt-3 ligand flk-2/flt-3 ligand flk-2/flt-3 ligand

CFU-GM

BFU-E CFU-Meg IL-3 IL-3 CFU-Eo CFU-Ba CFU-Mast Pre-B Cell Pre-T Cell
GM-CSF G-CSF
SCF SCF GM-CSF
IL-3 IL-3
GM-CSF GM-CSF
EPO IL-11
IL-6
MGDF/TPO IL-3 IL-3 IL-3 IL-7
CFU-M CFU-G GM-CSF SCF

IL-3 IL-3
CFU-E GM-CSF GM-CSF
IL-3 M-CSF G-CSF
GM-CSF
EPO
Megakaryocyte
IL-6

Monocyte SCF
IL-2
GM-CSF IL-7 IL-2
Reticulocyte M-CSF B Lymphocyte

Proplatelet

Red Blood Cell Platelet Macrophage Neutrophil Eosinophil Basophil Tissue Mast Cell Plasma Cell T Lymphocyte NK Cell

Figure 1. Hematopoietic tree (Figure courtesy of Amgen Inc., Thousand Oaks, CA)

Table 1. Summary of characteristics of human G-CSF


Characteristic
Location of gene Chromosome 17q11-22 (human)
Gene size Approximately 2500 nucleotides (approximately 2.5 kb)
Number of exons 5
Protein produced Proform
Number of amino acids in mature protein 174 (18.6 kDa)
Glycosylation site Thr 133
Disulfide bonds 2 (between 36 and 42, and 64 and 74)
Basic structure 4 helices arranged in antiparallel fashion; and sheet pleating
Receptor structure Single 150-kDa chain-forming homodimer
M. Foote and G. Morstyn 571

Table 2. Characteristics of the forms of recombinant human G-CSF


Generic name No. of amino acids Cell source Other information Marketing company
Filgrastim 175 E. coli Non-glycosylated Amgen, Kirin, Roche
Lenograstim 174 Chinese hamster ovary Glycosylated Chugai, Rhône-Poulenc
Nartograstim 174a E. coli Mutein form Kyowa-Hakko
Pegfilgrastim 175 E. coli Non-glycosylated, pegylated Amgen
RO 25–8315 174 E. coli Mutein form, pegylated Roche

a
Changes to amino acids 1, 3, 4, 5, and 17

2.5 kbp [84]. It is encoded by the CSF3 gene. One chro- of filgrastim results in the cleavage of the molecule near
mosomal gene exists per haploid genome. threonine 134 points to a role of glycosylation for prote-
The core protein of native human G-CSF has a molec- olytic protection and indicates that the residues along
ular weight of 18.8 kDa and the glycosylated protein this portion of the protein structure may serve as ‘handles’
20–23.5 kDa [76]. Filgrastim is a highly purified protein for proteolytic degradation [76]. Although it has been
consisting of a single 175-amino acid polypeptide; suggested that glycosylation of lenograstim increases
lenograsfim, lacking an N-terminal methiorryl residue, biologic activity in vitro compared with filgrastim [103],
has 174 amino acids. Lenograstim exists as two formu- this advantage has not been verified in randomized double-
lations, one using gelatin and one using human serum blind comparative studies [35]. Glycosylation possibly
albumin as a stabilizer. The gelatin lenograstim formu- influences the antigenicity of recombinant proteins [48].
lation reportedly allows faster neutrophil recovery after Several cell types in the human body produce G-CSF,
chemotherapy than lenograstim [90]. Although nar- including stromal cells, endothelial cells, fibroblasts,
tograstim also has 174 amino acids, the molecule has macrophages, and monocytes [19, 24, 61, 76, 79, 96,
been modified at the first, third, fourth, fifth, and 17th 105]. These cell types are widely distributed in the body,
amino acids [70]. Nartograstim is conjugated with an suggesting that G-CSF participates in the production
average of two polyethylene glycol chains per protein and functional enhancement of neutrophils that occur in
molecule [23]. Pegfilgrastim has a 20-kDa polyethylene response to local infection or other causes.
glycol molecule added to filgrastim, giving the protein In a variety of pathologic conditions, including expo-
an extended half-life in the serum [58]. sure to endotoxin, response to an infection or to neutro-
Native human G-CSF and lenograstim are O-glycosylated penia, the amount of circulating endogenous G-CSF in
[64, 86] whereas filgrastim and pegfilgrastim are not glyco- the blood increases [10, 40, 56, 98]. In humans, G-CSF
sylated. The sugar chains are responsible for the differences is the primary factor for the upregulation of neutrophils
in molecular mass. The presence and type of glycosylation of in infection and in various pathologic conditions with
the recombinant protein depends on the cellular source (i.e., decreased neutrophil counts [10]. It has been suggested
yeast, mammalian cell, or bacterial cell) [62]. Lenograstim that the Toll-like receptors on macrophages and den-
has heterogenous glycosylation: not all the sugar chains are dritic cells detect gram-negative bacteria and induce
identical. Not all structural components of native human IL-23 production that, in turn, induces IL-17 production
G-CSF have been identified, but it is known that its sugar by T cells, which then stimulates G-CSF production.
composition is not identical to that of lenograstim [72, 73]. Mechanistically, as G-CSF concentrations increase during
Native human G-CSF does not bind to concanavalin, sug- bacterial infections and circulate in the blood, neutro-
gesting that it does not have mannose-containing carbohy- poiesis is stimulated in the marrow. Hence, in cases of
drates [69]. When G-CSF was treated with neuraminidase, it gram-negative and fungal infections, serum G-CSF con-
showed reduced charge heterogeneity in isoelectric focusing, centrations are increased [40]. Indeed, the highest
suggesting sialic acid-containing O-glycosylation [68]. G-CSF concentrations have been found in patients with
Crystallography studies of filgrastim have shown that neutropenia and febrile neutropenia [10].
the glycosylation site (threonine 134) is attached to the G-CSF selectively stimulates the proliferation and
C-D loop at a distance from the active biologic sites [43, differentiation of neutrophil precursors by binding to a
74]. Although the glycosylation does not seem to have a specific cell-surface receptor [18]. The G-CSF receptor
role in the biologic function of the molecule, it may par- is expressed on cells of the neutrophil lineage from
tially protect the molecule from proteolytic degradation. myeloblast to the mature neutrophil and on a subset of
The observation that a limited proteolytic degradation cells of the monocyte lineage [56]. It has been reported
572 Granulocyte colony-stimulating factor: biology and clinical potential

that G-CSF receptor expression on cell types is not usu- Lenograstim also increases superoxide anion produc-
ally associated with its activity including endothelial tion in neutrophils in response to fMLP [93]. Neutrophils
cells, activated T lymphocytes, and nonhemopoietic from healthy volunteers treated with lenograstim showed
tumor cell lines. The role of these receptors is unclear. evidence of enhanced stimulation by agonists, including
Both filgrastim and lenograstim, as well as native HuG- adhesion to nylon fibers and physiologic substrates
CSF derived from a tumor cell line, have identical dose- [33, 36].
dependent affinity for G-CSF receptors (Fig. 2). The In vivo studies in animals and some studies in humans
binding of G-CSF to its receptor decreases the number have shown that rHuG-CSF causes a dose-dependent
of available surface receptors as the surface receptor increase in the number of neutrophils in the peripheral
complex is internalized and degraded. blood. This increase is thought to be a result of decreased
Both endogenous and recombinant G-CSFs work on maturation time, increased number of cell divisions, and
neutrophil precursor cells and mature neutrophils. accelerated release into the peripheral blood. The action
rHuG-CSF stimulates the proliferation, differentiation, of rHuG-CSF causes rapid dose-dependent increases in
and activation of committed progenitor cells of the neu- neutrophils and small or no effects on monocytes and
trophil lineage and reduces neutrophil maturation time eosinophils, respectively [50, 52, 63].
from 5 days to 1 day, leading to a rapid release of mature
neutrophils from the bone marrow into the circulation
[53, 54]. Neutrophils previously treated with rHuG-CSF
have normal intravascular half-life [8, 54]. In the pres-
Physiology
ence of rHuG-CSF, neutrophils have enhanced superox- G-CSF is an indispensable cytokine for normal murine
ide production in response to chemoattractants [1, 100]. myelopoiesis, as has been shown by knock-out-mouse
G-CSF also enhances chemotaxis by increasing the experiments [47]. Circulating neutrophil concentrations
binding of fMLP (formyl-methionyl-leucyl-phenylala- were reduced by 70–80% with less of a reduction in mar-
nine) [14] as well as enhancing anti-Candida activity. row stores of progenitors (50%) compared with those of
rHuG-CSF does not stimulate release of other cytokines normal mice. Despite appearing superficially healthy, these
by neutrophils and mononuclear cells [78]. mice have a diminished ability to mount neutrophilia

Figure 2. Inhibition of labeled filgrastim, lenograstim, and tumor cell-derived human G-CSF binding to neutrophils. Those data
demonstrate that the three forms have the same affinity for G-CSF receptors in vitro. (Figure courtesy of Amgen Inc., Thousand
Oaks, CA)
M. Foote and G. Morstyn 573

and monocytosis in response to infection and have a administration, and no dose sequence effects were observed
marked impairment in ability to control Listeria infec- in a crossover study [38].
tions. The observations from this study indicate that Administration of single 10, 30, 60,100, or 150 μg/kg
G-CSF is required for maintaining the normal quantita- doses of RO 258315 to healthy men produced dose-
tive balance of neutrophil production during steady-state dependent increases in peak neutrophil count [94]. The
granulopoiesis in vivo and implicate rHuG-CSF in emer- time to reach peak concentration and the area under the
gency granulopoiesis during infectious episodes. serum concentration time curve increased 100-fold over
the range of doses studied.
A healthy volunteer study was reported with pegfil-
grastim [58]. Pegfilgrastim was administered as a single
Pharmacokinetics injection of 30, 60, 100, or 300 μg/kg, and blood samples
Pharmacokinetic/pharmacodynamic data from the differ- were taken at specific time points for 48 h. Neutrophil
ent rHuG-CSF products are difficult to compare directly, counts increased in a dose-dependent manner, and the peak
as different study designs, doses, regimens, routes of neutrophil count attained and the length of the response
administration, and populations were evaluated. Table 3 were dose dependent.
summarizes these data.
Patients with Disease
Healthy Volunteer Studies Patients with cancer receiving filgrastim 11.5 μg/kg as a
Table 3 summarizes pharmacokinetic and pharmacody- 30-min IV infusion had a peak serum concentration of
namic data from healthy volunteer studies. Healthy 384 ng/ml [26, 27]. When patients with cancer received
men given single doses of 3.45 μg/kg filgrastim by SC bolus or SC infusion doses, serum concentrations of
30-min intravenous (IV) infusion had a mean serum filgrastim reflected rapid absorption [60]. The serum half-
concentration of 20.8 ng/ml 5 min after the end of infu- life of filgrastim has a t1/2 of 3.5 h [48]. With filgrastim,
sion [3]. The mean (SD) elimination half-life was 163 the maximum increase in neutrophil count can be achieved
(±7.4) min, and G-CSF concentrations returned to nor- by all routes of administration tested [60, 80].
mal values within 14–18 h. Patients with nonmyelogenous malignancies receiving
After single subcutaneous (SC) injections of lenograstim at 5, 10, or 20 μg/kg/day as a SC injection
lenograstim 10, 20, or 40 μg in healthy volunteers, Cmax had peak plasma concentrations of 10.1, 35.0, and
values were 0.09, 0.18, and 0.48 μg/L, respectively, 49.7 mg/l, respectively [25]. Cmax was reached within
within 3.5–4.5 h, peak serum concentrations were main- 4–8 h. The serum half-life of lenograstim was 3.0, 4.8,
tained for almost 4 h, and lenograstim was almost elimi- and 6.0 h, respectively, for the 5-, 10-, and 20-μg/kg per
nated from the serum by 48 h [81]. Lenograstim exhibits day cohorts.
dose-dependent pharmacokinetic characteristics with Results of a randomized, dose-escalation study with
peak serum concentrations after repeated doses (SC or pegfilgrastim in patients with nonsmall cell lung cancer
IV) that are proportional to the administered dose [13]. receiving chemotherapy were published [39]. Thirteen
Lenograstim does not appear to accumulate after repeated patients were randomly assigned to receive daily injections

Table 3. Summary of pharmacokinetic/pharmacodynamic data in normal volunteers. Pharmacokinetic


data are difficult to compare directly because of differences in study designs, doses, and populations
Generic Name Cmax tmax (h) AUC
Filgrastim (75 μg/kg dose)
a
1.65±0.80 ng/ml 5.5±1.8 14.3±4.3 (0–24 h, ng/L×h)
Lenograstimb (5 μg/kg dose) 10.3–11.9 mg/L 6.0 89.8–102.3 (0–24 h, mg/L×h)
Nartograstimc (2 μg/kg dose) 1.52±0.58* 4.0±2.0 12.7±4.78 ng×hr/ml
Pegfilgrastima (30 μg/kg dose) 43.6±20.0 ng/ml 9.50±3.51 887±336 (0–infinity, ng/L×h)
Ro 25–8315d 2.5±1.6 ng/ml 13±5.9 184±77 ng × h/ml

Cmax, peak serum concentration; tmax, time to Cmax; AUC, area under the serum drug concentration curve versus
time curve
*
Units not provided
a
Amgen data on file; bDunn and Goa [24]; cSuzuki et al. [92]; dVan der Auwera et al. [97]
574 Granulocyte colony-stimulating factor: biology and clinical potential

of filgrastim 5 μg/kg per day or a single injection of peg-


filgrastim at 30, 100, or 300 μg, 2 weeks before chemo-
Clinical Implications
therapy and 24 h after completion of chemotherapy. Much clinical data have been published about the various
Peak serum concentrations of filgrastim and the duration forms of rHuG-CSF and their use in numerous thera-
of increased serum concentrations were dose dependent. peutic areas. To cite all the important references in this
The concentration of serum filgrastim remained high for review paper is impossible, and we have concentrated
a longer duration in the setting of chemotherapy-induced on the pivotal phase 3 trials used in obtaining marketing
neutropenia until neutrophil recovery occurred. Patients authorization whenever possible. The interested reader
treated with pegfilgrastim had a higher prechemother- is directed to the cited reviews [21, 49, 59, 88, 101].
apy neutrophil count than patients treated with filgrastim
(who did not receive the study drug until after chemo-
therapy was completed). After chemotherapy, the neu-
Chemotherapy-induced Neutropenia
trophil count nadirs were similar among patients Neutropenia and infection are common dose-limiting
receiving filgrastim 5 μg/kg per day or pegfilgrastim effects of cancer chemotherapy. It has long been known
30 μg; higher neutrophil count nadirs were seen in that the risk of infection is directly related to the depth
patients receiving the higher doses of pegfilgrastim. and duration of neutropenia [6]. The severity of chemo-
therapy-induced neutropenia depends on the dose inten-
sity of the therapy and other factors. Often, oncologists
withhold or delay the next cycle of chemotherapy if a
Pharmacodynamics patient has low neutrophil counts. Such practice,
The exposure of filgrastim may be influenced by the although done to prevent infectious complications, can
increased neutrophils formed after administration of compromise otherwise effective cancer chemotherapy.
the cytokine [80]. Increased numbers of neutrophils Two randomized, placebo-controlled, double-blind
were shown to be associated with increased clearance studies involving more than 300 patients with small-cell
of filgrastim in patients with cancer, which suggests lung cancer receiving cyclophosphamide, doxorubicin,
that a negative feedback mechanism is involved in and etoposide (CAE) chemotherapy showed that fil-
maintaining neutrophil counts at optimal values [45]. grastim decreased the incidence, severity, and duration
The bioavailability of filgrastim was an average of 53% of severe neutropenia [15, 92]. These two studies
when administered at very low doses (1 μg/kg) to showed that, in the placebo control group, most of the
healthy volunteers [91]; when administered at thera- febrile neutropenic events occurred in the first cycle.
peutic doses, its bioavailability has been reported as Two randomized placebo-controlled phase 3 trials with
high as 80% [80]. lenograstim showed significant reductions in the median
Lenograstim has an absolute bioavailability of 30% duration of neutropenia in patients with inflammatory
after SC doses of 2–5 μg/kg, with an apparent volume of breast cancer [12] or non-Hodgkin’s lymphoma [29]. The
distribution of 1 L/kg [13]. The serum elimination half- median neutrophil nadir was higher in lenograstim-treated
life after SC administration is approximately 3 h, but it patients who had inflammatory breast cancer than in
is shorter after repeated IV infusion (1–1.5 h) [38]. patients administered placebo. Treatment with lenograstim
The absolute bioavailability of pegfilgrastim after SC was associated with reductions in culture-confirmed
administration is estimated to be approximately 15% [31]. infections during periods of neutropenia, shorter durations
The exposure of pegfilgrastim is affected by the neutrophil of hospitalization for infections, and reduced need for
status; pegfilgrastim concentrations for patients who expe- antibacterial drugs.
rienced severe neutropenia were sustained longer and In two double-blind randomized studies, use of peg-
higher than those who did not experience severe neutrope- filgrastim reduced neutropenia and its complications in
nia. Based on pharmacokinetic/pharmacodynamic model- patients receiving chemotherapy [31, 37]. In one study,
ing, clearance of pegfilgrastim is almost completely patients with high-risk stage II or stage III/IV breast
(>99%) regulated by neutrophil-mediated clearance. cancer received a single 6-mg injection of pegfilgrastim
While renal clearance plays a significant role in the elimi- or daily 5 μg/kg SC injections of filgrastim for up to four
nation of filgrastim, it is absent for pegfilgrastim, based on cycles of chemotherapy [31]. The single injection of
results from a bilateral nephrectomy rat study [58].The pegfilgrastim was as effective as a mean of 11 daily
predominant dependency of pegfilgrastim clearance on injections of filgrastim with respect to the observed mean
the neutrophil-mediated pathway allows pegfilgrastim to duration of grade 4 neutropenia. The incidence of febrile
be under highly efficient ‘self-regulation’. neutropenia was lower, but not statistically significantly,
M. Foote and G. Morstyn 575

in the pegfilgrastim group compared with the filgrastim neutropenia (neutrophil count <0.5 × 109/L) was 23 days
group. In the second study, patients received either for the control group, 11 days for the 10-μg/kg per day
100 μg/kg pegfilgrastim once per cycle of chemotherapy group, and 14 days for the 30-μg/kg per day group.
or daily injections of 5 μg/kg filgrastim [37]. Again, For the control group, compared with both treatment
single doses of pegfilgrastim were comparable to multiple groups combined, the difference in the number of days
doses of filgrastim in terms of duration of grade 4 neu- of neutropenia was statistically significant. Moreover,
tropenia and depth of neutrophil nadir counts in all filgrastim reduced the duration of fever and febrile
cycles. In both studies, pegfilgrastim and filgrastim were neutropenia.
well tolerated. A phase 3 trial of lenograstim was performed in 298
Since these pivotal randomized clinical trials were patients receiving either autologous or allogeneic bone
completed, additional studies were conducted. This has marrow transplantation [30]. The addition of lenograstim
led to major organizations such as ASCO, EORTC, 5 μg/kg per day produced 30% reduction relative to
NCCN, and ESMO evaluating these data and producing placebo in the median time to reach neutrophil counts of
guidelines. These data have been reviewed by an expert 0.5 × 109/L. Other significant reductions compared with
panel in Europe [32]. The ASCO evaluation was pub- placebo were seen in lenograstim-treated patients for
lished in 2006. There is agreement in all studies that median time in hospital, duration of antibiotics, and dura-
rhuG-CSF reduces the rate of febrile neutropenia by tion of parenteral nutrition. The incidence of infections,
50% what ever the background febrile neutropenia rate. febrile neutropenia, and septicemia were similar between
Much discussion, however, ensued about the high back- the two groups, but the median duration of infection and
ground febrile neutropenia rate in the initial registration febrile neutropenia were reduced with lenograstim.
trials and whether the use of rHuG-CSF was appropriate In another study with lenograstim [51], reduction of
in settings with a lower febrile neutropenia rate. One neutropenia was associated with a reduction in the number
issue in the early studies was the frequent measures of of days of fever and a shorter duration of parenteral anti-
neutrophil counts and temperature led to the identifica- biotic use after transplantation.
tion of neutropenia that might have been missed if blood Recombinant HuG-CSF alone or in combination with
counts were performed less frequently. Nevertheless, chemotherapy is an effective agent for recruiting periph-
recent studies were performed in settings in which the eral blood progenitor cells (PBPC) with long-term
febrile neutropenia rate was approximately 20% and so reconstituting ability [11, 22, 41, 82, 83]. In a histori-
current guidelines recommend the use of CSFs in set- cally controlled study, filgrastim-generated PBPC, in
tings in which the risk of febrile neutropenia is greater conjunction with autologous bone marrow transplanta-
than 20%. The existence of risk factors such as age, tion and daily filgrastim, accelerated recovery of neutro-
treatment intent such as dose intensity, and prior toxicity phil and platelet count [82]. In a study of 85 patients
from chemotherapy may increase risk. with relapsed Hodgkin’s disease [11], use of filgrastim
for mobilization resulted in an accelerated time to recov-
Bone Marrow or Stem Cell ery of granulocytes compared with nonmobilized PBPC
recipients. The use of mobilized PBPC resulted in an
Transplantation accelerated time to platelet engraftment compared with
Bone marrow or stem cell transplantation allows the use nonmobilized PBPC recipients. Costs were lower in
of very high doses of chemotherapy, with or without patients who received filgrastim-mobilized PBPC com-
radiotherapy, to eliminate cancer cells from patients with pared with patients who did not.
refractory tumors. After ablative therapy and before the Use of lenograstim for PBPC mobilization has pri-
patient’s bone marrow recovers full function, the patient marily been reported in abstract form. Patients with
often experiences profound pancytopenia that requires lymphoma previously treated with lenograstim had a
multiple transfusions of blood and blood products. median time to neutrophil recovery of 11–12 days after
In the pivotal phase 3 trial for filgrastim in the high- chemotherapy and infusion of lenograstim-assisted
dose chemotherapy and autologous bone marrow trans- autologous PBPC transplantation [99].
plantation setting, patients were administered filgrastim A comparison of lenograstim and filgrastim administered
10 or 30 μg by continuous IV infusion or placebo for 5 after chemotherapy to increase PBPC yield was reported
days, starting 1 day after bone marrow infusion [87]. as a 126-patient retrospective study [46]. No differences
The dose of filgrastim was adjusted as needed during were observed between groups for median CD34 cells
neutrophil recovery for a scheduled maximum of 28 harvested or number of leukophereses needed to obtain
days of administration. The median number of days of the 3 × 106 CD34 cells/kg required for transplantation.
576 Granulocyte colony-stimulating factor: biology and clinical potential

Severe Chronic Neutropenia infection, for reversal of clinically significant neutropenia,


and for maintenance of adequate neutrophil counts during
A phase 3 trial of filgrastim in patients with severe chronic treatment with antiretroviral therapy. A phase 3 study
neutropenia has shown long-term efficacy in boosting reported the effect of filgrastim on the incidence of severe
neutrophil counts, with hematologic and clinical benefits neutropenia in patients with advanced HIV infection and
sustained during prolonged maintenance treatment [16, its effect on the prevention of infectious morbidity [44].
17]. Of the 120 patients who received filgrastim, 108 had In this 24-week study of 258 patients, filgrastim was
a median neutrophil count of >1.5 × 109/L during the administered daily at 1 μg/kg and adjusted to as much as
study. Their bone marrow had increased proportions of 10 μg/kg, or was administered intermittently at 300 μg
maturing neutrophils. The incidence and duration of infec- daily 1–3 days per week. Patients in a control group
tion-related events was reduced by approximately 50% received filgrastim only if they developed severe neutro-
and the duration of antibiotic use was reduced by 70%. penia. Both daily and intermittent administration of
No phase 3 studies of lenograstim in patients with filgrastim lowered the incidence of bacterial infection
severe chronic neutropenia have been reported, but sev- rates compared with patients in the control group. The
eral small groups of patients reported have been treated filgrastim-treated patients had 31% fewer bacterial infec-
with lenograstim [21]. One European phase 2 study tions than did the control patients, suggesting that the use
reported that all 19 children treated with lenograstim had of filgrastim can reduce the risk of several bacterial infec-
neutrophil recovery to >1.0 × 109/L [20]. Normal bone tions and subsequent number of days of hospitalization.
marrow cytology was attained by ten of the 19 children. No phase 3 studies with lenograstim in the setting of
AIDS/HIV infection appear to have been published as
Bone Marrow Failure States full reports. One study using lenograstim in the setting of
AIDS showed that lenograstim could safely ameliorate
Both filgrastim and lenograstim are licensed in Japan the neutropenia associated with zidovudine treatment in
and China for the treatment of aplastic anemia [77]. patients with AIDS or AIDS-related complex [95].
Studies suggest that rHuG-CSF increases neutrophil
counts in some patients with moderate aplastic anemia;
however, patients with very severe hypoplasia generally Current Issues
do not respond to growth factors [5, 42]. The European Although both filgrastim and lenograstim have been
Group for Bone Marrow Transplantation (EBMT) marketed for more than a decade and pegfilgrastim was
Working Party discourages the use of rHuG-CSF as a approved in 2002, and are licensed for many uses in
single agent in patients with newly diagnosed aplastic countries around the world, work continues to fully
anemia [4]. Although published data do not fully sup- understand the mechanisms of action and long-term
port the use of growth factor as first-line treatment for results of chronic administration and to discover other
aplastic anemia, rHuG-CSF in combination with other conditions and disease states where rHuG-CSF may be
approaches for this disorder (e.g., with immunosuppres- useful. One issue that may not be fully resolved in the
sive therapy [76]) may have a role. perceived relationship between use of hematopoietic
The myelodysplastic syndromes are a group of neoplas- growth factors in several patient populations and the
tic hematopoietic disorders. In a phase 3 randomized study risk of developing leukemia [34]. It is not clear if the
involving 102 patients with RAEB or RAEB-t subtypes slightly increased risk of developing leukemia is due to
of myelodysplastic syndromes, filgrastim was efficacious the underlying disease concurrent with chemotherapy or
in increasing neutrophil counts [65], although imbalances if the use of a growth factor increases the risk.
in patient characteristics made the overall benefit, if any, Surveillance is on going and the benefits appear to out-
of the use of rHuG-CSF in this setting difficult to define. weigh any potential risks.
In a phase 2 study to demonstrate the efficacy of
lenograstim, most patients responded to IV doses of 2 or
5 μg/kg per day [104]. Administration of lenograstim Immunomodulation
increased neutrophil counts in 18 Japanese patients and Immune reconstitution depends upon hematopoietic
no patient progressed to acute myeloid leukemia. reconstitution as the first step, and hematopoietic recon-
stitution, of course, depends on the fine actions and inter-
actions of hematopoietic growth factors. Hematopoietic
AIDS/HIV Infection
growth factors, including G-CSF, may be important
Filgrastim has been approved in Australia, Canada, the modulators of immune reconstitution because of their
European Union, and Japan for use in patients with HIV ability to increase the number of leukocytes, recruit
M. Foote and G. Morstyn 577

immature leukocytes, cause maturation and augmenta- 6. Bodey GP, Buckley M, Sathe YS, Freierich EI. Quantitative rela-
tion of leukocytes, and suppress leukocytes [97]. tionships between circulating leukocytes and infection in patients
with acute leukemia. Ann Intern Med 1966;64:328–340.
7. Bronchud MH, Howell A, Crother D, et al. Phase I/II study of
Use in Patients with Sickle-cell Anemia recombinant human granulocyte colony-stimulating factor to
increase the intensity of treatment with doxorubicin in patients with
Increases in leukocyte counts may be associated with advanced breast and ovarian cancer. Br J Cancer 1989;60:121–128.
8. Bronchud MH, Potter MR, Morgenstern C, et al. In vitro and in
worsened prognosis in patients with sickle-cell anemia.
vivo analysis of the effects of recombinant human granulocyte
A study of 3,764 patients with sickle-cell disease, rang- colony-stimulating factor in patients. Br J Cancer 1988;58:64–69.
ing in age from birth to 66 years of age, was done to 9. Bronchud MH, Scarffe JH, Thatcher N, et al. Phase I/II study of
determine the life expectancy, median age at death, and recombinant human granulocyte colony-stimulating factor in
circumstances of death [75]. Patients with sickle-cell patients receiving intensive chemotherapy for small cell lung can-
cer. Br J Cancer 1987;56:809–813.
anemia who had hemoglobin values <7.1 g/dL (below
10. Cebon JS, Layton JE, Maher D, Morstyn G. Endogenous hae-
the tenth percentile) had a slightly higher risk of death mopoietic growth factors in neutropenia and infection. Br J
than those patients with increased white blood cell Haematol 1994;86:263–274.
counts (>1.5 × 109/L). 11. Chao NJ, Schriber JR, Grimes K, et al. Granulocyte colony-stimu-
lating factor ‘mobilized’ peripheral blood progenitor cells acceler-
ate granulocyte and platelet recovery after high-dose chemotherapy.
Blood 1993;81:2031–2035.
Conclusions 12. Chevallier B, Chollet P, Merrouche Y, et al. Lenograstim prevents
morbidity from intensive induction chemotherapy in the treatment
The discovery, isolation, and cloning of G-CSF and the of inflammatory breast cancer. J Clin Oncol 1995;13:1564–1571.
13. Chugai Pharma. UK. ABPI Compendium of Data Sheets and
subsequent production of rHuG-CSF have had profound Summaries of Product Characteristics. London: Datapharm
effects on the practice of medicine, particularly oncol- Publications, 1999.
ogy. The use of rHuG-CSF has allowed increased 14. Colgan SP, Gasper PW, Thrall MA, Boone TC, Blancquaert AMB,
administration of cytodoxic drugs, thereby allowing Bruyninckx WJ. Neutrophil function in normal and Chediak-
Higashi syndrome cats following administration of recombinant
increased efficacy of chemotherapy and eliminating the
canine granulocyte colony-stimulating factor. Exp Hamatol
dose-limiting toxicity of myelosuppression; however, 1992;20:1229–1234.
the treatment of many diseases, including nonmalignant 15. Crawford J, Ozer H, Stoller R, et al. Reduction by granulocyte
disease and non-neutropenic infections, also has been colony-stimulating factor of fever and neutropenia induced by
altered by the use of rHuG-CSF. Although rHuG-CSFs chemotherapy in patients with small-cell lung cancer. N Eng
J Med 1991;325:164–170.
have been commercially available for more than 2
16. Dale DC, Fier C, Welte K. Use of filgrastim (r-metHuG-CSF) in
decades, research continues to find new formulations severe chronic neutropenia. In: Morstyn G, Dexter TM, Foote MA,
that improve patient compliance or that have enhanced eds. Filgrastim (r-metHuG-CSF) in Clinical Practice. New York:
biologic activity. Marcel Dekker, 1998:121–147.
17. Dale DC, Bonilla MA, Davis MS, et al. A randomized controlled
phase III trial of recombinant human granulocyte colony-stimulat-
ing factor (Filgrastim) for treatment of severe chronic neutropenia.
References Blood 1993;81:2496–2502.
18. Demetri GD, Griffin JD. Granulocyte colony-stimulating factor
1. Asano S. Human granulocyte colony-stimulating factor: its basic and its receptor. Blood 1991;78:2791–2808.
aspects and clinical applications. Am J Ped Hematol/Oncol 19. Devlin IT, Devlin PE, Myamabo K, et al. Expression of granulo-
1991;13:400–413. cyte colony-stimulating factor by human cell lines. J Leukocyte
2. Asano S, Sato N, Mori M, et al. Detection and assessment of Biol 1987;41:302–306.
human granulocyte-macrophage colony-stimulating factor 20. Donadieu J, Boutard P, Bernatowski E, et al. A European phase II
(GM-CSF) producing tumours by heterotransplantation into nude study of recombinant human granulocyte colony-stimulating fac-
mice. Br J Cancer 1980;41:689–694. tor (lenograstim) in the treatment of severe chronic neutropenia in
3. Azuma J, Kurimoto T, Awata S, et al. Phase I study of KRN8601 children. Eur J Pediatr 1997;156:693–700.
(rhG-CSF) in normal healthy volunteers: safety and pharmacokinet- 21. Dunn CJ, Goa KL. Lenograstim. An update of its pharmacological
ics in single subcutaneous administration. Rinsho Iyaka 1989;5: properties and use in chemotherapy-induced neutropenia and
2231–2252. related clinical settings. Drugs 2000;59:681–717.
4. Bacigalupo A, Hows J, Gluckman E, et al. Bone marrow transplan- 22. Dürhsen U, Villeval JL, Boyd J, Kannourakis G, Morstyn G,
tation (BMT) versus immunosuppression for treatment of severe Metcalf D. Effects of recombinant human granulocyte colony-
aplastic anemia (SAA): a report of the EBMT SAA Working Party. stimulating factor on hematopoietic progenitor cells in cancer
Br J Haematol 1988;70:177–182. patients. Blood 1988;72:2074–2081.
5. Bessho M, Toyoda A, Itoh N, et al. Trilineage recovery by combi- 23. Eliason JF, Greway A, Tare N, et al. Extended activity in cynomolgus
nation therapy with recombinant human granulocyte colony- monkeys of a granulocyte colony-stimulating factor mutain conju-
stimulating factor (rhGCSF) and erythropoietin (rhEPO) in gated with high molecular weight polyethylene glycol. Stem Cells
severe aplastic anemia. Br J Haematol 1992;80:409–411. 2000;18:40–45.
578 Granulocyte colony-stimulating factor: biology and clinical potential

24. Ernst TJ, Ritchie AR, Demetri GD, et al. Regulation of granulocyte blood stem cell auto-grafting in children. Exp Hamatol 1993;21:
colony-stimulating factor mRNA levels in human blood monocytes 103–108.
is mediated primarily at the posttranscriptional level. J Biol Chem 42. Kojima S, Fukuda M, Miyajima Y, Matsuyama T, Horibe K.
1989;264:5700–5703. Treatment of aplastic anemia in children with recombinant human
25. Frampton JE, Lee CR, Faulds D. Filgrastim. A review of its phar- granulocyte colony-stimulating factor. Blood 1991;77:937–941.
macological properties and therapeutic efficacy in neutropenia. 43. Kuga T, Komatsu Y, Yamaski M, et al. Mutagenesis of human
Drug Eval 1995;48:731–760. granulocyte colony stimulating factor. Biochem Biophys Res
26. Gabrilove JL, Jakabowski A, Fain K, et al. Phase I study of granu- Commun 1989;159:103–111.
locyte colony-stimulating factor in patients with transitional cell 44. Kuritzkes DR, Parenti D, Ward D, et al. Filgrastim prevents severe
carcinoma of the urothelium. J Clin Invest 1988;82:1454–1461. neutropenia and reduces infective morbidity in patients with
27. Gabrilove JL, Jakubowski A, Scher H, et al. Effect of granulocyte advanced HIV infection: results of a randomized, multicenter, con-
colony-stimulating factor on neutropenia and associated morbidity trolled trial. AIDS 1998;12:65–74.
due to chemotherapy for transitional cell carcinoma of the urothe- 45. Layton JE, Hockman H, Sheridan WP, Morstyn G. Evidence for a
lium. N Engl J Med 1988;318:1414–1422. novel in vivo control mechanism of granulopoiesis: mature cell-
28. George S, Yunus F, Case D, et al. Fixed-dose pegfilgrastim is safe related control of regulatory growth factor. Blood 1989;74:
and allows neutrophil recovery in patients with nonHodgkin’s 1303–1307.
lymphoma. Leuk Lymphoma 2003;44:1691–1696. 46. Lefrere F, Bernard M, Audat F, et al. Comparison of lenograstim vs
29. Gisselbrecht C, Human B, Lepage E, et al. Placebo-controlled filgrastim administration following chemotherapy for peripheral
phase III study of lenograstim (glycosylated recombinant human blood stem cell (PBSC) collection: a retrospective study of 126
granulocyte colony-stimulating factor) in aggressive non-Hodg- patients. Leuk Lymphoma 1999;35:501–505.
kin’s lymphoma: factors influencing chemotherapy administration. 47. Lieschke GJ, Grail D, Hodgson G, et al. Mice lacking granulocyte
Leuk Lymphoma 1997;25:289–300. colony-stimulating factor have chronic neutropenia, granulocyte
30. Gisselbrecht C, Prentice HG, Bacigalupo A, et al. Placebo- and macrophage progenitor cell deficiency, and impaired neutro-
controlled phase III trial of lenograstim in bone-marrow transplan- phil mobilization. Blood 1994;84:1737–1746.
tation. Lancet 1994;343:696–700. 48. Lieschke GJ, Burgess AW. Granulocyte colony-stimulating factor
31. Green M, Koelbl H, Baselga J, et al. A randomized, double-blind and granulocyte-macrophage colony-stimulating factor (1). N
multicenter phase III study of fixed-dose single administration Engl J Med 1992;327:28–35.
pegfilgrastim versus daily filgrastim in patients receiving myelo- 49. Lieschke GJ, Burgess AW. Granulocyte colony-stimulating factor
suppressive chemotherapy. Ann Oncol 2003;14:29–35. and granulocyte-macrophage colony-stimulating factor (2). N
32. Gridelli C, Aapro MS, Barni S, et al. Role of colony stimulating Engl J Med 1992;327:99–106.
factors (CSFs) in solid tumours: results of an expert panel. Crit 50. Lieschke GJ, Cebon J, Morstyn G. Characterization of the clinical
Rev Oncol Hematol 2007;63:53–64. effects after the first dose of bacterially synthesized recombinant
33. Hakansson L, Hoglund M, Jonsson UB, et al. Effects of in vivo human granulocyte-macrophage colony-stimulating factor. Blood
administration of G-CSF on neutrophil and eosinophil adhesion. 1989;74A:2634–2643.
Br J Haematol 1997;98:603–611. 51. Linch DC, Scarrffe H, Proctor S, et al. Randomised vehicle-con-
34. Herschman D, Nugent AI, Jacobson JS, et al. Acute myeloid leu- trolled dose finding study of glycosylated recombinant human
kemia or myelodysplastic syndrome following use of granulocyte granulocyte colony-stimulating factor after bone marrow trans-
colony-stimulating factors during breast cancer adjuvant chemo- plantation. Bone Marrow Transplant 1993;11:307–311.
therapy. J Natl Cancer Inst 2007;99:196–205. 52. Lindemann A, Herrmann F, Oster W, et al. Hematologic effects of
35. Hoglund M. Glycosylated and non-glycosylated recombinant recombinant human granulocyte colony-stimulating factor in
human granulocyte colony-stimulating factor (rhG-CSF) what is patients with malignancy. Blood 1989;74:2644–2651.
the difference? Med Oncol 1998;15:229–233. 53. Lord BI, Gurney H, Chang J, et al. Haemopoietic cell kinetics in
36. Hoglund M, Hakansson L, Venge P. Effects of in vivo administra- humans treated with rGM-CSF. Int J Cancer 1992;50:26–31.
tion of G-CSF on neutrophil functions in healthy volunteers. Eur J 54. Lord BI, Bronchud MH, Owens S, et al. The kinetics of human
Haematol 1997;58:195–202. granulopoiesis following treatment with granulocyte colony-stim-
37. Holmes FA, O’Shaughnessy JA, Vukelja S, et al. Blinded, random- ulating factor. Proc Nad Acad Sci USA 1989;86:9499–9503.
ized, multicenter study to evaluate single administration pegfil- 55. Metcalf D. Clonal extinction of myelomonocytic leukaemia cells
grastim once per cycle versus daily filgrastim as an adjunct to by serum from mice infected with endodoxin. Int J Cancer
chemotherapy in patients with high-risk stage II or stage III/IV 1980;25:225–233.
breast cancer. J Clin Oncol 2002;20:727–731. 56. Metcalf D. The Molecular Control of Blood Cells. Cambridge,
38. Houston AC, Stevens LA, Cour V. Pharmacokinetics of glycosy- MA: Harvard University Press, 1988.
lated recombinant human granulocyte colony-stimulating factor 57. Metcalf D, Morstyn G. Colony stimulating factors: general biol-
(lenograstim) in healthy male volunteers. Br J Haematol ogy. In: De Vita V, ed. Biologic Therapy of Cancer. Philadelphia,
1999;47:279–284. PA: JB Lippincott, 1991:417–444.
39. Johnston E, Crawford J, Blackwell S, et al. Randomized, dose- 58. Molineux G, Kinstler O, Briddell B et al. A new form of filgrastim
escalation study of SD/01 compared with daily filgrastim in patients with sustained duration in vivo and enhanced ability to mobilize
receiving chemotherapy. J Clin Oncol 2000;18:2522–2528. PBPC in both mice and humans. Exp Hematol 1999;27:1724–1734.
40. Kawakami M, Tsutsumi H, Kumakawa I, et al. Levels of serum 59. Morstyn G, Dexter TM, Foote MA, eds. Filgrastim (r-metHuG-
granulocyte colony-stimulating factor in patients with infections. CSF) in Clinical Practice. New York: Marcel Dekker, 1998.
Blood 1990;76:1962–1964. 60. Morstyn G, Campbell L, Lieschke G, et al. Treatment of chemo-
41. Kawano Y,Takaue Y,Watanabe T et al. Effects of progenitor cell therapy-induced neutropenia by subcutaneously administered
dose and preleukapheresis use of human recombinant granulocyte granulocyte colony-stimulating factor with optimization of dose
colony-stimulating factor on the recovery of hematopoiesis after and duration of therapy. Clin Oncol 1989;7:1554–1562.
M. Foote and G. Morstyn 579

61. Morstyn G, Lieschke GJ, Calmar J, et al. Early clinical trials with interleukin-l-inducible production of colony-stimulating factors.
colony-stimulating factors. Cancer Invest 1989;7:443–456. Blood 1987;69:682–691.
62. Morstyn G, Burgess AW. Hemopoietic growth factors: a review. 80. Roskos LK, Charing EN, Vincent M, Foote MA, Morstyn G.
Cancer Res 1988;48:5624–5637. Pharmacology of filgrastim (r-metHuG-CSF). In: Morstyn G,
63. Morstyn G, Campbell L, Souza LM, et al. Effect of granulocyte Dexter TM, Foote MA, eds. Filgrastim (r-metHuG-CSF) in
colony-stimulating factor on neutropenia induced by cytotoxic Clinical Practice. New York: Marcel Dekker, 1998:51–71.
chemotherapy. Lancet 1988;1:667–672. 81. Sekino H, Moriya K, Sergano I, Wakabayashi K, Okazaki A.
64. Nagata S, Tsuchrya M, Asano S, et al. Molecular cloning and Recombinant human G-CSF (rG–CSF). Shinryo Shinyaku
expression of cDNA for human granulocyte colony stimulating 1989;26:32–104.
factor. Nature 1986;319:415–418. 82. Sheridan WP, Begley CG, Juttner CA, et al. Effect of peripheral-
65. Negrin RS, Greenberg PL. Filgrastim (r-metHuG-CSF) for the blood progenitor cells mobilised by filgrastim (G-CSF) on platelet
treatment of myelodysplastic syndromes. In: Morstyn G, Dexter recovery after high-dose chemotherapy. Lancet 1992;339:640–644.
TM, Foote MA, eds. Filgrastim (r-metHuG-CSF) in Clinical 83. Simmers RN, Webber LM, Shannon ME, et al. Location of the G-CSF
Practice. New York: Marcel Dekker, 1998:491–510. gene on chromosome 17 proximal to the break-point in the T(15;17)
66. Nicola NA, Metcalf D. Differential induction in leukemic cells by in acute promyelocytic leukemia. Blood 1987;70:330–332.
normal growth factor regulators: molecular and binding properties 84. Smith TJ, Khatcheressian J, Lyman GH, et al. 2006 update of
of purified granulocyte colony-stimulating factor. In: Bishop JM, recommendations for the use of white blood cell growth factors:
Rowley JD, Greaves M, eds. Genes and Cancer. New York: Alan R an evidence-based clinical practice guideline. J Clin Oncol.
Less, 1984:591–610. 2006;24:3187–3205.
67. Nicola NA. Hemopoietic cell growth factors and their receptors. 85. Souza LM, Boone TC, Gabrilove J, et al. Recombinant human
Ann Rev Biochem 1989;58:45–77. granulocyte colony-stimulating factor: effects on normal and
68. Nicola NA, Metcalf D, Matsumoto M, Johnson CR. Purification of leukemic myeloid cells. Science 1986;232:61–65.
a factor inducing differentiation in marine myelomonocytic leu- 86. Stahel RA, Jost LM, Cerny T, et al. Randomized study of recombi-
kaemia cells: identification as granulocyte colony-timulating fac- nant granulocyte colony-stimulating factor after high-dose chemo-
tor (G-CSF). J Biol Chem 1983;258:9017–9023. therapy and autologous bone marrow transplantation for high-risk
69. Nicola NA, Metcalf D. Biochemical properties of differentiation lymphoid malignancies. J Clin Oncol 1994;12:1931–1938.
factors for marine myelomonocytic leukemic cells in organ-condi- 87. Steward WP. Granulocyte and granulocyte-macrophage colony-
tioned media: Separation from colony stimulating factors. J Cell stimulating factors. Lancet 1993;342:153–157.
Physiod 1981;109:253–264. 88. Suzuki K, Tsunno M, Shishido A, Kuwabara T, Kobayashi S.
70. Nio Y, Shiraishi I, Tsubano M, et al. Comparative effects of a Phase I study of KW-2288 in normal healthy volunteers. J Clin
recombinant and a mutein type of granulocyte colony stimulating Ther Med 1991;7:1947–1971.
factor on the growth of fibrosarcoma with 5-fluorouracil chemo- 89. Takeshita A, Saito H, Toyama K, et al. Efficacy of a new formula-
therapy. Biotherapy 1992;4:81–86. tion of lenograstim (recombinant glycosylated human granulocyte
71. Nomura H, Imazeki I, Oheda M, et al. Purification and character- colony-stimulating factor) containing gelatin for the treatment of
ization of human granulocyte colony stimulating factor (GCSF). neutropenia after consolidation chemotherapy in patients with
EMBO J 1986;5:871–876. acute myeloid leukemia. Int J Hematol 2000;71:136–143.
72. Oheda M, Hasegawa M, Hatton K, et al. O-linked sugar chain of 90. Tanaka H, Kaneko T. Pharmacokinetic and pharmacodynamic
human granulocyte colony-stimulating factor protects it against comparisons between human granulocyte colony-stimulating fac-
polymerization and denaturation allowing it to retain its biological tor purified from human bladder carcinoma cell line 5637 culture
activity. Biol Chem 1990;265:11482–11485. medium and recombinant human granulocyte colony-stimulating
73. Oheda M, Huse S, Ono M, Ikenaka T. Structure of the sugar chains of factor produced in Escherichia coli. J Pharm ExpTher
recombinant human granulocyte colony-stimulating factor produced 1992;262:439–444.
by Chinese hamster ovary cells. J Biochem 1988;103:544–546. 91. Trillet-Lenoir V, Green J, Manegold C, et al. Recombinant granulo-
74. Osslund T, Boone T. Biochemistry and structure of filgrastim cyte colony stimulating factor reduces the infectious complications
(r-metHuG-CSF). In: Morstyn G, Dexter TM, Foote MA, eds. of cytotoxic chemotherapy. Eur J Cancer 1993;29A:319–324.
Filgrastim (r-metHuG-CSF) in Clinical Practice. New York: 92. Turzanski J, Crouch SP, Fletcher J, Hunter A. Ex vivo neutrophil
Marcel Dekker, 1998: 41–49. function in response to three different doses of glycosylated rHuG-
75. Platt OS, Brambilla DJ, Rosse WE, et al. Mortality in sickle cell CSF (Lenograstim). Br J Haematol 1997;96:46–54.
disease life expectancy and risk factors for early death. N Engl J 93. van der Auwera P, Platzer E, Xu ZX, et al. Pharmacodynamics and
Med 1994;330:1639–1644. pharmacokinetics of single doses of subcutaneous pegylated
76. Platzer E. Human hemopoietic growth factors. Eur J Haematol human G-CSF mutant (Ro 258315) in healthy volunteers: com-
1989;42:1–15. parison with single and multiple daily doses of filgrastim. Am J
77. Raghavachar A, Schrazenmeier H, Bacigalupo A. Use of filgrastim Hematol 2001;66:245–251.
(r-metHuG-CSF) in aplastic anemia. In: Morstyn G, Dexter TM, 94. Van der Wouw PA, van Leeuwen R, van Oers RH, et al. Effects of
Foote MA, eds. Filgrastim (r-metHuG-CSF) in Clinical Practice. recombinant human granulocyte colony-stimulating factor on leu-
New York: Marcel Dekker, 1998:533–551. copenia in zidovudine-treated patients with AIDS and AIDS-related
78. Rapoport AP, Abboud CN, Di Persio JF. Granulocyte-macrophage complex, a phase IIII study. Br J Haematol 1991;78:319–324.
colony-stimulating factor (GM-CSF) and granulocyte colony- 95. Vellenga E, Rambaldi A, Ernst TI, Ostapovicz D, Griffin JD.
stimulating factor (G-CSF): receptor biology, signal transduction, Independent regulation of M-CSF and C-CSF gene expression in
and neutrophil activation. Blood Rev 1992;6:43–57. human monocytes. Blood 1988;71:1529–1532.
79. Rennick D, Yang G, Gammell L, et al.. Control of hemopoiesis 96. Walker BD. Immune reconstitution: is there a potential role for fil-
by a bone marrow stroma cell clone: lipopolysaccharide- and grastim (r-metHuG-CSF)? J Hematother Stem Cell Res 1999;8:Sl 2.
580 Granulocyte colony-stimulating factor: biology and clinical potential

97. Watari K, Asano S, Shirafuji N, et al. Serum granulocyte colony- stimulating factor. Proc Natl Acad Sci USA 1985;82:
stimulating factor levels in healthy volunteers and patients with 1526–1530.
various disorders as estimated by enzyme immunoassay. Blood 102. World Health Organization (WHO). International collabora-
1989;73:117–122. tive study for the proposed international standards for granu-
98. Watts MI, Sullivan AM, Jamieson E, et al. Progenitor-cell mobili- locyte-macrophage colony-stimulating factor and granulocyte
zation after low-dose cyclophosphamide and granulocyte colony- colony-stimulating factor. WHO Technical Report Series,
stimulating factor: an analysis of progenitor-cell quantity and quality 1994.
and factors predicting for these parameters in 101 pretreated patients 103. Yamamoto Y, Klein TW, Friedman H, Kimura S, Yamaguchi H.
with malignant lymphoma. J Clin Oncol 1997;15:535–546. Granulocyte colony-stimulating factor potentiates anti-Candida
99. Weisbart RH, Golde DW. Physiology of granulocyte and mac- alhicans growth inhibitory activity of polymorphonuclear cells.
rophage colony-stimulating factors in host defense. Hematol FEM Immunol Med Microbiol 1993;7:15–22.
Oncol Clin N Am 1989;3:401–409. 104. Yoshida Y, Hirashima K, Asano S, et al. A phase II trial of recom-
100. Welte K, Gabrilove J, Bronchud MH, Platzer E, Morstyn G. binant human granulocyte colony-stimulating factor in the myel-
Filgrastim (r-metHuG-CSF): the first 10 years. Blood odysplastic syndromes. Br J Haematol 1991;78:378–84.
1996;86:1907–1929. 105. Zsebo KM, Cohen AM, Murdock DC, et al. Recombinant human
101. Welte K, Platzer E, Lu L, et al. Purification and biochemical granulocyte colony-stimulating factor: molecular and biological
characterization of human pluripoent hematopoietic colony- characterization. Immunobiology 1986;172:175–184.
18 Granulocyte-macrophage colony-stimulating
factor
MARYANN FOOTE AND GEORGE MORSTYN

Introduction 35,000 daltons depending upon the degree of glycosyla-


tion. The gene has been expressed using recombinant
Granulocyte-macrophage colony-stimulating factor DNA technology in bacteria (Escherischia coli), Chinese
(GM-CSF) is one of the colony-stimulating factors hamster ovary (CHO) cells, and yeast (Saccharomyces
whose name is derived from its major target cell lin- cerevisiae) resulting in the availability of large amounts
eages. Since the original studies, which characterized of glycosylated and nonglycosylated material for clinical
its ability to stimulate the clonal proliferation of myel- trials. Small differences in the amino acid sequence, as
oid precursors, however, a broader range of biologic well as major differences in the degree of glycosylation,
effects on mature, effector cells, and the immune sys- can be found among the preparations. The growth factor
tem, have been identified. These expanded areas of produced by expression of the cDNA in bacteria is not
activity have carried its more recent clinical application glycosylated [16], but that expressed in mammalian
beyond that of ‘colony-stimulating factor’ into the cells and yeast demonstrates variable degrees of glyco-
realm of immunomodulation. sylation [20]. Both the glycosylated and nonglycosy-
GM-CSF was initially purified from media used to lated forms are active in vitro or in vivo, and have
culture an HTLV-II-infected, T lymphoblastoid cell line similar activity to the natural form of the protein, but the
(Mo) [5], but was subsequently found to be produced by degree of glycosylation plays a role in inducing antibody
a variety of cell types including macrophages, endothe- formation, as well as altering some specific biologic
lial cells, and certain mesenchymal cells. Although basal activities and toxicities.
serum concentrations of GM-CSF are often not measur-
able in normal adults, its elaboration is readily inducible
in ‘producer’ cells by immune stimuli and many of the
mediators of inflammation.
Physiology
In 1985, GM-CSF was the first myeloid hematopoi- Both endogenous and recombinant GM-CSF generate
etic growth factor, to have its gene sequenced and cloned predominantly granulocyte and monocyte colonies in
[20, 92]. GM-CSF is used for the native form of the semisolid culture systems. It acts as a potent stimulus for
molecule or when it is not necessary to distinguish a the growth of uncommitted and committed bone marrow
specific form of the recombinant molecule, which is progenitors including colony-forming unit–granulocyte,
referred to as rHuGM-CSF. erythrocyte, monocyte, and megakaryocyte (CFU–
GEMM) [61, 77]. It is a potent inducer of granulocyte-
monocyte differentiation, and potentiates the growth of
Biochemistry and Structure human blast-forming units–erythroid (BFU-E) in the
presence of erythropoietin in vitro.
The gene for GM-CSF has been mapped to the long At the level of mature cells, GM-CSF enhances the
arm of chromosome 5 (5q21–32) [51] in the cluster function of neutrophilic and eosinophilic granulocytes,
region of genes for other growth factor proteins and their and of monocyte/macrophages. Exposure of neutrophils
cell-surface receptors, such as interleukin-3 (IL-3) [56], to GM-CSF produces increased expression of cellular
IL-4 [93], IL-5 [83], macrophage colony-stimulating adhesion molecules [5, 23, 41] such as CDllb, increased
factor and its receptor (the c-fms proto-oncogene expression of class II MHC molecules [38, 63], increased
product) [70], and the receptor for platelet-derived number of FMLP receptors [88], and increased fMLP-
growth factor [93]. induced superoxide production [87], chemotaxis, anti-
The gene for GM-CSF encodes a protein of 127 body dependent cellular cytotoxicity (ADCC), and
amino acids whose molecular weight varies from l4 to phagocytosis. In vitro, it was reported to inhibit random

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 581
© Springer Science + Business Media B.V. 2009
582 Granulocyte-macrophage colony-stimulating factor

neutrophil granulocyte migration (8,334) and in vivo, not glycosylated, is available in Europe and other
when administered by continuous infusion, it prevented countries.
neutrophils from migrating into areas of inflammation [69].
GM-CSF prolongs the survival of neutrophilic and Pharmacodynamics and
eosinophilic granulocytes in vitro [60] by inhibiting Pharmacokinetics
programmed cell death [11], and enhances microbicidal
activity and their leukotriene synthesis [79]. Exposure Several early pharmacodynamic studies defined the
of macrophages to GM-CSF increases expression of pharmacology of rHuGM-CSF [58]. Radiolabeling of
cell-surface adhesion molecules such as CD11a, b, and leukocytes suggested that a transient leukopenia is due
c; and Fc RII (CDw32) receptors involved in phagocy- to sequestration of the cells within the lung. In vitro and
tosis [72, 91]. In vitro, it enhances ADCC, phagocyto- in vivo studies have suggested that this effect may be
sis, microbicidal and tumoricidal activity [39, 44, 89], due to a change in cell surface adhesion molecules
and the synthesis and secretion of other cytokines on the leukocytes such as those identified by CD11b
including those related to inflammation such as tumor [5, 80], which is upregulated on neutrophils, as well as
necrosis factor (TNF) and IL-1. LAM-1, which is downregulated, and adherence to
GM-CSF exerts its biologic activity by binding to spe- endothelial cells of the blood vessels [41].
cific transmembrane surface receptors, which are subse- The major effect of rhGM-CSF on hematopoiesis is
quently internalized, on the target cells [24, 33]. These the leukocytosis that was reported to be dose dependent
receptors have been detected on mature neutrophils, in most of the early studies [58] when either glycosy-
monocytes/macrophages, some lymphocytes, normal lated or nonglycosylated rHuGM-CSF was administered
bone marrow progenitors, dendritic cells (DC), fibroblasts, daily subcutaneously (SC) or by prolonged intravenous
endometrial and endothelial cells, fresh leukemic cells, (IV) or SC infusions. The pattern of the leukocytosis in
and leukemic cell lines [13, 14,18, 66]. GM-CSF itself can many of these trials appeared as a biphasic response: an
downregulate these receptors on cells such as neutrophils, initial increase that frequently plateaued during the first
monocytes, and normal bone marrow myeloid cells few days of treatment, followed by a decline. With con-
in vitro [18]. tinued dosing, however, another increase in leukocytes,
Crosslinking experiments have revealed molecular which often contained more immature myeloid cells
weights of 75–156 kD for the receptors. At least two such as myelocytes, promyelocytes, and myeloblasts,
types of GM-CSF receptors have been identified: one was observed. Appearance of monocytes and eosino-
with high affinity (20–100 pmol/L) and one with low phils generally occurred later in the course of treatment
affinity (1 nmol/L), but as many as four types may exist and at higher doses of rHuGM-CSF. Morphologic
on AML cells [13]. Two subunits (alpha and beta) changes in neutrophils included toxic granulation with
comprise the heterodimer receptor and both have been an increase in leukocyte alkaline phosphatase, promi-
cloned [46]. The alpha subunit is specific to the GM-CSF nent Döhle bodies, cytoplasmic vacuolization and, at
receptor. At least two distinct regions within the higher doses, increasing numbers of hypersegmented
cytoplasmic domain of the common beta subunit have neutrophils. At the higher doses of rHuGM-CSF in
been shown to be responsible for different signals [74]. patients with normal marrows, the cellularity of the
A membrane proximal region of approximately 60 bone marrow was increased with a left-shift and an
amino acids has been shown to be essential for induction increase in the myeloid-to-erythroid ratio [50, 58].
of c-myc and activation of DNA replication, which An important observation made during these early
involves the tyrosine phosphorylation of a Janus kinase trials, which later found significant clinical application,
(JAK2) and activation of its in vitro kinase activity [71]. was the dose-dependent increase in peripheral blood
A second, distal region of about 140 amino acids is erythroid and myeloid progenitors [59]. Bone marrow
required for activation of Ras, Raf-1, MAP kinase and progenitors were found to be unchanged or decreased,
p70 S6 kinase, and induction of c-fos and c-jun [74]. probably somewhat dependent upon the degree of
expansion of the non-colony forming myeloid mononu-
clear cells [62].
Pharmacokinetic studies were conducted using both
Pharmacology glycosylated and nonglycosylated rHuGM-CSF by
Both glycosylated and nonglycosylated forms of radioimmunoassay and by bioassay. Route of adminis-
rHuGM-CSF have been studied clinically. Glycosylated tration affected the peak serum concentration, area
rHuGM-CSF (sargramostim), produced in yeast, is under the concentration-time curve, and the time during
available in the United States. Molgramostim, which is which GM-CSF was detectable in the serum.
Maryann Foote and George Morstyn 583

Clinical Implications of chemotherapy have been used to supplement bone


marrow after autologous bone marrow transplantation
Much clinical data have been published about the various [36]; to support dose-intensified chemotherapy when
forms of rHuGM-CSF and their uses in many therapeutic growth factor alone is not adequate [76]; and when autol-
areas. It is impossible to cite all the important papers, and ogous bone marrow alone was used for hematopoietic
we have concentrated on reports of pivotal studies. support [27, 43]. Although the early trials were conducted
primarily in patients undergoing autologous transplant,
the technique has been applied to normal donors and for
Chemotherapy-induced Neutropenia allogeneic stem cell transplant harvesting.
Many clinical trials have studied the ability of rHuGM- PBPC were initially used in combination with autolo-
CSF to abrogate the hematologic toxicity of chemother- gous bone marrow, and were found to reduce the time to
apy (both standard and dose-intensified) and bone hematologic reconstitution [12, 36, 68]. Durable reconsti-
marrow/stem cell transplantation. Most of the early tution was demonstrated using PBPC alone [37, 68, 76].
studies were not randomized studies, included relatively CSF-mobilized PBPC are preferred because of the ability
small numbers of patients, compared the results of to harvest great numbers of PBPC, the simplicity of har-
cycles of chemotherapy given without growth factor to vesting, the ability to harvest from patients with a history
cycles given with growth factor, and cycles of chemo- of pelvic irradiation and/or other causes of poor bone
therapy given with growth factor compared with data of marrow reserve, and the rapidity of engraftment [9].
historical controls. In most studies, rHuGM-CSF did For autologous, allogeneic, and syngeneic transplan-
not completely eliminate profound leukopenia. In gen- tation, rHuGM-CSF has been administered after the con-
eral, the neutrophil nadir occurred earlier in the courses ditioning regimen and reinfusion of bone marrow and/or
of chemotherapy in which rHuGM-CSF was adminis- PBPC to shorten the duration and reduce the severity of
tered than in the courses without. Higher doses of the neutropenia, and thus decrease the requirement for sup-
growth factor in some studies decreased the depth of the portive care, and the duration of hospitalization [10, 55,
absolute neutrophil count (ANC) nadir, and frequently 64, 65].
the duration of neutropenia and time to an ANC > 1.0 ×
109/L was shortened at during the first cycle of chemo-
therapy administered with rHuGM-CSF. The results
Bone Marrow Failure States
with respect to the ability to reduce the morbidity (hema- GM-CSF has multilineage activity, in vitro and some-
tologic, and in some cases the incidence of infection and what more variably in vivo. It can also enhance effector
mucositis) of dose intensification was variable. cell function, and induces differentiation of some leuke-
Furthermore, even in the larger randomized trials [6, 15, mic cell lines. Based on these activities, it was thought
35, 53], efficacy was variable. In most cases, duration of to be a natural candidate to study as therapy for patients
neutropenia was reduced at least during the first cycle; with myelodysplastic syndromes (MDS), who frequently
however, persistence of this effect on subsequent cycles, have bi- and tri-lineage cytopenias, as well as dysfunc-
and impact on infections was much more variable. tional granulocytes. The focus of most of these studies
was improvement in blood counts, and determination if
Bone Marrow and Stem Cell myeloblasts were stimulated [3, 26, 32, 40, 49, 84, 85].
A wide range of doses were administered by a variety of
Transplantation dosing schedules for all of the preparations of rHuGM-
One of the areas of more intense research has been derived CSF. The reported studies, for the most part, have
from the ability to mobilize peripheral blood stem cells included relatively small numbers of patients. Their dis-
(PBPC). Although it is difficult to collect PBPC from a ease status covered the spectrum from refractory anemia
patient who is in a hematologic steady-state, the number (RA) to the poorer prognosis refractory anemia with
of circulating stem cells increases during blood cell count excess of blasts (RAEB), refractory anemia with excess
recovery after treatment with chemotherapy, especially of blasts in transformation (RAEB-t), and chronic myel-
after cyclophosphamide. The early trials showed that omonocytic leukemia (CMMoL). In general, these studies
CSF alone could increase the number of circulating PBPC reported comparable increases in neutrophil counts but
[80], and when administered after chemotherapy the no consistent improvement in the other hematopoietic
number was enhanced even further [78]. cell lines [29]. An increase in bone marrow and periph-
The PBPC harvested by leukopheresis during rHuGM- eral blood myeloblasts and conversion to acute myelog-
CSF priming without chemotherapy or while a patient is enous leukemia (AML) occurred more often in patients
receiving growth factor to abrogate the myelosuppression with RAEB and RAEB-t, but in many cases, the number
584 Granulocyte-macrophage colony-stimulating factor

of myeloblasts decreased and/or returned to baseline of these agents in the pediatric population was extremely
upon discontinuation of the growth factor. In a number of limited by concerns about their long-term effects. One
patients, the drug was discontinued due to progression of the earliest areas of investigation for myeloid growth
of disease or to side effects. Several investigators tried to factors in pediatric patients was chronic severe neutro-
circumvent the issues of toxicity and myeloblast stimu- penia [31, 90]. Because of the limited numbers of studies
lation by manipulating the dosing of the GM-CSF [54]. using these agents, that permitted enrollment of children,
the safety and efficacy was not established until consid-
erably later than when the same was defined for adults.
Acute Leukemia In pediatric oncology, these agents were used as adjuncts
Use of hematopoietic growth factors has taken a number to chemotherapy and radiation to facilitate engraftment
of approaches in patients with acute leukemia: for facili- after transplantation, and to mobilize PBPC. Guidelines
tating recovery from the myelosuppressive effects of for their use in children were not available until a
induction and/or consolidation chemotherapy to reduce European expert panel convened to define them and
morbidity; as a means of sensitizing myeloid leukemic finally published their recommendations in 1998 [75].
cells to chemotherapy by recruitment of these cells into Until that time, guidelines published by ASCO were
S-phase followed by treatment with a cycle-specific extended to children, despite the lack of clinical trials to
drug, for harvesting PBPC (after intensive chemotherapy) support those recommendations [1, 2]. The ASCO
that may be used for stem cell support after bone marrow guidelines for clinical use of CSFs: 2000 update made
ablation; and as differentiation/maturation therapy in no significant changes to its previous recommendations
myeloid leukemia. There has been appropriate concern for their use in the pediatric population [67].
about possibly stimulating the leukemic population, Clinical trials using G- and GM-CSF as mobilizing
especially in AML, with rHuGM-CSF since such activity agents for PBPC have demonstrated efficacy for collect-
has been reported in vitro [42, 62, 86]. Sargramostim ing autologous stem cells that can reduce the period of
showed reduction in early deaths due to fungal infections neutropenia compared with bone marrow. Based on
and reduction in neutropenia in a small randomized trial such studies, the European guidelines recommended the
that led to its registration in this setting. routine use of CSF in children for this purpose. On the
The second approach taken in the use of myeloid other hand, despite the now common use of CSF to
growth factors in patients with AML has been an attempt mobilize PBPC from normal donors in the adult popula-
to cycle activate leukemic cells, thus sensitizing them to tion, there has been significant hesitation in extending
cycle-specific chemotherapy, such as cytosine arabino- that practice to the pediatric population. The limited
side, and potentially improving remission rates [4, 8, 17, information available regarding the long-term effects of
25, 30, 47]. None of the studies showed an absolute CSF in normal pediatric subjects resulted in the state-
change >15% in the S-phase fraction of cells during ment by the European panel that such use was
treatment with rHuGM-CSF; in general the percent contraindicated.
change was small.
The use of rHuGM-CSF in acute lymphoblastic leu-
kemia (ALL) has been less controversial, but even fewer
Immunomodulation
studies have been reported. The complex nature of most GM-CSF has a broad spectrum of activities that can con-
ALL chemotherapy regimens, which include frequent tribute to the immune response in humans. Early in the
dosing, and the difficulty in determining appropriate studies characterizing this protein, it was recognized that
growth factor dosing, has undoubtedly contributed to GM-CSF could induce production of other cytokines
the limited number of clinical trials. and growth factors such as IL-1, IL-6, and tumor necro-
sis factor (TNF) that also contribute to the expansion of
B and T cells, as well as induce or coinduce TNF-α gene
expression with interferon (IFN)-γ in monocytes [19, 45]
Current Issues and in conjunction with IL-2, costimulates T-cell prolif-
eration [73].
Use in Children As the biology of DC has been better defined, it has
Although G- and GM-CSF were placed into clinical trials become evident that in vitro and possibly in vivo,
in the late 1980s and approved by the FDA in 1991, GM-CSF is a cofactor in the differentiation and func-
most clinical trials have been conducted in adult patients. tional activities of these cells. DC are antigen-presenting
As is commonly the case with new agents, the evaluation cells that have a critical role in T-cell immune responses
Maryann Foote and George Morstyn 585

[7, 11, 15, 82]. DC can be differentiated from CD34+


hematopoietic progenitor cells in human bone marrow,
References
peripheral blood, and cord blood [21, 22] when cultured 1. American Society of Clinical Oncology. Recommendations for the
use of hematopoietic colony-stimulating factors: evidence-based,
with GM-CSF plus TNF. They can be cultured from clinical practice guidelines. J Clin Oncol 1994;12:2471–2508.
monocytes using GM-CSF plus IL-4. Antigens are pre- 2. American Society of Clinical Oncology. Update of recommenda-
sented on their surface in association with class II MHC tions for the use of hematopoietic colony-stimulating factors:
molecules, to be recognized by T4 helper cells. The evidence-based clinical practice guidelines. J Clin Oncol 1996;14:
1957–1960.
CD4+ cells participate in the development of B cells,
3. Antin JH, Smith BR, Holmes W, Rosenthal DS. Phase I/II study of
antigen-specific cytotoxic T8 cells; and macrophages, recombinant human granulocyte-macrophage colony-stimulating
eosinophils, and natural killer (NK) cells, which are factor in aplastic anemia and myelodysplastic syndrome. Blood
antigen nonspecific [7]. All these cells may be involved 1988;72:705–713.
in an immune response. In addition to its activity on the 4. Archimbaud E, Fenaux P, Reiffers J, et al. Granulocyte-macrophage
colony-stimulating factor in association to timed-sequential chemo-
function of DC, GM-CSF increases expression of class
therapy with mitoxantrone, etoposide, and cytarabine for refractory
II MHC molecules on the DC, which are essential to acute myelogenous leukemia. Leukemia 1993;7:372–377.
recognition. It is an enhancer of costimulatory molecule 5. Arnout MA, Wang EA, Clark SC, Sieff CA. Human recombinant
expression – such as B7 and adhesion molecules like granulocyte-macrophage colony-stimulating factor increases cell-
ICAM that are necessary for the interaction of antigen to-cell adhesion and surface expression of adhesion-promoting
surface glycoproteins on mature granulocytes. J Clin Invest
presenting cells and T cells. 1986;78:597–601.
6. Bajorin D, Nichols CR, Schmoll H-J, et al. Recombinant human
granulocyte-macrophage colony-stimulating factor as an adjunct
Vaccine Therapy to conventional-dose ifosfamide based chemotherapy for patients
with advanced or relapsed germ cell tumors: A randomized trial. J
Since the mid 1990s, many nonclinical and clinical studies Clin Oncol 1995;13:79–86.
have used genetic engineering techniques to harness the 7. Banchereau J, Briere F, Caux C, et al. Immunology of dendritic
immunomodulatory activity of GM-CSF to augment cells. Annu Rev Immunol 2000;18:767–811.
antitumor immunity. Using tumor cell lines or autologous 8. Bettelheim P, Valent P, Andreeff M, et al. Recombinant human
granulocyte-macrophage colony stimulating factor in combination
tumor cells genetically modified to produce GM-CSF,
with standard induction chemotherapy in de novo acute myeloid
investigators have shown that this immunostimulatory leukemia. Blood 1991;77:700–711.
protein can enhance tumor immunogenicity in a variety 9. Bishop MR, Anderson JR, Jackson JD, et al. High-dose therapy
of tumors. Replicating tumor cells are transduced with and peripheral blood progenitor cell transplantation: Effects of
viral supernatants harvested from CRIP packaging cell recombinant human granulocyte-macrophage colony-stimulating
factor on the autograft. Blood 1994;83:610–616.
lines transfected with MFG-S-human GM-CSF. They
10. Blazar BR, Kersey JH, McGlave PB et al. In vivo administration
are irradiated and administered as vaccine to tumor- of recombinant human granulocyte/macrophage colony stimulating
containing animals or patients. Delayed-type hypersen- factor in acute lymphoblastic leukemia patients receiving purged
sitivity skin responses were demonstrated against autografts. Blood 1989;73:849–857.
irradiated autologous cancer cells after vaccination [52]. 11. Brach MA, deVos S, Gruss HJ, Herrmann F. Prolongation of sur-
vival of human polymorphonuclear neutrophils by granulocyte-
Biopsies of vaccine sites showed recruitment of DC, T macrophage colony-stimulating factor is caused by inhibition of
cells, and eosinophils [48, 81], regression of metastatic programmed cell death. Blood 1992;80:2920–2924.
lesions was observed in some studies [28, 52, 57] and 12. Brandt SJ, Peters WP, Atwater SK, et al. Effect of recombinant
prolongation of survival [25, 48]. human granulocyte-macrophage colony stimulating factor on
hematopoietic reconstitution after high dose chemotherapy and
autologous bone marrow transplantation. N Engl J Med 1988;318:
869–876.

Conclusions 13. Budel LM, Elbaz O, Hoogerbrugge H, et al. Common binding


structure for granulocyte macrophage colony-stimulating factor
and interleukin-3 on human acute myeloid leukemia cells and
rHuGM-CSF has found limited adoption for the pre-
monocytes. Blood 1990;75:1439–1445.
vention of neutropenia and infections in the induction 14. Budel LM, Touw IP, Delwel R, Clark SC, Lowenberg B.
therapy of acute myeloid leukemia. It has been used Interleukin-3 and granulocyte-monocyte colony-stimulating factor
for the mobilization of progenitor cells for PBPC receptors on human acute myelocytic leukemia cells and relationship,
transplantation. Its effects on other cells of the immune to the proliferative response. Blood 1989;74:565–571.
15. Bunn PA, Crowley J, Kelly K, et al. Chemoradiotherapy with or
system acting as a vaccine adjuvant and in DC therapy
without granulocyte-macrophage colony-stimulating factor in the
are not approved by the FDA for clinical use. In the treatment of limited-stage small-cell lung cancer: A prospective
future, rHuGM-CSF in these settings may have great phase III randomized study of the Southwest Oncology Group. J Clin
utility. Oncol 1995;13:1632–1641.
586 Granulocyte-macrophage colony-stimulating factor

16. Burgess AW, Begley CG, Johnson GR, et al. Purification and prop- 31. Ganser A, Ottman OG, Erdmann H, et al. The effect of recombinant
erties of bacterially synthesized human granulocyte macrophage human granulocyte-macrophage colony-stimulating factor on neu-
colony stimulating factor. Blood 1987;69:43–51. tropenia and related morbidity in chronic severe neutropenia. Ann
17. Cannistra SA, DiCarlo J, Groshek P, et al. Simultaneous adminis- Intern Med 1989;111:887–892.
tration of granulocyte-macrophage colony-stimulating factor and 32. Ganser A, Volkers B, Greher J, et al. Recombinant human granulo-
cytosine arabinoside for the treatment of relapsed acute myeloid cyte-macrophage colony-stimulating factor in patients with myelo-
leukemia. Leukemia 1991;5:230–238. dysplastic syndromes – A phase I/II trial. Blood 1989;73:31–37.
18. Cannistra SA, Groshek P, Garlick R, et al. Regulation of surface 33. Gasson JC, Kaufman SE, Weisbart RH, Tomonaga M, Golde DW.
expression of the granulocyte/macrophage colony stimulating fac- High-affinity binding of granulocyte-macrophage colony stimulating
tor receptor in normal human myeloid cells. Proc Natl Acad Sci factor to normal and leukemic human myeloid cells. Proc Natl Acad
USA 1990;87:97. Sci USA 1986;83:669–673.
19. Cannistra SA, Rambaldi A, Spriggs DR, Herrmann F, Kufe D, 34. Gasson JC, Weisbart RH, Kaufman SE, et al. Purified human gran-
Griffin JD. Human granulocyte-macrophage colony-stimulating ulocyte-macrophage colony-stimulating factor: Direct action on
factor induces expression of the tumor necrosis factor gene by the neutrophils. Science 1984;226:1339–1342.
U937 cell line and by normal human monocytes. J Clin Invest 35. Gerhartz HH, Engelhard M, Meusers P, et al. Randomized, double-
1987;79:1720–1728. blind, placebo-controlled, phase III study of recombinant human
20. Cantrell MA, Anderson D, Ceretti DP, et al. Cloning sequence and granulocyte-macrophage colony-stimulating factor as adjunct to
expression of human granulocyte/macrophage colony stimulating induction treatment of high-grade malignant non-Hodgkin’s lym-
factor. Proc Natl Acad Sci USA 1985;73:6250–6254. phomas. Blood 1993;82:2329–2339.
21. Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. GM-CSF 36. Gianni AM, Bregni M, Stern AC, et al. Granulocyte-macrophage
and TNF-alpha cooperate in the generation of dendritic Langerhans colony-stimulating factor to harvest circulating hemopoietic stem
cells. Nature 1992;360:258–261. cells for autotransplantation. Lancet 1989;580–584.
22. Caux C, Massacrier C, Vanbervliet B, et al. CD34+ hematopoietic 37. Gianni AM, Tarella C, Siena S, et al. Durable and complete
progenitors from human cord blood differentiate along two inde- hematopoietic reconstitution after autografting of rhGM-CSF
pendent dendritic cell pathways in response to granulocyte- exposed peripheral blood progenitor cells. Bone Marrow Transplant
macrophage colony-stimulating factor plus tumor necrosis factor 1990;6:143–145.
alpha: II. Functional analysis. Blood 1997;90:1458–1470. 38. Gosselin EJ, Wardwell K, Rigby WF, Guyre PM. Induction of
23. Devereux S, Bull HA, Campos-Costa D, et al. Granulocyte mac- MHC class II on human polymorphonuclear neutrophils by granu-
rophage colony stimulating factor induced changes in cellular adhe- locyte/macrophage colony-stimulating factor, INF-gamma and
sion molecule expression and adhesion to endothelium: in-vitro and IL-3. J Immunol 1993;151:1482–1490.
in-vivo studies in man. Br J Haemoto1 1989;71:323–330. 39. Grabstein KH, Urdal DL, Tushinski RJ, et al. Induction of mac-
24. DiPersio J, Billing P, Kaufman S, et al. Characterization of the rophage tumoricidal activity by granulocyte-macrophage colony-
human granulocyte-macrophage colony-stimulating factor recep- stimulating factor. Science 1986;232:506–508.
tor. J Biol Chem 1988;263:1834–1841. 40. Gradishar W, Le Beau MM, O’Laughlin R, et al. Clinical and cyto-
25. Dunussi-Joannopoulos K, Dranoff G, Weinstein HJ, Ferrara JL, genetic responses to GM-CSF in therapy-related myelodysplastic
Bierer BE, Croop JM. Gene immunotherapy in murine acute myel- syndrome. Blood 1992;80:2463–2470.
oid leukemia: granulocyte-macrophage colony-stimulating factor 41. Griffin JD, Spertini O, Ernst TJ, et al. Granulocyte-macrophage
tumor cell vaccines elicit more potent antitumor immunity com- colony-stimulating factor and other cytokines regulate surface
pared with B7 family and other cytokine vaccines. Blood expression of the leukocyte adhesion molecule-1 on human neu-
1998;91:222–230. trophils, monocytes, and their precursors. J lmmunol 1990;145:
26. Economopoulos T, Papageorgiou E, Stathakis N, et al. Treatment 576–584.
of myelodysplastic syndromes with human granulocyte-mac- 42. Griffin JD, Young D, Herrmann F, et al. Effects of recombinant
rophage colony-stimulating factor (GM-CSF) or GM-CSF com- human GM-CSF on proliferation of clonogenic cells in acute
bined with low-dose cytosine arabinoside. Eur J Haemato1 myeloblastic leukemia. Blood 1986;67:1448–1453.
1999;49:138–142. 43. Haas R, Ho AD, Bredthauer U, et al. Successful autologous trans-
27. Elias AD, Ayash L, Anderson KC, et al. Mobilization of peripheral plantation of blood stem cells mobilized with recombinant human
blood progenitor cells by chemotherapy and granulocyte-macrophage granulocyte-macrophage colony-stimulating factor. Exp Hematol
colony-stimulating factor for hematologic support after high-dose 1990;18:94–98.
intensification for breast cancer. Blood 1992;79:3036–3044. 44. Handman E, Burgess AW. Stimulation by granulocyte-macrophage
28. Ellem KA, O’Rourke MG, Johnson GR, et al. A case report: colony-stimulating factor of Leishmania tropica killing by mac-
immune responses and clinical course of the first human use of rophages. J Immunol 1979;120:1134–1137.
granulocyte/macrophage-colony-stimulating-factor-transduced 45. Hart PH, Whitty GA, Piccoli DS, Hamilton JA. Synergistic activa-
autologous melanoma cells for immunotherapy. Cancer Immunol tion of human monocytes by granulocyt-macrophage colony-stim-
Immunother 1997;44:10–20. ulating factor and IFN-gamma: Increased TNF-alpha but not IL-1
29. Estey EH, Kurzrock R, Talpaz M, et al. Effects of low doses of activity. J Immunol 1988;141:1516–1521.
recombinant human granulocyte-macrophage colony stimulating 46. Hayashida K, Kitamura T, Gorman DM, et al. Molecular cloning
factor (GM-CSF) in patients with myelodysplastic syndromes. Br of a second subunit of the receptor for human granulocyte-
J Haematol 1991;77:291–295. macrophage colony-stimulating factor (GMCSF): Reconstitution
30. Estey E, Thall PF, Kantarjian H, et al. Treatment of newly diag- of a high-affinity GM-CSF receptor. Proc Natl Acad Sci USA
nosed acute myelogenous leukemia with granulocyte-macrophage 1990;87:9655–9659.
colony-stimulating factor (GM-CSF) before and during continuous- 47. Heil G, Chadid L, Hoelzer D, et al. GM-CSF in a double-blind ran-
infusion high-dose ara-C+ daunorubicin: Comparison to patients domized, placebo controlled trial in therapy of adult patients with
treated without GM-CSF. Blood 1992;79:2246–2255. de novo/acute myeloid leukemia (AML). Leukemia 1995;9:3–9.
Maryann Foote and George Morstyn 587

48. Herrlinger U, Kramm CM, Johnston KM, et al. Vaccination for 64. Nemunaitis J, Singer JW, Buckner CD, et al. Use of recombinant
experimental gliomas using GM-CSF-transduced glioma cells. human granulocyte-macrophage colony-stimulating factor in
Cancer Gene Ther 1997;4:345–352. autologous marrow transplantation for lymphoid malignancies.
49. Hermann F, Lindemann A, Klein H, et al. Effect of recombinant Blood 1988;72:834–836.
human granulocyte-macrophage colony-stimulating factor in 65. O’Day SJ, Rabinowe SIN, Neuberg D, et al. A phase II study of
patients with myelodysplastic syndrome with excess blasts. continuous infusion recombinant human granulocyte-macrophage
Leukemia 1989;3:335–338. colony-stimulating factor as an adjunct to autologous bone mar-
50. Herrmann F, Schulz G, Lindemann A, et al. Yeast expressed gran- row transplantation for patients with non-Hodgkin’s lymphoma in
ulocyte-macrophage colony-stimulating factor in cancer patients: first remission. Blood 1994;83:2707–2714.
A phase Ib clinical study. Behring Inst Mitt 1988;83:107–118. 66. Onetto-Pothier N, Aumont N, Haman A, et al. Characterization of
51. Huebner K, Isobe M, Croce CM, et al. The human gene encoding granulocyte-macrophage colony-stimulating factor receptor on the
GM-CSF is at 5q21-q32, the chromosome region deleted in the blast cells of acute myeloblastic leukemia. Blood 1990;75:59–66.
5q- anomaly. Science 1985;230:1282–1285. 67. Ozer H, Armitage JO, Bennett CL, et al. Update of recommenda-
52. Jaschke E, Zabernigg A, Gattringer C. Recombinant human gran- tions for the use of hematopoietic colony-stimulating factors:
ulocyte-macrophage colony-stimulating factor applied locally in Evidence-based, clinical practice guidelines. J Clin Oncol
low doses enhances healing and prevents recurrence of chronic 2000;18:3558–85.
venous ulcers. Int J Dermatol 1999;38:380–386. 68. Peters WP, Rosner G, Ross M, et al. Comparative effects of granu-
53. Kaplan LD, Kahn JO, Crowe S, et al. Clinical and virologic effects locyte-macrophage colony-stimulating factor (GM-CSF) and
of recombinant human granulocyte-macrophage colony-stimulat- granulocyte colony-stimulating factor (G-CSF) on priming periph-
ing factor in patients receiving chemotherapy for human immuno- eral blood progenitor cells for use with autologous bone marrow
deficiency virus-associated non-Hodgkin’s lymphoma: results of a after high-dose chemotherapy. Blood 1993;81:1709–1719.
randomized trial. J Clin Oncol 1999;9:929–940. 69. Peters WP, Stuart A, Affronn ML, et al. Neutrophil migration is
54. Kurzrock R, Talpaz M, Gomez JA, et al. Differential dose-related defective during recombinant human granulocyte-macrophage
haematological effects of GM-CSF in pancytopenia: evidence sup- colony-stimulating factor infusion after autologous bone marrow
porting the advantage of low-over high-dose administration in transplantation in humans. Blood 1988;72:1310–1315.
selected patients. Br J Haematol 1991;78:352–358. 70. Pettenati MJ, Le Beau MM, Lemons RS, et al. Assignment of
55. Lazarus HM, Andersen J, Chen MG, et al. Recombinant granulo- CSF-1 to 5q33.1: Evidence for the clustering of genes regulating
cyte-macrophage colony-stimulating factor after autologous bone hematopoiesis and for their involvement in the deletion of the long
marrow transplantation for relapsed non-Hodgkin’s lymphoma: arm of chromosome 5 in myeloid disorders. Proc Natl Acad Sci
Blood and bone marrow progenitor growth studies. A phase II USA 1987;84:2970–2974.
Eastern Cooperative Oncology Group trial. Blood 1991;78:830. 71. Quelle FW, Sato N, Witthuhn BA, et al. JAK2 associates with the
56. Le Beau MM, Epstein ND, O’Brien SJ, et al. The interleukin 3 beta c chain of the receptor for granulocyte-macrophage colony-
gene is located on human chromosome 5 and is deleted in myeloid stimulating factor, and its activation requires the membrane-prox-
leukemias with a deletion of 5q. Proc Natl Acad Sci USA 1987;84: imal region. Mol Cell Biol 1994;14:4335–4341.
5913–5917. 72. Ross AA, Cooper BW, Lazarus HM, et al. Detection and viability
57. Leong SP, Enders-Zohr P, Zhou YM, et al. Recombinant human of tumor cells in peripheral blood stem cell collections from breast
granulocyte macrophage-colony stimulating factor (rhGM-CSF) and cancer patients using immunocytochemical and clonogenic assay
autologous melanoma vaccine mediate tumor regression in patients techniques. Blood 1993;82:2605–2610.
with metastatic melanoma. J Immunother 1999;22:166–174. 73. Santoli D, Clark SC, Kreider BL, Maslin PA, Rovera G.
58. Lieschke GJ, Maher D, Cebon J, et al. Effects of bacterially syn- Amplification of IL-2-driven T cell proliferative by recombinant
thesized recombinant human granulocyte macrophage colony- human IL-3 and granulocyte-macrophage colony-stimulating fac-
stimulating factor in patients with advanced malignancy. Ann Int tor. J Immunol 1988;141:519–526.
Med 1989;110:357–364. 74. Sato N, Sakamaki K, Terada N, Arai K, Miyajimi A. Signal trans-
59. Lieschke GJ, Maher D, O’Connor M, et al. Phase I study of intra- duction by the high-affinity GM-CSF receptor: two distinct cyto-
venously administered bacterially synthesized granulocyte-mac- plasmic regions of the common beta subunit responsible for
rophage colony-stimulating factor and comparison with different signaling. EMBO J 1993;12:4181–4189.
subcutaneous administration. Cancer Res 1990;50:606–614. 75. Schaison G, Eden OB, Henze G, et al. Recommendations on the
60. Lopez AF, Williamson J, Gamble JR, et al. Recombinant human use of colony-stimulating factors in children: conclusions of a
granulocyte-macrophage colony-stimulating factor stimulates in vitro European panel. Eur J Pediatr 1998;157:955–966.
mature human neutrophil and eosinophil function, surface receptor 76. Shea TC, Mason JR, Storniolo AM, et al. Sequential cycles of
expression and survival. J Clin Invest 1986;78:1220–1228. high-dose carboplatin administered with recombinant human
61. Metcalf D, Begley CG, Johnson GR, et al. Biologic properties in granulocyte-macrophage colony stimulating factor and repeated
vitro of a recombinant human granulocyte-macrophage colony infusions of autologous peripheral-blood progenitor cells: A novel and
stimulating factor. Blood 1986;67:37–45. effective method for delivering multiple courses of dose-intensive
62. Miyauchi J, Kelleher CA, Yang YC, et al. The effects of three therapy. J Clin Oncol 1992;10:464–473.
recombinant growth factors, IL-3, GM-CSF, and G-CSF, on the 77. Sieff C, Emerson SG, Donahue RE, et al. Human recombinant
blast cells of acute myeloblastic leukemia maintained in short-term granulocyte-macrophage colony-stimulating factor: a multi-lineage
suspension culture. Blood 1987;70:657–663. hematopoietin. Science 1985;230:1171–1173.
63. Mudzinski SP, Christian TP, Guo TL, Cirenza E, Hazlett KR, 78. Siena S, Bregni M, Brando B, et al. Circulation of CD34+
Gosselin EJ. Expression of HLA-DR (major histocompatibility hematopoietic stem cells in the peripheral blood of high-dose
complex class II) on neutrophils from patients treated with granu- cyclophosphamide-treated patients: Enhancement by intravenous
locyte-macrophage colony-stimulating factor for mobilization of recombinant human granulocyte-macrophage colony-stimulating
stem cells. Blood 1995;86:2452–2453. factor. Blood 1989;74:1905–1914.
588 Granulocyte-macrophage colony-stimulating factor

79. Silberstein DS, Owen WF, Gasson JC, et al. Enhancement of human 86. Vellenga E, Young DC, Wagner K, Wiper D, Ostapovicz D, Griffin
eosinophil cytotoxicity and leukotriene synthesis by biosynthetic JD. The effects of GM-CSF and G-CSF in promoting growth of
(recombinant) granulocyte-macrophage colony-stimulating factor. clonogenic cells in acute myeloblastic leukemia. Blood
J Immunol 1986;137:3290–3294. 1987;69:1771–1776.
80. Socinski MA, Cannistra SA, Sullivan R, et al. Human granulocyte- 87. Weisbart RH, Golde DW, Clark SC, et al. Human granulocyte-
macrophage colony stimulating factor induces expression of the macrophage colony-stimulating factor is a neutrophil activator.
CD11 b surface adhesion molecule on granulocytes in vivo. Blood Nature 1985;314:361–363.
1988;72:691–697. 88. Weisbart RH, Golde DW, Gasson JC. Biosynthetic human
81. Soiffer R, Lynch T, Mihm M, et al. Vaccination with irradiated GM-CSF modulates the number and affinity of neutrophil. F-met-
autologous melanoma cells engineered to secrete human granulocyte- leu-phe receptors. J Immunol 1986;137:3584–3587.
macrophage colony-stimulating factor generates potent antitumor 89. Weiser WY, Van Niel A, Clark SC, et al. Recombinant human
immunity in patients with metastatic melanoma. Proc Natl Acad granulocyte-macrophage colony-stimulating factor activates intra-
Sci USA 1998;95:13141–13146. cellular killing of Leishmania donovani by human monocyte-
82. Steward WP, Scarffe JH, Dirix LY, et al. Granulocyte-macrophage derived macrophage. J Exp Med 1987;166:1436–1446.
colony stimulating factor (GM-CSF) after high-dose melphalan in 90. Welte K, Zeidle C, Reiter A, et al. Differential effects of granulo-
patients with advanced colon cancer. Br J Cancer 1990;61: cyte-macrophage colony stimulating factor in children with severe
749–754. congenital neutropenia. Blood 1990;75:1056–1063.
83. Sutherland GR, Baker E, Callen DF, et al. Interleukin-5 is at 5q31 91. Williams MA, Kelsey SM, Collins PW, Gutteridge CN, Newland AC.
and is deleted in the 5q- syndrome. Blood 1988;71:1150–1152. Administration of rHuGM-CSF activates monocyte reactive oxygen
84. Thompson JA, Lee DJ, Kidd P, et al. Subcutaneous granulocyte- species secretion and adhesion molecule expression in vivo in patients
macrophage colony-stimulating factor in patients with myelodys- following high-dose chemotherapy. Br J Haematol 1995;90:31–40.
plastic syndromes: Toxicity, pharmacokinetics, and hematologic 92. Wong GG, Witek JS, Temple PA, et al. Human GM-CSF molecular
effects. J Clin Oncol 1989;7:629–637. cloning of the complementary DNA and purification of the natural
85. Vadhan-Raj S, Keating M, LeMaistre A, et al. Effects of recombi- and recombinant proteins. Science 1985;228:810–815.
nant human granulocyte-macrophage colony-stimulating factor in 93. Yarden Y, Escobedo JA, Kuang WJ. Structure of the receptor for
patients with myelodysplastic syndromes. N Engl J Med 1988;317: platelet-derived growth factor helps define a family of closely
1545–1551. related growth factor receptors. Nature 1986;323:226–232.
19 Cancer gene therapy
DONALD J. BUCHSBAUM, C. RYAN MILLER, LACEY R. MCNALLY,
AND SERGEY A. KALIBEROV

gene therapy strategies for the treatment of cancer are


Introduction summarized in Table 1. However, insufficient informa-
Cancer is the product of a multi-step process involving tion is available to evaluate the therapeutic efficacy of
an accumulation of genetic alterations in somatic cells these strategies in most clinical trials. The first clinical
[203]. Advances in the understanding of the molecular gene transfer trial involved the transfer of gene-marked
basis of neoplasia and host–tumor relationships have tumor-infiltrating lymphocytes into patients with
resulted in new cancer therapy strategies with transla- advanced cancer. There are now approximately 190
tion into clinical trials. Gene therapy has emerged as a active gene therapy protocols which involve thousands
treatment strategy which relies on the introduction of of cancer patients [39]. About 50% have been designed
genetic material to cure, slow the progression, or possi- to augment an immune response against cancers by vac-
bly even prevent a variety of diseases including genetic, cine or direct cytokine gene transduction and about 20
infectious (HIV in particular), cardiovascular, and protocols involve a drug-sensitization strategy. Approved
arthritic diseases [11, 66]. Gene therapy has shown protocols exist for overcoming multi-drug resistance in
potential for the treatment of cancer. A number of can- breast and ovarian cancer patients receiving myelosup-
cer gene therapy approaches have been developed based pressive chemotherapy.
on direct correction of genetic lesions. These “mutation
compensation” approaches, designed to correct the
molecular lesions underlying neoplastic transformation
and progression, have demonstrated efficacy in the con-
Gene Transfer Vectors
text of in vitro and preclinical model systems. Although Vectors are vehicles to carry the genetic material. The
many therapeutic genes exist an efficient, non-toxic vectors for modification of tumor cells or normal tissues
gene delivery system is not currently available. The effi- should allow definitive therapeutic or preventive inter-
ciency and accuracy of gene delivery remain the most ventions. To achieve this end, there is a need for gene
significant barriers to the success of cancer gene therapy delivery vectors capable of efficient and selective gene
[200]. However, preclinical studies have demonstrated transfer to tumor cells in vivo. One of the major prob-
approaches that may have promise in the clinical set- lems has been a low level of gene transfer in human gene
ting. The approaches that have been explored include therapy clinical trials. Recent advances in the develop-
genetic immunopotentiation with cytokine gene transfer ment of vectors for gene transfer make it likely that gene
and tumor cell vaccination, molecular chemotherapy therapy will have an increasing role in the clinical treat-
with prodrug activation by suicide genes or an increase ment of cancer. Vectors are being engineered that will
in tumor cell sensitivity to chemotherapy or radiation target specific tumor cell types. Vector requirements
therapy, protection of bone marrow cells from the toxic depend on the specific approach and disease. Gene transfer
effects of chemotherapeutic drugs, inhibition of acti- vectors are either nonviral or viral [96].
vated oncogenes by antisense treatment, transfer of Viruses are currently the most effective means of
tumor suppressor genes, and pro-apoptotic gene therapy. gene delivery and can be manipulated to express thera-
Each of these approaches have been translated into clin- peutic genes or to replicate specifically in certain cells.
ical cancer therapy trials [32, 66]. Viral vectors used in in vivo pre-clinical studies have
Gene therapy is likely to play a prominent role in the included retrovirus, adenovirus, adeno-associated virus,
treatment of cancer in the future. The purpose of this and lentivirus. Each has limitations, and studies leading
article is to provide an update on research in gene ther- to a better understanding of virus–cell interactions are
apy for the treatment of cancer. Several excellent recent needed to improve vector technology. Some viruses,
reviews of cancer gene therapy have been published such as adenovirus, only result in transient production
[32, 48, 64, 66, 105, 168]. In this paper, current cancer of the therapeutic protein as they do not incorporate the

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 589
© Springer Science + Business Media B.V. 2009
590 Cancer gene therapy

Table 1. Current strategies for cancer gene therapya


Strategy Specific agent Reference
Mutation Compensation
Induction of tumor suppressor genes P53, Rb, BRCA-1 [32, 33, 66, 141, 170]
Pro-apoptotic and cell cycle gene therapy bax, bak, Fas ligand, caspase-9, p21 [88, 101, 148, 149, 211]
Inactivation of oncogenes
Intracellular knockout of growth factor receptors erbB-2 [8, 43, 66]
Antisense k-ras, c-myc, TGFß [66]
Genetic immunopotentiation
Genetic modification of tumor cells cytokines, B7, MHC [41, 50, 66, 201, 219]
Genetic modification of immune effector cells Cytokines [66, 167]
Molecular chemotherapy
Suicide gene therapy HSV-tk, CD [5, 9, 66, 72, 108, 146, 168, 169,
183, 216]
Chemoprotection of bone marrow MDR [36, 126]
Inhibition of angiogenesis flt-1, flk-1, endostatin, angiostatin [13, 52, 104, 174]
Replicative vector oncolysis Adp53, herpes virus [66, 102, 123, 187, 193]
Chemosensitization and radiosensitization bax, bcl-2, erbB-2, p53, cytokines, [14, 30, 31, 42, 47,51, 66, 70, 89, 110,
angiostatin, HSV-tk, CD 117, 143, 151, 154, 169, 180, 181, 216]

a
Reprinted with permission from Mary Ann Liebert, Inc.

Table 2. Characteristics of most commonly used vectorsa


Cell division Duration of
Type Insert size Genome required expression Advantages Disadvantages
Retrovirus 5–7 kb RNA Yes Long-term Potential to integrate into Insertional mutagenesis
genome of target cell
Adenovirus 7–35 kb DNA No Transient Relatively high Local tissue inflammation
transduction efficiency and immune response
into normal and tumor
cells; easy production
and at high titers;
tropism can be modified
Adeno- 2–4 kb DNA No Long-term High transduction Insertional mutagenesis;
associated- efficiency into muscle difficulties with production;
virus and brain do not work in all organs
Herpes simplex Up to 30 kb DNA Yes Transient High transduction Difficult to obtain long-term
virus efficiency gene expression; difficult
to target
Nonviral vectors No limitation RNA or DNA No Transient Repetitive and safe Low efficiency gene transfer
administration feasible

a
Adapted from [66, 105]; Reprinted with permission from Mary Ann Liebert, Inc.

genes into the chromosomes of the target cells [125]. The characteristics of the most commonly used vectors
Other vectors, such as adeno-associated viruses, are used in cancer gene therapy are summarized in Table 2.
limited in the size of therapeutic genes that can be incor-
porated or produce low levels of protein [208]. The
mechanisms by which adeno-associated virus integrates
Retroviral Vectors
into the human genome are unclear, and HIV lentiviral Retroviral vectors have been used for many gene transfer
vectors may have biosafety issues. Major limitations of protocols because of their ability to produce stable, high
different vector systems include biosafety risk, random level gene expression in many cell types as a consequence
integration into the genome, need for cell proliferation, of their integration into the target cell genome. There
potential for immune responses to the vector, lack of a are several limitations of retroviral vectors, including
tissue-specific response, and unknown duration of effect. the size of the inserted gene, low titers, difficulty in
Donald J. Buchsbaum et al. 591

large-scale production, and they can only insert their gene expression [66, 105]. The recognition of CAR
genes into the genome of actively dividing cells. Vectors deficiency in most human carcinomas has resulted in the
derived from lentiviral retroviruses can transduce gene generation of Ad vectors capable of CAR independent
expression in both dividing and nondividing cells [96], gene transfer [36, 66]. Strategies that have been devel-
and have been used for cancer gene therapy in animal oped to achieve this utilize retargeting ligands as well as
models [45]. The issues and assays needed to ensure genetic capsid modifications. This has allowed infection
patient safety with this new vector system are being and gene transfer to otherwise refractory tumor cells,
defined [22, 49, 156]. Efforts are ongoing to increase the and resulted in therapeutic gain in preclinical models.
infectivity of retroviruses through the development of These new vectors are currently being evaluated in
retroviral vectors which display ligands on their surface human clinical trials [107].
for which receptors are present on target cells. Serious Replication-competent adenovirus vectors, which
adverse events in some clinical trials have highlighted cause cytolysis as part of their natural life cycle, repre-
safety concerns when retroviral viral vectors are used for sent an emerging technology that shows considerable
gene transfer [142]. promise as a novel treatment option, particularly for
armed vectors which carry therapeutic genes [135]. The
use of replication competent viruses is an attractive
Adenoviral Vectors strategy for tumor therapy because the virus can repli-
Adenoviral vectors for gene transfer are adenoviruses cate and spread in situ, exhibiting oncolytic activity
(Ad) that have been genetically modified through dele- through a direct cytopathic effect, and produce higher
tions of the viral genome to create space for insertion of levels of therapeutic gene expression achieved as a
a foreign transgene [96]. Both replication-incompetent result of adenovirus replication.
and replicative Ad vectors have been used in clinical
cancer gene therapy trials. There is no risk for insertion
mutagenesis since they do not incorporate into the
Adeno-associated Viruses
genome. An entire issue of Human Gene Therapy was Adeno-associated viruses (AAV) have received consid-
devoted to Ad vector safety and toxicity [134]. Ad vec- erable attention for gene transfer studies and cancer
tors produce higher levels of gene expression and can be gene therapy [95, 197]. AAV are capable of infecting
produced in greater quantities than retroviral vectors both dividing and nondividing cells, producing stable
[96]. Another advantage of Ad vectors is their ability to and efficient integration of viral DNA into the host
infect both dividing and nondividing cancer cells. A dis- genome, and achieving long-term gene expression. Only
advantage is that Ad vectors tend to be recognized as minimal viral gene products are retained in current vec-
foreign and therefore elicit host cellular and neutralizing tors so that they are not very immunogenic and do not
humoral immune responses directed against the viral markedly elicit host cytotoxic T-cell immune responses
capsid. These inflammatory reactions limit the efficacy against vector-transduced cells. AAV vectors may be
of repetitive administrations of Ad vectors and compro- useful for antiangiogenic gene therapy and cytokine
mise the persistence of transduced cells in vivo [48]. gene transfer and in vivo immunization approaches
A solution to overcoming the immune response has been [140, 157]. However, major obstacles in the develop-
to produce Ad vectors deleted of all Ad gene products. ment of effective recombinant AAV mediated gene
Although gene expression is transient following Ad therapy are infection specificity and gene targeting.
infection, since the transferred genes do not integrate
into the genome, this produces a biosafety advantage.
To sustain transgene expression levels, repeat adminis-
Herpes Simplex Virus
trations of the Ad vectors are required. Analysis of a The herpes simplex virus (HSV) has been used to deliver
variety of primary human tumors has demonstrated therapeutic genes to some forms of brain cancer and
deficiencies in the primary receptor for Ad, coxsackie prostate adenocarcinoma [96, 202]. HSV displays a
and Ad receptor (CAR), which explains the lack of gene broad host cell range and its cellular receptors, heparan
transfer in many human clinical trials. sulfate (HS), herpes virus entry mediator (HVEM), and
Modification of Ad vectors may reduce expression of nectin-1 and 2, are widely expressed on the cell surface
endogenous Ad proteins and diminish immune responses of numerous cell types. Herpes viruses have been engi-
to Ad vectors. Second generation Ad vectors have been neered to replicate selectively in tumors but poorly or
developed to reduce the host immune response against not at all in normal tissues. The problems with HSV-
viral proteins and to increase the duration of therapeutic mediated gene transfer include the cytopathic nature of
592 Cancer gene therapy

HSV, the difficulty in maintaining long-term expression DNA is only transiently maintained. This approach has
of inserted genes, and low infection efficiency [105]. also been used for the generation of cancer vaccines.
This suboptimal result may reflect viral gene deletions, Nonviral vectors are attractive with respect to ease of
which can reduce the replicative potential of viruses large-scale production and lack of specific immune
in tumor cells. For example, deletion of the γ34.5 response [111].
gene significantly reduced viral growth even in rapidly
dividing cells [194]. Biosafety concerns of HSV relate
to the fact that wild-type virus is highly pathogenic and
cerebral injection causes fatal encephalitis. Toxic and/or
Targeting of Viral Vectors
pathogenic properties of the virus must, therefore, be Transductional Targeting
disabled prior to its use as a gene delivery vector.
Several studies have shown that viral vectors can be tar-
geted to specific cell types after attachment of ligands
Other Viral Vectors (e.g. transferrin, folate, epidermal growth factor, fibro-
Vaccinia viruses, poxviruses, baculoviruses, and RNA blast growth factor, peptides, and single chain antibod-
replicons derived from poliovirus [12] have also been ies (sFv) that bind to tumor cell surface receptors) to the
investigated for use in the delivery of genes for thera- viral capsid, either by chemical conjugation employing
peutic purposes. A variety of replicative viruses have the Fab fragment of an anti-knob monoclonal antibody,
been employed for gene transfer including parvovirus, or by genetic engineering for transductional regulation
herpes virus, reovirus, and Ad [66]. The antitumoral of vector infection (Fig. 1) [16, 36, 68, 86]. Tropism-
effect is achieved by virtue of the replicative cycle of modified Ad vectors can infect cells that are refractory
the virus to produce oncolysis selectively in tumor cells. to transduction by the native Ad, resulting in enhanced
An attractive aspect of replicative viruses is their ability gene transfer and therapeutic benefit in vivo [162].
to achieve an amplified effect as a result of their capac- Efforts have been made to genetically engineer the viral
ity to spread and infect tumor cells within solid tumors capsid proteins to contain cell-targeting ligands. Our
[7, 37, 65]. Replicative viral vectors have been tested in group and others have modified the carboxy terminus of
human clinical gene therapy trials. Vector modifications the Ad fiber protein to incorporate peptides or growth
to overcome viral receptor deficiency and to achieve factors with specificity for tumor cellular receptors [36,
tumor replicative specificity are being investigated. 66]. Ad vectors which have been engineered to incorpo-
rate a RGD motif at the carboxy terminus or in the HI
loop for binding to tumor cell surface integrins have
Nonviral Vectors shown markedly enhanced transduction of human cancer
Nonviral vectors have been another promising area of cell lines and primary tumor cells, which express low
vector development. Several lipid-, peptide-, and poly- levels of the CAR receptor [36, 65, 94]. Combined target-
mer-based systems are being investigated for gene deliv- ing of Ad to glioma cell surface integrins and epidermal
ery [34, 73, 144]. Liposomes have been used for gene growth factor receptors increased gene transfer into
transfer of foreign MHC genes in a clinical trial. The primary glioma cells [66]. It has been shown that Ad
liposomes can be modified to provide for selectivity in vectors can be targeted to vascular receptors by using
cell targeting. However, the levels of gene expression peptide-based molecular adaptors [196]. Alternatively,
are limited by the liposomal degradation of the internal- the therapeutic gene can be placed under the control of
ized particles. Systemic administration of cationic lipid/ a tissue- or tumor-specific promoter which is activated
plasmid complexes yields predominantly transfection of in tumor cells but not normal cells, and therefore restrict
the lungs, with the major transfected cell type being expression to the tumor cell.
endothelial cells [13]. Improved cationic liposomes
delivered the p53 tumor suppressor gene to primary and
metastatic murine and human lung tumors in mice and
Transcriptional Targeting
this was associated with suppressed tumor growth and Targeted expression of therapeutic genes has been
prolonged animal survival with minimal toxicity [160]. obtained using transcriptional regulatory sequences from
Direct injection of naked DNA into tumors using tumor-specific genes that are ectopically expressed in
mechanical methods has been shown to result in gene cancers, viral genes expressed in virus-associated can-
transfer and expression [105]. A problem is the inability cers, and tissue-specific genes expressed in cancers
to transduce a large number of cells and the fact that the and their tissues of origin [125]. Examples include the
Donald J. Buchsbaum et al. 593

a b c

Anti-fiber
antibody Vector with TSP
(Fab) gene construct

Ligand Peptide Peptide

Primary Cell surface Receptors


Receptors
receptor receptor
(CAR) (CAR)
TSP Gene
Specific Transcriptional
Regulatory Sequence
Tumor Cell Tumor Cell
Gene Expression

Figure 1. Strategies to target Ad vector gene transfer include (a) transductional targeting to modify Ad binding tropism to the
CAR receptor in which a ligand that binds to tumor cell surface receptors is conjugated to a Fab of an anti-knob monoclonal
antibody, (b) transductional targeting by genetic engineering of a peptide with specificity for tumor cellular receptors into the
knob of the Ad fiber protein, and (c) transcriptional targeting using a tumor-specific promoter (TSP) to produce targeted
expression of a therapeutic gene in a tumor cell with specific transcriptional regulatory sequences in combination with genetically
modified Ad. Reprinted with permission from Mary Ann Liebert, Inc.

alpha-fetoprotein promoter for hepatocellular carcinoma,


the hTERT promoter in telemorase positive tumors, the Mutation Compensation
PSA, PSMA, and probasin promoters for prostate can-
cer, and the MUC1 and erbB-2 promoters for breast can-
Induction of Tumor Suppressor Genes
cer [61, 125]. The incorporation of these promoters into The p53 protein is a multifunctional regulator of cell
gene therapy vectors allows selective expression of the growth and plays a critical role in regulating the cell
transduced therapeutic gene. Other examples of induc- cycle. When the p53 gene is mutant or absent, the lack
ible promoters that have been investigated are hypoxia- of control of p21 results in lack of inhibition of cyclin
responsive elements that are induced by hypoxia which dependent kinases and unregulated cell growth. The p53
is present in many solid tumors [82], and radiation protein also plays a central role in the apoptosis path-
responsive promoters that provide a three- to four-fold way. A large proportion of human cancers carry impaired
induction in gene expression in the radiation field [19]. p53 gene function. In certain tumor types, dysfunctional
Regulatory sequences from genes expressed in tumor p53 has been associated with more aggressive tumor
endothelial cells and from cell cycle-regulated genes are growth, enhanced tendency for tumor metastases, and
also candidates for transcriptional targeting [125]. poor survival rates. The approach of p53 gene transfer
For example, we have developed Ad vectors encoding has been used by a number of investigators [141]. When
apoptosis-inducing (bax or TRAIL) genes under control p53 was transfected into tumor cells and overexpressed,
of the VEGF or flt-1 promoter, respectively, and apoptosis and growth arrest has been observed. It was
angiogenesis-suppressing soluble VEGF receptor 2 gene shown that transfecting normal p53 to cells with deleted
under control of the VEGF promoter element. Using VEGF or mutated p53 decreased anchorage independent
and VEGFR1 promoter for targeting of gene expression growth in vitro and tumorigenicity in animals [32].
in VEGF/VEGFR1 positive tumor cells as well as tumor- The approach taken by Clayman et al. [32] to p53
associated blood vessel endothelial cells significantly gene replacement therapy has been to inject head and
increased therapeutic efficacy of gene therapy in human neck squamous cell cancer with an Ad vector encoding
prostate tumor xenograft models [90–92]. wild-type p53. Patients with head and neck cancer have
594 Cancer gene therapy

accessible lesions allowing for direct delivery of vectors. totic and anti-apoptotic proteins is modified in order
Intratumoral injection of Ad containing wild-type p53 to increase apoptosis. Human tumor cells have been
produced growth inhibition of established head and transduced with bax, bak, and Fas ligand genes in Ad vec-
neck squamous cell tumors in animal models [32]. The Ad tors and shown to undergo apoptosis [88, 101, 148, 149].
vector was not toxic to normal squamous oral epithelial In addition, the gene for caspase-9 has been used in an Ad
cells or normal fibroblasts in culture, and minimal toxicity vector to induce apoptosis in human prostate cancer cells
was produced in animal models. Studies performed in and suppress tumor growth in nude mice [211]. A tumor-
mouse models of non-small cell lung cancer also have specific apoptotic effect of TRAIL (tumour necrosis fac-
shown a significant therapeutic effect of Adp53 gene tor-related apoptosis-inducing ligand) has been shown, in
therapy [32]. Studies have shown that vascular endothelial which the TRAIL gene was delivered using Ad or AAV
growth factor (VEGF) expression is reduced in cancer and shown to have significant anti-tumour efficacy in
cells following Adp53 infection, resulting in inhibition animal models of aggressive primary and metastatic
of angiogenesis in vivo [32]. cancer [119]. A limitation of these approaches is that
Phase I clinical trials of Adp53 gene transfer have every cell of the primary tumor and metastases be affected
been completed [32, 33]. The studies demonstrated that by the treatment, unless these approaches are used in
Adp53 gene transfer is safe and well tolerated. Transient combination with other therapeutic modalities.
p53 expression occurred in solid tumors despite high Induction of apoptosis following p21 gene transfer
levels of antibodies to Ad. The most frequently observed which was originally identified as a molecule that regu-
toxicities were fever, chills, and injection site pain [32]. lates transition from the G1 phase to the S phase of the
A Phase II study of 170 patients with recurrent head and cell cycle has been reported [87].
neck squamous cell cancer further established the safety
and lack of toxicity of direct intratumoral Adp53 injections
[32]. A small percentage of patients had stable disease.
Inactivation of Oncogenes
Randomized trials of Adp53 gene therapy in head and Although most oncogenes are silenced after fetal devel-
neck cancer patients are currently ongoing. opment to prevent abnormal tissue growth, cancer cells
Other clinical trials have confirmed the safety and effi- may propagate by activating or amplifying oncogenes.
cacy of retroviral or Ad p53 gene replacement therapy in One form of cancer gene therapy is the targeted disrup-
non-small cell lung cancer, prostate cancer, recurrent tion of tumor oncogenes by: (1) inhibition of oncogene
ovarian carcinoma, and esophageal cancer [32, 170, transcription; (2) reduction of mRNA translation into
177]. The results of a recent Phase II study indicated that protein; and (3) interference with protein function.
intratumoral Ad p53 provided no additional benefits in Inhibition of erbB-2, and blockade of k-ras, c-myc,
patients receiving first-line chemotherapy for advanced c-fos, TGFß and insulin-like growth factor 1 are
non-small cell lung cancer [176]. In addition, other approaches that are being investigated clinically [66].
replacement strategies that are being investigated clini- Transcription of dominant oncogenes has been inhibited
cally include induction of Rb and BRCA-1 gene expres- using triplex-forming oligonucleotides. Ad gene E1A
sion [65]. Preclinical studies showed expression of the that inhibits transcription of the human c-erbB-2 pro-
wild-type Rb and BRCA-1 proteins after gene transfer moter suppressed tumorigenicity and metastatic poten-
and reversion of the malignant phenotype, often associ- tial induced by the erbB-2 oncogene. Translation of
ated with the induction of apoptosis in tumor cells [141]. oncogene messenger RNA has been blocked using spe-
Another tumor suppressor gene that has been investi- cific antisense sequences [66]. These include antisense
gated is PTEN [129]. Multiple gene replacements have treatment against k-ras in lung cancer, c-myc in breast
been examined as a potential treatment of cancer and and prostate cancer, and TGFß in glioma. The erbB-2
have generally resulted in additive or synergistic effects. oncoprotein has been inhibited with the use of intracel-
Combination delivery of both p16 and p53 into cancer lular antibodies. In this regard, Deshane et al. [43] from
cells by adenovirus resulted in an additive or synergistic our group showed that intracellular expression of an anti
apoptotic effect for treatment of cancer [100, 173]. erbB-2 sFv following Ad gene transfer resulted in down-
regulation of cell surface erbB-2 expression, and cyto-
Pro-apoptotic and Cell Cycle Gene toxicity in human ovarian cancer cells both in vitro and
in vivo in an animal model. This approach was translated
Therapy into a clinical Phase I trial in patients with ovarian cancer
Another approach that is receiving attention is pro-apop- [8]. Of the 13 patients evaluable for response, 5 (38%)
totic cancer gene therapy in which the ratio of pro-apop- had stable disease and 8 (62%) had progressive disease.
Donald J. Buchsbaum et al. 595

Genetic Immunopotentiation the level of gene transfer and avoiding toxicity com-
pared to in vivo gene transfer. However, the level of gene
Genetic immunopotentiation strategies are designed to transfer into tumor and immune effector cells in clinical
achieve active immunization against tumor associated trials has so far been limited [66].
antigens by gene modification of tumor cells to enhance
their immunogenicity, or enhance the anti-tumor activity
of immune system cells [53]. Genetic immunopotenta-
Genetic Modification of Immune
tion is most utilized for the treatment of melanoma. Effector Cells
Tumor vaccines are used to initiate an immune response Cells of the immune system including lymphocytes, NK
against an unrecognized or poorly antigenic tumor. cells, and dendritic cells have been genetically modified
Treatment with unmethylated cytosine–phosphate–gua- to augment their capacity to kill tumors [66]. Tumor
nine (CpG) activates plasmacytoid dendritic cells, thus infiltrating lymphocytes were the first immune cells to
releasing IFN alpha and boosting T-cell and natural killer be genetically transduced and used in a human gene
cell responses as well as activating conventional myeloid therapy trial against melanoma [167]. Although attempts
dendritic cells to treat early stage melanoma [131]. have been made to modify their binding tropism [66],
Alternatively, metastatic melanoma treatment with retro- the modest localization and toxicity of these lympho-
viral vectors armed with IFN-gamma is currently in cytes remains a limitation for this form of therapy. Using
phase I clinical trials as a tumor cell vaccine [60]. dendritic cell vaccines requires the cells to be loaded
with antigen for presentation to elicit an immune
Genetic Modification of Tumor Cells response to tumor [175]. The first reported dendritic cell
vaccination study was for indolent lymphoma with other
It has been hypothesized that vaccination against the trials for lymphoma [78]. In the case of glioma, tumors
tumor by exposing tumor antigens to the immune sys- have been targeted by bone marrow-derived dendritic
tem in a more favorable context will result in tumor cells loaded with glioma cell mRNA resulting in a spe-
rejection [66]. A major focus of research is the identifi- cific T cell response to protect against intracerebrally
cation of new tumor antigens and the development of implanted glioma tumor cells [114]. Numerous studies
cancer vaccines and new anticancer drugs. The first continue to evaluate the effectiveness of dendritic cell
clinical trials with cDNA vaccination with carcinoem- vaccines in preventing tumor relapses and extending
bryonic antigen demonstrated limited benefit. Cytokine patients’ survival [218].
or co-stimulatory molecule B7 gene modified tumor
cells and defined tumor antigens have also been used.
Numerous studies have shown that tumor cells can be
transfected with various cytokine genes and become tar- Molecular Chemotherapy
gets for specific immune rejection [201]. Transduction Evolution of Molecular Chemotherapy
of a cytokine gene into tumor cells elicits an inflamma-
tory host reaction that impairs tumor growth [124, 207].
Paradigm
This approach has been used in clinical trials [63, 201]. The narrow therapeutic index of drug toxicity to tumor
Several investigators have shown that exposure to tumor versus normal tissues, has significantly limited conven-
cells induced to express B7 can be highly effective at tional systemic chemotherapy and necessitated further
enabling normal animals to reject subsequent challenge drug development research aimed at designing more
by tumor cells, but this approach has been much less selective chemotherapeutic agents.
effective at enabling tumor-bearing animals to kill their Because systemic toxicity is the main limitation to
pre-existing tumor [50]. In a melanoma trial with autol- cytotoxic chemotherapy, one approach towards increasing
ogous irradiated tumor cells transduced with GM-CSF, its therapeutic index is through research on alternative
20% of patients who received three vaccinations were forms of drug delivery. One promising alternative
alive after a follow-up of 3–5 years [41]. Live attenuated employs gene transfer to engineer either (1) increased
viruses have been used in vaccines. Cationic liposomes tumor cell sensitivity or (2) decreased normal tissue
that encapsulate DNA expression plasmids which sensitivity to systemically administered cytotoxic agents.
encode allogeneic MHC molecules have been used to This approach, which has been termed molecular chemo-
generate tumor vaccines in situ [219]. In these approaches therapy, gene-directed enzyme-prodrug therapy (GDEPT),
the genetic modification of tumor cells and effector or suicide gene therapy involves delivery of a specific
immune cells can be performed ex vivo, thus enhancing enzyme that can produce cell death through the conversion
596 Cancer gene therapy

of an inactive non-toxic prodrug into a cytotoxic drug randomized controlled trial, the most comprehensive
metabolite. Specifically targeted expression of the prod- cancer gene therapy clinical trial to date, published by
rug-activating enzyme avoids systemic toxicity, and the GLI-328 International Study Group in November
results in high drug concentration in the tumor mass and 2000 [159]. This study assessed the efficacy of standard
an improved therapeutic index compared to systemic therapy, consisting of surgery plus fractionated external
drug administration. The key element of a GDEPT therapy beam radiotherapy, versus standard therapy plus adju-
system is a gene that encodes an enzyme, which con- vant, intratumoral HSV-tk retroviral gene therapy at the
verts a prodrug to an active cytotoxic drug. Importantly, time of surgery in 248 patients with previously untreated
prodrug-activating enzymes are normally absent or (grade IV) glioblastoma multiforme (GBM). No signifi-
poorly expressed in mammalian cells. This means tumor- cant differences were found between the two treatment
targeting of gene therapy, using specific delivery vehicles, arms in either overall median survival (354 versus 365
restricts enzyme expression to the transduced tumor cells days, respectively, p > 0.05) or progression-free survival
and adjacent surrounding tumor cells through diffusion (183 versus 180 days, respectively, p > 0.05). At autopsy
of the drug metabolite to generate a bystander effect. or post-therapy biopsy, 7 of 17 (41%) tumors and 1 of
13 (8%) normal brains tested positive for retroviral vec-
tor DNA by PCR. Thus, anecdotes and case reports of
From Concept to Clinical Trials response notwithstanding [198], 20 years of preclinical
A number of enzyme-prodrug (EP) systems have been and clinical testing of retroviral vector-mediated, HSV-
investigated for cancer gene therapy over the last 20 tk-based cancer gene therapy failed to achieve signifi-
years. Of the nine classes of traditional chemotherapeutic cant clinical benefit in patients with GBM.
agents, alkylating agents, anthracyclines, antibiotics, As of April 2008, 121 clinical trials of enzyme-prod-
antimetabolites, camptothecins, platinum agents, podo- rug cancer gene therapy had been initiated in 14 differ-
phyllotoxins, taxanes, and vinca alkaloids, EP systems ent countries for over 20 different tumor types [1].
have been described for five. A comprehensive overview Despite the major setbacks to the field in the early 2000s,
of these systems and their development in cell culture and the number of new enzyme-prodrug clinical trials
animal model systems was outlined in the previous edition worldwide continues to grow at approximately the same
of this text [21]. Here, we will focus on the clinical devel- rate (six new trials per year). The majority (77 of 90,
opment of these approaches over the past 5 years. 86%) have utilized either retroviruses or adenoviruses
One of the most widely studied suicide gene-prodrug to effect gene delivery. Although retroviruses remain the
systems for cancer gene therapy utilizes the herpes single most common gene transfer vector employed
simplex virus-thymidine kinase (HSV-tk) in combina- worldwide (Fig. 2a), its use has plateaued in the United
tion with anti-herpetic drugs such as a 9-[2-hydroxy-1- States since 1996 (Fig. 2b) and no new enzyme-prodrug
(hydroxymethyl)-ethoxy]-methylguanine (ganciclovir; trials for cancer have been presented at the NIH
GCV) or its analogues (acyclovir and valacyclovir). The Recombinant DNA Advisory Committee (RAC) since
HSV-tk/GCV system is characterized by the effective 2000 [2]. Problems with retrovirus vectors are well-
phosphorylation of GCV into a toxic compound that is established [190] and alternative vectors, including rep-
incorporated into DNA during its replication. This incorpo- lication-competent adenovirus and herpes simplex
ration into guanine sites in newly synthesized DNA chains viruses (HSV) are becoming increasingly employed.
causes termination of synthesis and the selective killing HSV-tk/GCV remains the most actively investigated
of dividing cells by activation of apoptosis pathways. enzyme-prodrug combination, but alternative systems,
The first report of GDEPT for cancer appeared in the including cytosine deaminase (CD) and the combina-
literature in 1986 and involved HSV-tk and the prodrug tion of HSV-tk and CD either as separate or fused trans-
GCV [132]. HSV-tk GDEPT was tested in the first clini- genes, are becoming more frequent, both in the US and
cal gene therapy trial for cancer in humans, which began worldwide (Fig. 3). CD/5-fluorocytosine (CD/5-FC)
patient accrual in 1993 [145] and paved the way for a was proposed as an alternative enzyme-prodrug system
series of similar clinical trials involving retrovirus- in 1992, shortly after the initiation of phase I clinical
mediated delivery of HSV-tk. Initial studies in animal trials with HSV-tk/GCV [133]. In contrast to GCV,
models suggested that the main limitation to this strategy 5-FC, a non-toxic antifungal agent utilized clinically for
was the limited HSV-tk gene transfer efficiency to tumor CNS mycoses, produces a well-characterized, highly
cells mediated by murine fibroblast-based retroviral vector effective chemotherapeutic agent, 5-FU, upon intratumoral
producer cells. These findings were later confirmed in a conversion by CD. Moreover, 5-FU is freely diffusible
large, multinational phase III prospective, open-label, and, unlike GCV, does not require gap junction-mediated
Donald J. Buchsbaum et al. 597

Figure 2. Enzyme-prodrug cancer gene therapy clinical trials worldwide (a) and in the United States (b). The cumulative
number of clinical trials were determined from the gene medicine [1] and NIH Recombinant DNA Advisory Committee (RAC)
databases [2] and stratified by gene transfer vector employed. Abbreviations: Ad, adenovirus; HSV, herpes simplex virus
598 Cancer gene therapy

Figure 3. Enzyme-prodrug cancer gene therapy clinical trials worldwide (a) and in the United States (b). The cumulative number
of clinical trials were determined from the gene medicine [1] and NIH Recombinant DNA Advisory Committee (RAC) databases
[2] and stratified by transgene delivered. Abbreviations: CD, cytosine deaminase; CYP1B1, cytochrome P450 1B1; HSV, herpes
simplex virus; HSV-tk*, genetically modified HSV-thymidine kinase transgene or co-delivery of HSV-tk and other transgenes;
TK, thymidine kinase
Donald J. Buchsbaum et al. 599

intracellular communication to elicit a potent bystander bearing orthotopic, intracranial xenografts of human
effect [67, 179]. glioblastomas after 5-FC therapy and efficacy was further
5-FU is also a well-established, clinically useful radi- potentiated by concomitant fractionated external-beam
osensitizing agent commonly employed for chemoradi- XRT [93].
ation of gastrointestinal (GI) malignancies [189]. We Co-delivery of cDNAs encoding multiple prodrug
have previously demonstrated that adenovirus (Ad)- metabolizing enzymes, either as separate or fused coding
mediated expression of E. coli CD (AdCMVCD), sequences, is another promising approach first described
together with 5-FC, a combination termed virus-directed for the two most common EP systems, HSV-tk/GCV and
EP therapy (VDEPT), and external beam radiation CD/5-FC. The CD-TK fusion gene proved to be synergis-
(XRT) significantly enhanced in vitro cytotoxicity and tic when co-expressed in tumor cells from two separate
in vivo tumor growth control in both GI [151, 152] and coding sequences [6]. Freytag and colleagues extended
non-GI [128] tumors relative to those receiving this approach by combining enzyme-prodrug and onco-
AdCMVCD/5-FC alone. Moreover, incorporation of lytic viral therapy through use of a CD-TK fusion con-
AdCMVCD/5-FC into clinically appropriate fraction- struct delivered by a replication-competent adenovirus
ated XRT schemes significantly enhanced local tumor (Ad5-CD/TKrep) [166]. Freytag, Kim, and colleagues at
growth control relative to single high-dose XRT [181]. Henry Ford Hospital have initiated three phase I clinical
One promising approach towards generating more trials with Ad5-CD/TKrep in patients with prostate carci-
potent enzymes for EP gene therapy involves utilizing noma, either alone [54], with concomitant conformal
either homologous enzymes from different microbial XRT [59], or with intensity-modulated radiation therapy
species or mutated endogenous enzymes that possess (IMRT) [54]. CD-TK gene therapy proved to be safe in
improved kinetics, facilitating enhanced prodrug con- all three trials. Although the first trial was a phase I dose-
version. For example, CD/5-FC EP systems have been escalation trial and was not powered to determine effi-
described using CD from at least three different micro- cacy, preliminary analyses of 5-year follow-up data from
organisms: the bacterium E. coli [133], the yeast 16 patients treated with direct in situ injection of 1010–
Saccharomyces cerevisiae [98] and Candida albicans [10]. 1012 Ad5-CD/TKrep particles and a 1 week course of
Lawrence and colleagues demonstrated the catalytic GCV/5-FC showed an increase in the prognostically
superiority of recombinant CD derived from the yeast meaningful surrogate endpoint of mean prostate-specific
Saccharomyces cerevisiae (yCD) relative to E. coli CD antigen (PSA) doubling time (PSADT) [59]. The authors
(bCD) [98] and that the favorable enzyme kinetics of demonstrated a significant increase in PSADT from 16 to
yCD translate into (1) enhanced tumor regression after 31 months. Androgen suppression therapy (AST) is a pal-
systemic 5-FC [98], (2) 19F-NMR-based detection of liative approach for patients with treatment-refractory
CD activity [182], and (3) radiosensitization [99]. prostate carcinoma and is associated with significant
We have recently described benefits of the second patient morbidity. The authors demonstrated that the pro-
approach, specifically mutation of E. coli CD [93]. jected onset of AST in the treated cohort of patients was
Substitution of alanine (A) for aspartic acid (D) at posi- delayed by 2 years [59]. The exact mechanisms of the
tion 314 of CD increased the relative specificity of the apparent therapeutic effect will require additional clinical
mutant CD-D314A enzyme to 5-FC in comparison with trials and follow-up correlative molecular studies.
wild-type CD [121] and increased prodrug conversion However, at a time of decreased pharmaceutical company
and cytotoxicity to human glioma cells in vitro [93]. We interest in enzyme-prodrug gene therapy and tight fund-
constructed the AdbCD-D314A vector, encoding mutant ing of biomedical research, these results have been hailed
bCD-D314A gene and investigated mutant bCD gene in the gene therapy and urological communities [191] as
transfer in an Ad-directed molecular chemotherapy evidence that this line of research merits continued finan-
approach for treatment of human glioma xenografts. cial support and multi-institutional testing of this approach
This study demonstrated that AdbCD-D314A infection in phase II clinical trials.
resulted in increased 5-FC-mediated cell killing, compared
with AdbCDwt. Therapy with a replication-incompetent
adenovirus encoding CD-D314A with 5-FC and XRT
showed significant reduction in clonogenic glioma
Inhibition of Angiogenesis
cell outgrowth in vitro and subcutaneous xenograft Extensive preclinical and clinical data support the con-
growth in vivo compared to wild-type CD/5-FC/XRT. cept that tumor growth is dependent on angiogenesis
Finally, increased 5-FC conversion efficiency of this and that VEGF plays a central role in this process.
CD-D314A system led to improved survival of mice Inhibition of angiogenesis is one of the major targets for
600 Cancer gene therapy

anti-cancer gene therapy strategies [52]. An advantage Several immuno-cytokines and or chemokines have
of the anti-angiogenic approach is the highly amplified been employed in anti-angiogenic gene therapy strategies.
death of a large number of tumor cells when deprived of Interferon alpha (IFN-alpha) gene delivery by electropo-
their vasculature, which can partially overcome current ration produced regression of squamous cell carcinoma
limitations in the number of tumor cells modified by (SCVII) tumors in 50% of the mice and increased sur-
gene transfer in vivo, as well, it offers an alternative vival time more than two fold [112]. Similarly, inhibition
means of tackling multidrug-resistant tumors that have of tumorigenicity and metastasis of human bladder car-
proved intractable to conventional chemotherapies cinoma in athymic mice by Ad-mediated IFN-beta gene
because unlike cancer cells, endothelial cells are stable therapy is also attributed to the inhibition of angiogenesis
and do not mutate [20]. An additional advantage is the [85]. IFN-gamma gene therapy employing retrovirus
direct access of vectors delivered systemically to vascu- completely eradicated intracranial C6 glioma tumors in
lar endothelial cells. Both suppression of cellular angio- an immunocompetent mouse model [171]. The IFN-
genic signals and augmentation of inhibitors of gamma mediated tumoricidal activity is due to an appar-
angiogenesis have proven to be feasible strategies in ent interplay of B and T cell components of the immune
preclinical tumor models [104]. It has been shown that system, as well as the inhibition of tumor angiogenesis.
Ad-mediated anti-VEGF therapy using a gene encoding Further, Ad-mediated gene therapy of proliferin-related
a soluble VEGFR1/flt-1, a naturally encoded, alterna- protein (PRP), a strong inhibitor of endothelial cell
tively spliced form of flt-1 VEGF receptor, can be used migration or interferon inducible protein (IP10), a sup-
to control tumor growth [52, 122]. Treatment with pressor of capillary tube formation, significantly sup-
AdVEGF-sKDR, encoding a soluble VEGF receptor 2 pressed murine melanoma tumor growth [163]. A recent
under control of the VEGF promoter, significantly study also reported that gene transfer of human IP10
inhibited the proliferation and migration of human vas- using replication-competent retroviral (RCR) vectors
cular endothelial cells and prostate cancer cells. markedly reduced growth in xenograft and syngeneic
AdVEGF-sKDR also sensitized cancer cells to ionizing mouse models associated with a marked reduction in
radiation. In vivo tumor therapy studies demonstrated microvessel density [185].
significant inhibition of tumor growth in mice that Interleukin-8 (IL-8) is a mediator of angiogenesis.
received combined AdVEGF-sKDR infection and radi- Based on this mechanism, an Ad-mediated antisense
ation versus AdVEGF-sKDR alone or radiation therapy IL-8 gene therapy significantly inhibited human bladder
alone [92]. Similarly, suppression of other angiogenic carcinoma in athymic nude mice [84]. Similarly, treat-
factors such as basic fibroblast growth factor (bFGF) by ment of subcutaneous and intracranially established rat
Ad mediated expression of antisense basic fibroblast C6 cell glioma by retroviral delivery of interleukin-4
growth factor (bFGF-AS) resulted in significant inhibi- (IL-4) in situ, resulted in tumor growth arrest and was
tion of transitional cell carcinoma growth [83]. RNasin, associated with eosinophil infiltration and inhibition of
the placental ribonuclease inhibitor, is known to have angiogenesis [172].
anti-angiogenic activity through the inhibition of angio- Matrix metalloproteinases (MMPs) play a critical role
genin and bFGF. A plasmid-mediated gene therapy in degradation of endothelial basement membrane which
approach employing Rnasin in B16 murine melanoma are required to initiate angiogenesis, thus promoting
cell lines significantly inhibited angiogenesis and tumor angiogenesis and cancer progression. TIMP-2 is a natural
metastatic progression [18]. MMP inhibitor. Ad-mediated TIMP-2 gene therapy
Angiopoietins regulate blood vessel assembly and significantly reduced tumor growth rates by 60–80%,
mediate their activity through the receptor tyrosine angiogenesis by 25–75%, and increased survival by 90%
kinases Tie-1 and Tie-2. Administration of an Ad vector in murine lung, colon, and human breast cancer models
expressing a soluble Tie-2 receptor (AdExTek) an in mice [108]. TIMP-1 gene transfer delivered by an AAV
endothelium-specific receptor tyrosine kinase, which is vector inhibited angiogenesis via reduced endothelial
capable of blocking Tie-2 activation, significantly inhib- cell migration and invasion in vitro and inhibited tumor
ited the growth rate of tumors in mice with two different growth in a murine xenotransplant model [222].
well established primary tumors, a murine mammary Treatment with this low dose of AdhTIMP-2 produced a
carcinoma (4T1) or a murine melanoma (B16F10.9), synergistic effect in combination with TNF/IFN-gamma
64% and 47%, respectively. It has also been shown that using the murine B16BL6 melanoma model [199].
administration of AdExTek therapy inhibited tumor Angiogenesis is regulated by several angiogenic
metastasis when delivered at the time of surgical exci- agents and at multiple levels [103]. The anti-angiogenic
sion of primary tumors [116]. molecules function by different mechanisms, including
Donald J. Buchsbaum et al. 601

endothelial cell proliferation, migration, protease activity, primary tumor growth and development of metastases in
and tubule formation. Therefore, it can be inhibited by the lungs of mice [13]. Further, retroviral-mediated
different methods either by a combination of angiogenic delivery of angiostatin and endostatin in both murine
inhibitors or combination of tumor suppressor agents leukemia and melanoma models produced enhanced
and angiogenic inhibitors, and/or combination of immu- antitumor efficacy [174]. Also, Ad-mediated murine
notherapy and angiogenesis inhibitors. For example, ret- endostatin gene therapy prolonged survival and in 25%
roviral mediated angiostatin and endostatin combination of the mice completely prevented tumor growth in a pro-
gene therapy showed synergistic anti-tumor activity and phylactic human colon/liver metastasis xenograft murine
survival in murine leukemia and melanoma models with model [25]. However, high dose intravenous delivery of
complete loss of tumorigenicity in 40% of animals in the Ad-mediated human endostatin was associated with
leukemia model [174]. Thrombospondin I has been severe acute toxicity in mice that included loss of weight,
shown to inhibit angiogenesis. However, a synergistic bleeding, death of animals [204].
inhibition of MDA-MB-435 breast cancer tumor growth Although there is convincing proof of concept in ani-
has been shown with a combination gene therapy of lipo- mal models that an anti-angiogenesis gene therapy
somes complexed with p53 and thrombospondin I encod- approach can be used to treat cancer, realization of its
ing plasmids BAP-TSPf and BAP-p53 when compared full potential for the treatment of a broad array of dis-
to either BAP-p53 or BAP-TSPf alone [214]. eases will require several challenging technical hurdles
Antigen specific cancer immunotherapy and anti- such as duration of expression, induction of immune
angiogenesis have emerged as two attractive strategies response, cytotoxicity of the vectors and tissue specific-
for cancer treatment. An innovative approach that com- ity to be overcome and safety concerns to be alleviated
bines both mechanisms which has potent anti-tumor [24, 94, 113, 168, 206].
activity has been reported using Calreticulin (CRT)
which has the ability to enhance MHC class I presenta-
tion and exhibit an antiangiogenic effect [28]. Another
study reported that treatment of murine breast carcinoma
Replicative Vector Oncolysis
with combined Ad-mediated murine angiostatin and One approach to overcome suboptimal tumor transduc-
Ad-mediated murine IL-12 resulted in 96% of the ani- tion of non-replicative viral vectors is the use of condi-
mals developing initial regression with 54% undergoing tionally replicative viral vectors, in which replication
total regression of the tumor. Further, mice were resistant competent virus selectively replicates within tumor cells
to rechallenge and developed strong CTL response [71]. but not in normal tissues. Release of progeny virions
One novel form of gene therapy has been reported from the initially infected tumor cells would infect
wherein a cell based anti-angiogenic gene therapy neighboring tumor cells and thereby spread throughout
approach has been employed. Fibroblasts were retrovi- the tumor volume. In addition, the use of viruses that
rally transduced to overexpress thrombospondin-2 to have a lytic life cycle would result in virus-mediated
inhibit human squamous cell carcinoma, malignant mel- oncolysis [66].
anoma, and Lewis lung carcinoma growth [184]. The currently available replicating vectors for cancer
However, a similar study using hematopoietic stem cells oncolytic virotherapy include Ad or HSV-1 and can be
transduced with retrovirus encoding a secretable form divided into two large groups: genetically engineered
of endostatin did not show inhibition of neoangiogene- viruses with enhanced selectivity and/or decreased tox-
sis or anti-tumor activity [150]. Another study employed icity (including a wide variety of vectors with specific
a gene therapy approach with murine bone marrow- deletions in viral genes or that encode essential genes
derived cells encoding soluble flk-1 to inhibit tumor under control of tumor specific promoters) and vectors
growth. Significant reduction in tumor size was reported engineered to function as therapeutic gene delivery
when challenged with tumor within 3 months after vehicles by incorporating an expression cassette con-
transplantation with bone marrow-derived cells encod- taining a transgene to improve the antitumor efficacy of
ing truncated soluble flk-1 [40]. oncolytic virotherapy (including GM-CSF or CD/TK
The induction of several inhibitors of angiogenesis genes). Importantly, some of these vectors have already
has been carried out by transfection of cells with the been explored in extensive preclinical studies with clinical
Thrombospondin-1 gene, or by using viral vectors that evaluation in patients showing significant antitumor
encode the genes for soluble platelet factor-4 and effects. As of April 2008, 28 clinical trials had been initi-
angiostatin [52]. Cationic lipid based delivery of endostatin ated employing Ad, HSV-1, Vaccinia Virus and Poliovirus
gene sequences and its expression in muscle suppressed for different tumor types [1].
602 Cancer gene therapy

Since the E1B 55kD gene product in Ad vectors is driving the E1B gene. In nu/nu mice carrying LNCaP
responsible for p53 binding and inactivation, it was xenografts, a single tail vein injection of CG7870 elimi-
hypothesized that an E1B 55kD deletion mutant Ad nated tumors within 4 weeks [221]. Also, this agent was
would be unable to inactivate p53 in normal cells and evaluated in a phase I trial of patients with hormone-
thus would be unable to replicate efficiently. Because refractory metastatic prostate cancer. Systemic injection
p53 is absent in many tumors, the replication of a mutant of CG7870 produced a transient minor decrease of pros-
Ad would be selective in tumors. A selectively replica- tate-specific antigen levels [178]. Studies using an Ad
tion-competent E1B 55kD gene-deleted Ad, dl1520 vector encoding the E1A gene under transcriptional
(ONYX-015) has been injected into solid tumors of control of the human telomerase reverse transcriptase
patients whose tumors carry mutant p53. Clinical trials promoter have shown that viral genome replication
using ONYX-015 alone produced minimal tumor and productive infection is primarily restricted to telom-
responses in patients with head and neck cancer while erase-positive tumor cells. Administration of the virus
combination treatment with chemotherapy produced a into nude mice bearing human prostate xenografts pro-
63% response rate [102]. duced significant tumor reduction [81].
In recent years, two major strategies have been devel- The second strategy is to genetically modify the fiber
oped for specific retargeting of conditionally replicating protein to present a tumor-specific binding ligand.
Ad vectors. In the first one, transcription of essential Progress in this direction has included the successful
viral genes has been controlled by replacing the native insertion of small peptides such as an RGD motif in the
viral promoters with tumor or tissue-specific promoters. Ad fiber, resulting in Ad retargeting [15]. It was shown
A number of oncolytic Ads have been generated in that the efficacy of a replicating Ad can be improved by
which the gene essential for viral replication is under incorporating a RGD peptide motif into the fiber protein
the control of exogenous promoters that are preferen- [186]. Also, an Ad that secretes a fusion molecule con-
tially active in tumor cells [77, 158, 197]. One group has sisting of the extracellular domain of CAR (sCAR) and
produced a conditionally replicative Ad in which the epidermal growth factor (EGF) was constructed.
expression of E1A is controlled by the midkine pro- Infection of tumor cells with a sCAR-EGF-retargeted
moter that induced tumor cell killing of neuroblastoma replication-competent virus system resulted in increased
and Ewing’s sarcoma cells [3]. oncolysis in vitro and a therapeutic benefit against tumor
CN706 is an oncolytic conditionally replicating Ad xenografts [76].
vector, encoding the E1A gene under the control of an In an attempt to improve both the efficacy and safety of
exogenous minimal enhancer/promoter construct oncolytic virotherapy, a replication-competent Ad vector
derived from the 5′ flank of the human PSA gene pro- encoding a CD and HSV-tk fusion gene was constructed.
moter [165]. CN706 was tested in a phase I clinical trial, A single injection of a replicative HSV-tk vector in
in which virus was intratumorally injected into patients established s.c. human glioma xenografts resulted in a
with locally recurrent prostate cancer without dose-lim- significant reduction of tumor growth. The addition of
iting toxicity [44]. Another example of a specifically ganciclovir produced an additional slowing of tumor
targeted replication-competent Ad vector, Ad-hOC-E1, growth and prolonged survival [138]. Phase I studies
which contains a single bidirectional human osteocalcin demonstrated the safety of intraprostatic administration
(hOC) promoter to drive both the early viral E1A and of this Ad in combination with conventional-dose three-
E1B genes was constructed. This vector selectively rep- dimensional conformal radiation therapy in patients with
licated in OC-expressing prostate cancer cells and viral intermediate to high risk prostate cancer [54, 57, 59].
replication was enhanced at least ten-fold with vitamin Herpes simplex virus 1 (HSV-1) vectors have also
D3 exposure. Unlike Ad-sPSA-E1, an Ad vector with been developed that replicate conditionally in dividing
viral replication controlled by a strong super prostate- tumor cells based on mutations engineered in the viral
specific antigen (sPSA) promoter which only replicates genome [193]. So far, four oncolytic HSV vectors have
in PSA-expressing cells with androgen receptor (AR), been tested in several clinical trials with encouraging
Ad-hOC-E1 retarded the growth of both androgen- results. G207 is an attenuated/replication-competent
dependent and androgen-independent prostate cancer HSV-1 mutant that lacks both copies of ICP34.5 (RL1)
cells irrespective of their basal level of AR and PSA gene and contains an insertion of lacZ in the ICP6
expression [60]. CG7870, a prostate selective replica- (UL39) gene [130]. These gene modifications offer a
tion-competent Ad with improved efficacy, contains the new modality in cancer therapy through the ability of
prostate-specific rat probasin promoter, driving the E1A G207 to selectively replicate within and kill malignant
gene, and the human prostate-specific enhancer/promoter, cells with minimal toxicity to normal tissues [106].
Donald J. Buchsbaum et al. 603

G207 induced a significant inhibition of the tumor in tumor cell resistance, or by enhancing intratumoral
growth alone and in combination with radiation in production of cytotoxic drugs [154, 223].
human prostate tumor xenograft models [202]. Based Ionizing radiation and many chemotherapies depend
on positive results in preclinical brain tumor models, on wild-type p53 function for their cytotoxic effect.
G207 has been tested in clinical trials in patients with Thus, restoration of wild-type p53 function in tumor
glioblastoma, with some evidence of disease stabiliza- cells can be used to potentiate the effects of radiation
tion [123]. A gamma-34.5 gene deleted HSV1716 vector therapy and chemotherapy [47, 213]. Animal studies
was also evaluated in patients with recurrent malignant have shown that the effects of cisplatin can be synergistic
glioma following surgical resection in phase I/II trials in combination with Adp53 gene transfer in human lung
[74, 147, 161] and in patients with metastatic melanoma cancer cells in vivo. A trial was carried out in non-small
[120]. NV1020 HSV-1 mutant vector has been tested cell lung cancer patients receiving Adp53 alone, or pre-
followed by chemotherapy in 12 patients with colorectal ceded by cisplatin. Clinical responses were observed,
cancer hepatic metastases with some minor responses and progression-free survival was prolonged with the
[114]. All the patients in these clinical trials tolerated combined treatment [169]. Colon and nasopharyngeal
the treatment well. The safety of ICP34.5-deleted HSV cancer cells transfected with Adp53 showed increased
has been shown in multiple clinical studies. sensitivity to radiation [110, 169]. Studies of Adp53
Recently, a phase I clinical trial with a second-gen- gene therapy combined with surgery, chemotherapy, or
eration oncolytic HSV expressing granulocyte mac- radiation therapy have been initiated in patients with
rophage colony-stimulating factor (Onco VEXGM-CSF) head and neck cancer and non-small cell lung cancer
has been conducted. This virus was administered intra- [17, 51, 139, 188]. Results show that Adp53 is well tol-
tumorally in patients with cutaneous or s.c. deposits of erated and produces efficacious treatment in combination
breast, head and neck and gastrointestinal cancers, and with radiation and/or chemotherapy agents [62].
malignant melanoma [79]. Improved oncolysis was Restoring or enhancing the capacity of tumor cells to
achieved through the use of a more potent clinical isolate undergo apoptosis through genetic modification of Bax
of HSV for construction of the virus, and, in addition or Bcl-2 expression has resulted in tumors being more
to the deletion of ICP34.5, the expression of the US11 sensitive to chemotherapeutic drugs and radiation ther-
gene as an immediate early rather than late gene was apy [14, 64, 210]. The cellular transcription factor E2F1
used since this has previously been shown to boost promotes apoptosis. Intratumoral injection of E2F1-
tumor-selective virus replication [23]. Also, co- expressing Ad vector in combination with gemcitabine
expression of GM-CSF has shown promising preclinical produced a significant reduction in pancreatic tumor
and clinical results [192, 205]. Incorporation of a gene xenograft size [164]. Transduction of the caspase-3 gene
encoding a cell membrane fusion glycoprotein of the in rat hepatoma cells in the liver with an Ad vector
gibbon ape leukemia virus into the HSV genome sig- induced extensive apoptosis and reduced tumor volume
nificantly increased the antitumor potency in mouse when combined with etoposide administration [215].
models of primary and metastatic human prostate A HSV type 1 mutant in combination with mitomycin C
cancer [137]. exhibited a synergistic cytotoxic effect against non-
small cell lung cancer cells in vitro and an additive effect
against tumor xenografts [195].
Transcriptional targeting via radiation inducible pro-
Chemosensitization moters in a vector can selectively produce gene transfer
in a tumor in combination with radiation therapy [31].
and Radiosensitization Promoters triggered by radiation confine the cytotoxic
The limitations of surgery and radiation therapy con- effect to the high dose target volume. Examples of radi-
cern treatment of cancer metastases. The problem with ation inducible promoters include various early response
chemotherapy is its low therapeutic ratio for many genes (c-jun, c-fos, Egr-1, and NFκB), which can be
tumors and the development of multi-drug resistance. coupled to genes which produce proteins that enhance
Gene therapy offers the potential to enhance radiobio- radiosensitivity. Similarly, the Egr-1 promoter in an Ad
logical effects through various mechanisms, and com- vector carrying the TNF-α gene has been activated by
bination treatment with radiation therapy might amplify several chemotherapy drugs [118].
tumor cell killing with selected gene transfer events. Tumor injection of an Ad expressing IL-12 and B7.1
Chemosensitization can be achieved by genetic induc- following fractionated radiation therapy resulted in a
tion of apoptosis, by inhibition of molecules involved greater therapeutic effect in murine tumor models than
604 Cancer gene therapy

with either treatment alone [117]. Expression of IL-3 in Ad as a single agent therapy, favorable interactions with
transfected murine tumors increased their response to the cytotoxic drugs cisplatin and 5-FU were reported in
radiation, and systemic administration of IL-3 gene phase II and III trials with recurrent squamous cell car-
transduced tumor cells in combination with local radia- cinoma of the head and neck [29, 58, 59, 97, 209].
tion therapy resulted in enhanced efficacy [30]. Rat Synergistic effects between oncolytic Ad vectors and
glioma cells transduced to express HSV-tk and treated chemotherapeutic drugs (e.g. paclitaxel, docetaxel, dox-
with prodrug acyclovir or bromovinyldeoxyuridine had orubicin, cisplatin, and 5-FU) have been reported in ani-
enhanced sensitivity to radiation in vitro and in vivo mal models and in clinical trials [75, 102, 220].
[31]. Weichselbaum et al. showed that irradiation medi- Combination with radiation therapy allowed a 50-fold
ates enhancement of HSV replication and produces decrease in the dose of the oncolytic Ad in a prostate
enhanced antitumor activity [4, 127]. It was shown that xenograft model [26, 44]. Combination of oncolytic Ad
HSV-tk/ganciclovir suicide gene therapy, vector-based with radiotherapy significantly increased antitumor effi-
TNF-α expression, and radiosurgery was more effective cacy against prostate cancer xenografts compared to
in controlling human glioblastoma xenografts than single either agent alone [46].
or dual-component protocols [143]. Triple therapy with
Ad.Egr-TNF, radiation, and temozolomide produced
increased survival in mice bearing glioma xenografts
compared with dual treatment [217]. A phase I trial of a
Current Limitations and Future
replication-deficient Ad vector expressing TNF-α under Directions of Cancer Gene
control of the Egr-1 promoter in combination with radi-
ation therapy in patients with soft tissue sarcomas of the
Therapy
extremity was well tolerated and produced a high num- The development of sophisticated molecular biological
ber of objective responses [136]. The combination of techniques over the past 25 years and their application
radiation therapy and angiostatin gene therapy produced to the study of the molecular mechanisms of cancer
enhanced antitumor effects in a rat glioma model [70]. have made gene therapy both a logical and practical
The combination treatment with anti angiogenic agents extension of classic treatment paradigms that is poised
and chemotherapy or radiation therapy has been shown to make major advances in new treatments for cancer in
to produce an enhanced anti-tumor effect in preclinical the near future.
models [89, 212]. Based on this observation, genetic The major limitation of gene therapy for the treat-
modification of tumor vascular endothelial cells would ment of cancer arises from the relative inefficiency of
be expected to produce an enhanced therapeutic effect current vectors in transducing target cells, the inability
in combined modality therapy. to transfer therapeutic genes into sufficient numbers of
Several chemotherapy drugs are proven radiosensi- target cells in situ to elicit the desired biological effect,
tizers. One such drug, 5-FU, when produced by the the development of both vector- and transgene-induced
CD/5-FC suicide gene therapy approach following humoral and cellular immunity, and the inability of vec-
intratumoral injection of an Ad encoding the CD gene, tors to selectively localize and transduce tumor cells fol-
has been shown by our group to enhance the effects of lowing systemic administration. Both viral and non-viral
radiation therapy in preclinical models of cholangiocar- vectors are rapidly cleared from the circulation, primar-
cinoma, glioma, pancreatic cancer, and colon cancer ily by hepatic sequestration, following intravenous
[93, 151, 181]. Thus, strategies to increase chemosensi- injection. These four limitations pose significant prob-
tivity and radiosensitivity by gene transfer have poten- lems for the development of cancer gene therapy
tially wide applicability for the treatment of cancer employing systemic vector administration. Future
clinically. Freytag et al. evaluated the efficacy and toxic- research is likely to be focused on generating modified
ity of replicative Ad-mediated suicide gene therapy in vectors with reduced toxicity and immunogenicity,
combination with radiation therapy in a preclinical increased transduction efficiency, increased duration
model of prostate cancer, and in prostate cancer clinical and regulation of gene expression, and enhanced vector
trials [54–56, 58, 59]. specificity and targeting [66, 105]. Refinements directed
Replication-selective oncolytic Ad have been con- towards these goals will be made with replicative viral
structed to target tumor tissue for selective replication vectors capable of both therapeutic gene delivery and
and amplification at the tumor site with limited replica- direct oncolysis [102]. A universal gene delivery system
tion in normal cells, minimizing toxic side effects. has yet to be identified, so that further optimization of
Although low efficacy was documented with oncolytic existing vector delivery systems is likely to occur.
Donald J. Buchsbaum et al. 605

Small interfering RNA (siRNA) has been used to


knock down or silence gene expression. This has been
References
accomplished using chemically synthesized oligonuco- 1. Gene therapy clinical trials database. J Gene Med [web site].
March 2008. Available at: http://www.wiley.co.uk/genetherapy/
leotides delivered in nanoparticles and liposomes, or clinical/. Accessed April 27, 2008, 2008.
vector-based siRNA [80, 155]. Targets have included 2. Genetic Modification Clinical Research Information System
oncogenes, and genes involved in angiogenesis, metas- (GeMCRIS). National Institutes of Health. Available at: http://
tasis, anti-apoptosis, and resistance to chemotherapy. www.gemcris.od.nih.gov/. Accessed April 25, 2008.
3. Adachi Y, Reynolds PN, Yamamoto M, Wang M, Takayama K,
Further research into this promising gene therapy tech-
Matsubara S, Muramatsu T, and Curiel DT. A midkine promoter-
nique is required. based conditionally replicative adenovirus for treatment of pediatric
Gene-modified stem cells have been used for cancer solid tumors and bone marrow tumor purging. Cancer Res 2001;
therapy [27, 153]. Their ability to traffic to tumor sites 61: 7882–7888.
makes them an attractive gene delivery vehicle. They 4. Advani SJ, Mezhir JJ, Roizman B, and Weichselbaum RR.
ReVOLT: radiation-enhanced viral oncolytic therapy. Int J Radiat
may be useful in conjunction with immunotherapy and
Oncol Biol Phys 2006; 66: 637–646.
gene therapy for treating a variety of cancers. 5. Aghi M, Hochberg F, and Breakefield XO. Prodrug activation
The targeting of tumor blood vessels should be a enzymes in cancer gene therapy. J Gene Med 2000; 2: 148–164.
useful approach for the treatment of a variety of cancers. 6. Aghi M, Kramm CM, Chou T-C, Breakefield XO, and Chiocca
Issues that need to be addressed are highly efficient EA. Synergistic anticancer effects of ganciclovir/thymidine kinase
and 5-fluorocytosine/cytosine deaminase gene therapies. J Natl
gene delivery and long-term expression of the anti- Cancer Inst 1998; 90: 370–380.
angiogenic genes in metastatic cancer. The development 7. Alemany R, Balagué C, and Curiel DT. Replicative adenoviruses
of targeted vectors should aid in the development for cancer therapy. Nature Biotech 2000; 18: 723–727.
of this approach. Gene-directed enzyme prodrug 8. Alvarez RD, Barnes MN, Gomez-Navarro J, Wang M, Strong TV,
therapy uses transcriptional differences between normal Arafat W, Arani RB, Johnson MR, Roberts BL, Siegal GP, and
Curiel DT. A cancer gene therapy approach utilizing an anti-erbB-2
and cancer cells to drive the selective expression of a single-chain antibody-encoding adenovirus (AD21): A phase I
metabolic “suicide gene” that is able to convert trial. Clin Cancer Res 2000; 6: 3081–3087.
a nontoxic prodrug into its toxic metabolite. A variety 9. Alvarez RD, Gomez-Navarro J, Wang M, Barnes MN, Strong TV,
of enzyme prodrug systems have been evaluated in Arani RB, Arafat W, Hughes JV, Siegal GP, and Curiel DT.
Adenoviral-mediated suicide gene therapy for ovarian cancer. Mol
phase I/II clinical trials using either retrovirus or
Ther 2000; 2: 524–530.
adenovirus to deliver the enzyme to tumor cells [38]. 10. Anderson LM, Krotz S, Weitzman SA, and Thimmapaya B. Breast
The evaluation of gene therapy in combination with cancer-specific expression of the Candida albicans cytosine deam-
surgery, radiation therapy, and chemotherapy is likely to inase gene using a transcriptional targeting approach. Cancer Gene
undergo further development. Ther 2000; 7: 845–852.
11. Anderson WF. Human gene therapy. Nature 1998; 392: 25–30.
Gene therapy will continue to be applied to many
12. Ansardi DC, Porter DC, Jackson CA, Gillespie GY, and Morrow
fields of medicine, and particularly to the treatment of CD. RNA replicons derived from poliovirus are directly oncolytic
cancer. Whereas human gene therapy trials for cancer for human tumor cells of diverse origins. Cancer Res 2001; 61:
have not yet yielded clear benefit, these studies highlight 8470–8479.
the deficiencies of current approaches. Gene therapy will 13. Anwer K, Meaney C, Kao G, Hussain N, Shelvin R, Earls RM,
Leonard P, Quezada A, Rolland AP, and Sullivan SM. Cationic
likely become an integral component of a multimodality lipid-based delivery system for systemic cancer gene therapy.
strategy for the treatment of cancer. Improvements in Cancer Gene Ther 2000; 7: 1156–1164.
vectors that increase the specificity and duration of gene 14. Arafat WO, Gómez-Navarro J, Xiang J, Barnes MN, Mahasreshti P,
expression, the reduction of immunogenicity and toxic- Alvarez RD, Siegal GP, Badib AO, Buchsbaum D, Curiel DT, and
ity, and targeted vector delivery are required for advance- Stackhouse M. An adenovirus encoding proapoptotic bax induces
apoptosis and enhances the radiation effect in human ovarian cancer.
ment of the clinical practice of gene therapy with an Mol Ther 2000; 1: 545–554.
increase in response rate. As knowledge of the molecular 15. Barnett BG, Crews CJ, and Douglas JT. Targeted adenoviral vectors.
basis of cancer accumulates, and as genetic screening Biochim Biophys Acta 2002; 1575: 1–14.
programs are introduced to identify high-risk patients, it 16. Bartlett JS. Prospects for the development of targeted adeno-associated
virus (AAV) vector systems. Tumor Targeting 1999; 4: 143–149.
might be possible to use gene therapy for early treat-
17. Baselga J. New horizons: Gene therapy for cancer. Anti-Cancer
ment, including vaccination against cancer. Drugs (Suppl 1) 1999; 10: S39–S42.
18. Botella-Estrada R, Malet G, Revert F, Dasi F, Crespo A, Sanmartin
Acknowledgements We acknowledge research support from O, Guillen C, and Alino SF. Antitumor effect of B16 melanoma
the National Institutes of Health (P20 CA101955). We are cells genetically modified with the angiogenesis inhibitor rnasin.
grateful to Sally Lagan for preparation of the manuscript. Cancer Gene Ther 2001; 8: 278–284.
606 Cancer gene therapy

19. Brade AM, Ngo D, Szmitko P, Li P-X, Liu F-F, and Klamut HJ. 36. Curiel DT. Strategies to adapt adenoviral vectors for targeted
Heat-directed gene targeting of adenoviral vectors to tumor cells. delivery. Ann N Y Acad Sci 1999; 886: 158–171.
Cancer Gene Ther 2000; 7: 1566–1574. 37. Curiel DT. The development of conditionally replicative adenovi-
20. Brower V. Tumor angiogenesis – new drugs on the block. Nat ruses for cancer therapy. Clin Cancer Res 2000; 6: 3395–3399.
Biotechnol 1999; 17: 963–968. 38. Dachs GU, Tupper J, and Tozer GM. From bench to bedside for
21. Buchsbaum DJ, Miller CR, Mahasreshti PJ, and Curiel DT. Cancer gene-directed enzyme prodrug therapy of cancer. Anticancer
gene therapy. In: Oldham RK (ed). Principles of Cancer Drugs 2005; 16: 349–359.
Biotherapy, 4th edn. Kluwer: Dordrecht, 2003, pp. 583–613. 39. Dang CV, Gerson SL, Litwak M, and Padarathsingh M. Gene
22. Buchschacher GL, Jr., and Wong-Staal F. Development of lentiviral therapy and translational cancer research. Clin Cancer Res 1999;
vectors for gene therapy for human diseases. Blood 2000; 95: 5: 471–474.
2499–2504. 40. Davidoff AM, Ng CY, Brown P, Leary MA, Spurbeck WW, Zhou
23. Cassady KA, Gross M, and Roizman B. The herpes simplex virus J, Horwitz E, Vanin EF, and Nienhuis AW. Bone marrow-derived
US11 protein effectively compensates for the gamma1(34.5) gene cells contribute to tumor neovasculature and, when modified to
if present before activation of protein kinase R by precluding its express an angiogenesis inhibitor, can restrict tumor growth in
phosphorylation and that of the alpha subunit of eukaryotic trans- mice. Clin Cancer Res 2001; 7: 2870–2879.
lation initiation factor 2. J Virol 1998; 72: 8620–8626. 41. de Gast GC, Gallee MPW, Spits H, Gerritsen WR, Batchelor D,
24. Channon KM, and George SE. Improved adenoviral vectors: cau- Clift S, Nooijen WJ, Kroon BBR, Berns AM, and Rankin EM.
tious optimism for gene therapy. Q J Med 1997; 90: 105–109. Immunological, pathological, and long-term clinical data of vac-
25. Chen CT, Lin J, Li Q, Phipps SS, Jakubczak JL, Stewart DA, cination with autologous granulocoyte-macrophage colony-stimu-
Skripchenko Y, Forry-Schaudies S, Wood J, Schnell C, and lating factor-transduced tumor cells in metastatic melanoma.
Hallenbeck PL. Antiangiogenic gene therapy for cancer via sys- Cancer Gene Ther 2000; 7: 1204.
temic administration of adenoviral vectors expressing secretable 42. Deshane J, Cabrera G, Grim JE, Siegel GP, Pike J, Alvarez RD,
endostatin. Hum Gene Ther 2000; 11: 1983–1996. and Curiel DT. Targeted eradication of ovarian cancer mediated by
26. Chen Y, DeWeese T, Dilley J, Zhang Y, Li Y, Ramesh N, Lee J, intracellular expression of anti-erbB-2 single-chain antibody.
Pennathur-Das R, Radzyminski J, Wypych J, Brignetti D, Scott S, Gynecol Oncol 1995; 59: 8–14.
Stephens J, Karpf DB, Henderson DR, and Yu DC. CV706, a pros- 43. Deshane J, Grim J, Loechel S, Siegal GP, Alvarez RD, and Curiel
tate cancer-specific adenovirus variant, in combination with radio- DT. Intracellular antibody against erbB-2 mediates targeted tumor
therapy produces synergistic antitumor efficacy without increasing cell eradication by apoptosis. Cancer Gene Ther 1996; 3: 89–98.
toxicity. Cancer Res 2001; 61: 5453–5460. 44. DeWeese TL, van der Poel H, Li S, Mikhak B, Drew R, Goemann
27. Cheng T. Toward ‘SMART’ stem cells. Gene Ther 2008; 15: M, Hamper U, DeJong R, Detorie N, Rodriguez R, Haulk T,
67–73. DeMarzo AM, Piantadosi S, Yu DC, Chen Y, Henderson DR,
28. Cheng WF, Hung CF, Chai CY, Hsu KF, He L, Ling M, and Wu Carducci MA, Nelson WG, and Simons JW. A phase I trial of
TC. Tumor specific immunity and antiangiogensis generated by a CV706, a replication-competent, PSA selective oncolytic adenovi-
DNA vaccine encoding calreticulin linked to a tumor antigen. Clin rus, for the treatment of locally recurrent prostate cancer following
Invest 2001; 5: 669–678. radiation therapy. Cancer Res 2001; 61: 7464–7472.
29. Cheong SC, Wang Y, Meng JH, Hill R, Sweeney K, Kirn D, 45. Diaz RM, Bateman A, Emiliusen L, Fielding A, Trono D, Russell
Lemoine NR, and Hallden G. E1A-expressing adenoviral E3B SJ, and Vile RG. A lentiviral vector expressing a fusogenic glyco-
mutants act synergistically with chemotherapeutics in immuno- protein for cancer gene therapy. Gene Ther 2000; 7: 1656–1663.
competent tumor models. Cancer Gene Ther 2008; 15: 40–50. 46. Dilley J, Reddy S, Ko D, Nguyen N, Rojas G, Working P, and Yu
30. Chiang C-S, Hong J-H, Wu YC, McBride WH, and Dougherty GJ. DC. Oncolytic adenovirus CG7870 in combination with radiation
Combining radiation therapy with interleukin-3 gene immuno- demonstrates synergistic enhancements of antitumor efficacy
therapy. Cancer Gene Ther 2000; 7: 1172–1178. without loss of specificity. Cancer Gene Ther 2005; 12: 715–722.
31. Chmura SJ, Advani SJ, Kufe DW, and Weichselbaum RR. 47. Dorigo O, Turla ST, Lebedeva S, and Gjerset RA. Sensitization of
Strategies for enhancing viral-based gene therapy using ionizing rat glioblastoma multiforme to cisplatin in vivo following restora-
radiation. Radiat Oncol Investig 1999; 7: 261–269. tion of wild-type p53 function. J Neurosurg 1998; 88: 535–540.
32. Clayman GL. The current status of gene therapy. Semin Oncol 48. Dranoff G. Cancer gene therapy: Connecting basic research with
2000; 27: 39–43. clinical inquiry. J Clin Oncol 1998; 16: 2548–2556.
33. Clayman GL, Frank DK, Bruso PA, and Goepfert H. Adenovirus- 49. Dropulic B. Lentivirus in the clinic. Mol Ther 2001; 4: 511–512.
mediated wild-type p53 gene transfer as a surgical adjuvant in 50. Eck SL. The prospects for gene therapy. Hosp Pract 1999; 34:
advanced head and neck cancers. Clin Cancer Res 1999; 5: 67–75.
1715–1722. 51. Fahraeus R, Fischer P, Krausz E, and Lane DP. New approaches to
34. Cohen H, Levy RJ, Gao J, Fishbein I, Kousaev V, Sosnowski S, cancer therapies. J Pathol 1999; 187: 138–146.
Slomkowski S, and Golomb G. Sustained delivery and expression 52. Feldman AL, and Libutti SK. Progress in antiangiogenic gene
of DNA encapsulated in polymeric nanoparticles. Gene Ther 2000; therapy of cancer. Cancer 2000; 89: 1181–1194.
7: 1896–1905. 53. Finn OJ, and Lotze MT. Cellular immunity and immunotherapy of
35. Cowan KH, Moscow JA, Huang H, Zujewski JA, O’Shaughnessy cancer. Clin Cancer Res (Suppl) 2001; 7: 759s–989s.
J, Sorrentino B, Hines K, Carter C, Schneider E, Cusack G, Noone 54. Freytag SO, Khil M, Stricker H, Peabody J, Menon M, DePeralta-
M, Dunbar C, Steinberg S, Wilson W, Goldspiel B, Read EJ, Venturina M, Nafziger D, Pegg J, Paielli D, Brown S, Barton K, Lu
Leitman SF, McDonagh K, Chow C, Abati A, Chiang Y, Chang M, Aguilar-Cordova E, and Kim JH. Phase I study of replication-
YN, Gottesman MM, Pastan I, and Nienhuis A. Paclitaxel chemo- competent adenovirus-mediated double suicide gene therapy for
therapy after autologous stem-cell transplantation and engraftment the treatment of locally recurrent prostate cancer. Cancer Res
of hematopoietic cells transduced with a retrovirus containing the 2002; 62: 4968–4976.
multidrug resistance complementary DNA (MDR1) in metastatic 55. Freytag SO, Movsas B, Aref I, Stricker H, Peabody J, Pegg J,
breast cancer patients. Clin Cancer Res 1999; 5: 1619–1628. Zhang Y, Barton KN, Brown SL, Lu M, Savera A, and Kim JH.
Donald J. Buchsbaum et al. 607

Phase I trial of replication-competent adenovirus-mediated suicide effects of radiotherapy and angiostatin gene therapy in glioma tumor
gene therapy combined with IMRT for prostate cancer. Mol Ther model. Proc Natl Acad Sci USA 2000; 97: 6698–6703.
2007; 15: 1016–1023. 71. Gyorffy S, Palmer K, Podor TJ, Hitt M, and Gauldie J. Combined
56. Freytag SO, Paielli D, Wing M, Rogulski K, Brown S, Kolozsvary treatment of a murine breast cancer model with type 5 adenovirus
A, Seely J, Barton K, Dragovic A, and Kim JH. Efficacy and toxic- vectors expressing murine angiostatin and IL-12: A role for com-
ity of replication-competent adenovirus-mediated double suicide bined anti-angiogenesis and immunotherapy. J Immunol 2001;
gene therapy in combination with radiation therapy in an orthoto- 166: 6212–6217.
pic mouse prostate cancer model. Int J Radiat Oncol Biol Phys 72. Haisma HJ, van Beusechem W, Curiel DT, Pinedo HM, and
2002; 54: 873–885. Gerritsen WR. Targeted enzymes for molecular chemotherapy.
57. Freytag SO, Rogulski KR, Paielli DL, Gilbert JD, and Kim JH. Cancer Gene Ther 2000; 7: 1208.
A novel three-pronged approach to kill cancer cells selectively: 73. Han S-o, Mahato RI, Sung YK, and Kim SW. Development of
Concomitant viral, double suicide gene, and radiotherapy. Hum biomaterials for gene therapy. Mol Ther 2000; 2: 302–317.
Gene Ther 1998; 9: 1323–1333. 74. Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser
58. Freytag SO, Stricker H, Peabody J, Pegg J, Paielli D, Movsas B, M, Hadley D, Patterson J, Brown SM, and Rampling R. HSV1716
Barton KN, Brown SL, Lu M, and Kim JH. Five-year follow-up of injection into the brain adjacent to tumour following surgical
trial of replication-competent adenovirus-mediated suicide gene resection of high-grade glioma: safety data and long-term survival.
therapy for treatment of prostate cancer. Mol Ther 2007; 15: Gene Ther 2004; 11: 1648–1658.
636–642. 75. Heise C, Lemmon M, and Kirn D. Efficacy with a replication-
59. Freytag SO, Stricker H, Pegg J, Paielli D, Pradhan DG, Peabody J, selective adenovirus plus cisplatin-based chemotherapy: depen-
DePeralta-Venturina M, Xia X, Brown S, Lu M, and Kim JH. dence on sequencing but not p53 functional status or route of
Phase I study of replication-competent adenovirus-mediated double- administration. Clin Cancer Res 2000; 6: 4908–4914.
suicide gene therapy in combination with conventional-dose three- 76. Hemminki A, Dmitriev I, Liu B, Desmond RA, Alemany R, and
dimensional conformal radiation therapy for the treatment of Curiel DT. Targeting oncolytic adenoviral agents to the epidermal
newly diagnosed, intermediate- to high-risk prostate cancer. growth factor pathway with a secretory fusion molecule. Cancer
Cancer Res 2003; 63: 7497–7506. Res 2001; 61: 6377–6381.
60. Fujii S, Huang S, Fong TC, Ando D, Burrows F, Jolly DJ, 77. Hernandez-Alcoceba R, Pihalja M, Qian D, and Clarke MF. New
Nemunaitis J, and Hoon DS. Induction of melanoma-associated oncolytic adenoviruses with hypoxia- and estrogen receptor-regu-
antigen systemic immunity upon intratumoral delivery of inter- lated replication. Hum Gene Ther 2002; 13: 1737–1750.
feron-gamma retroviral vector in melanoma patients. Cancer Gene 78. Hsu FJ, Benike C, Fagnoni F, Liles TM, Czerwinski D, Taidi B,
Ther 2000; 7: 1220–1230. Engleman EG, and Levy R. Vaccination of patients with B-cell
61. Fujiwara T, Urata Y, and Tanaka N. Telomerase-specific oncolytic lymphoma using autologous antigen-pulsed dendritic cells. Nat
virotherapy for human cancer with the hTERT promoter. Curr Med 1996; 2: 52–58.
Cancer Drug Targets 2007; 7: 191–201. 79. Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ,
62. Gabrilovich DI. INGN 201 (Advexin): adenoviral p53 gene ther- Harrington KJ, James ND, Love CA, McNeish I, Medley LC,
apy for cancer. Expert Opin Biol Ther 2006; 6: 823–832. Michael A, Nutting CM, Pandha HS, Shorrock CA, Simpson J,
63. Galanis E, Hersh EM, Stopeck AT, Gonzalez R, Burch P, Spier C, Steiner J, Steven NM, Wright D, and Coombes RC. A phase I study
Akporiaye ET, Rinehart JJ, Edmonson J, Sobol RE, Forscher C, of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex
Sondak VK, Lewis BD, Unger EC, O’Driscoll M, Selk L, and virus expressing granulocyte macrophage colony-stimulating factor.
Rubin J. Immunotherapy of advanced malignancy by direct gene Clin Cancer Res 2006; 12: 6737–6747.
transfer of an interleukin-2 DNA/DMRIE/DOPE lipid complex: 80. Huang C, Li M, Chen C, and Yao Q. Small interfering RNA therapy
phase I/II experience. J Clin Oncol 1999; 17: 3313–3323. in cancer: mechanism, potential targets, and clinical applications.
64. Gomez-Navarro J, Arafat W, and Xiang J. Gene therapy for carci- Expert Opin Ther Targets 2008; 12: 637–645.
noma of the breast: Pro-apoptotic gene therapy. Breast Cancer Res 81. Huang Q, Zhang X, Wang H, Yan B, Kirkpatrick J, Dewhrist MW,
2000; 2: 32–44. and Li CY. A novel conditionally replicative adenovirus vector tar-
65. Gomez-Navarro J, and Curiel DT. Conditionally replicative adeno- geting telomerase-positive tumor cells. Clin Cancer Res 2004; 10:
viral vectors for cancer gene therapy. Lancet Oncol 2000; 1: 1439–1445.
148–158. 82. Ido A, Uto H, Moriuchi A, Nagata K, Onaga Y, Onaga M, Hori T,
66. Gomez-Navarro J, Curiel DT, and Douglas JT. Gene therapy for Hirono S, Hayashi K, Tamaoki T, and Tsubouchi H. Gene therapy
cancer. Eur J Cancer 1999; 35: 867–885. targeting for hepatocellular carcinoma: Selective and enhanced
67. Greco O, and Dachs GU. Gene directed enzyme/prodrug therapy suicide gene expression regulated by a hypoxia-inducible enhancer
of cancer: historical appraisal and future prospectives. J Cell linked to a human α-fetoprotein promoter. Cancer Res 2001; 61:
Physiol 2001; 187: 22–36. 3016–3021.
68. Grifman M, Trepel M, Speece P, Gilbert LB, Arap W, Pasqualini 83. Inoue K, Perrotte P, Wood CG, Slaton JW, Sweeney P, and Dinney CP.
R, and Weitzman MD. Incorporation of tumor-targeting peptides Gene therapy of human bladder cancer with adenovirus-mediated
into recombinant adeno-associated virus capsids. Mol Ther 2001; antisense basic fibroblast growth factor. Clin Cancer Res 2000; 6:
3: 964–975. 4422–4431.
69. Grill J, van Beusechem VW, Van Der Valk P, Dirven CMF, 84. Inoue K, Wood CG, Slaton JW, Karashima T, Sweeney P, and
Leonhart A, Pherai DS, Haisma HJ, Pinedo HM, Curiel DT, and Dinney CP. Adenoviral-mediated gene therapy of human bladder
Gerritsen WR. Combined targeting of adenoviruses to integrins cancer with antisense interleukin-8. Oncol Rep 2001; 8:
and epidermal growth factor receptors increases gene transfer into 955–964.
primary glioma cells and spheroids. Clin Cancer Res 2001; 7: 85. Izawa JI, Sweeney P, Perrotte P, Kedar D, Dong Z, Slaton JW,
641–650. Karashima T, Inoue K, Benedict WF, and Dinney CP. Inhibition of
70. Griscelli F, Li H, Cheong C, Opolon P, Bennaceur-Griscelli A, tumorigenicity and metastasis of human bladder cancer growing
Vassal G, Soria J, Soria C, Lu H, Perricaudet M, and Yeh P. Combined in athymic mice by interferon-beta gene therapy results partially
608 Cancer gene therapy

from various antiangiogenic effects including endothelial cell 100. Kim SK, Wang KC, Cho BK, Chung HT, Kim YY, Lim SY, Lee
apoptosis. Clin Cancer Res 2002; 8: 1258–1270. CT, and Kim HJ. Interaction between p53 and p16 expressed by
86. Jiang A, and Dornburg R. In vivo cell type-specific gene delivery adenoviral vectors in human malignant glioma cell lines.
with retroviral vectors that display single chain antibodies. Gene J Neurosurg 2002; 97: 143–150.
Ther 1999; 6: 1982–1987. 101. Kirkman W, III, Chen P, Schroeder R, Feneley MR, Rodriguez R,
87. Kadowaki Y, Fujiwara T, Fukazawa T, Shao J, Yasuda T, Itoshima Wickham TJ, King CR, and Bruder JT. Transduction and apopto-
T, Kagawa S, Hudson LG, Roth JA, and Tanaka N. Induction of sis induction in the rat prostate, using adenovirus vectors. Hum
differentiation-dependent apoptosis in human esophageal Gene Ther 2001; 12: 1499–1512.
squamous cell carcinoma by adenovirus-mediated p21sdi1 gene 102. Kirn D. Clinical research results with dl1520 (Onyx-015), a rep-
transfer. Clin Cancer Res 1999; 5: 4233–4241. lication-selective adenovirus for the treatment of cancer: What
88. Kagawa S, Pearson SA, Ji L, Xu K, McDonnell TJ, Swisher SG, have we learned?. Gene Ther 2001; 8: 89–98.
Roth JA, and Fang B. A binary adenoviral vector system for 103. Kleinman HK and Liau G. Gene therapy for antiangiogenesis.
expressing high levels of the proapoptotic gene bax. Gene Ther J Natl Cancer Inst 2001; 93: 965–967.
2000; 7: 75–79. 104. Kong HL, and Crystal RG. Gene therapy strategies for tumor
89. Kakeji Y, and Teicher BA. Preclinical studies of the combination antiangiogenesis. J Natl Cancer Inst 1998; 90: 273–286.
of angiogenic inhibitors with cytotoxic agents. Invest New Drugs 105. Kouraklis G. Gene therapy for cancer: From the laboratory to the
1997; 15: 39–48. patient. Dig Dis Sci 2000; 45: 1045–1052.
90. Kaliberov S, Buchsbaum DJ, Gillespie GY, Curiel DT, Carpenter 106. Kramm CM, Rainov NG, Sena-Esteves M, Barnett FH, Chase M,
M, Arafat WO, and Stackhouse MA. Adenovirus-mediated trans- Herrlinger U, Pechan PA, Chiocca EA, and Breakefield XO.
fer of Bax driven by the vascular endothelial growth factor pro- Long-term survival in a rodent model of disseminated brain
moter induces apoptosis in lung cancer cells. Mol Ther 2002; 6: tumors by combined intrathecal delivery of herpes vectors and
190–198. ganciclovir treatment. Hum Gene Ther 1996; 7: 1989–1994.
91. Kaliberov SA, Kaliberova L, Stockard CR, Grizzle WE, and 107. Krasnykh V, Dmitriev I, Gomez-Navarro J, Belousova N,
Buchsbaum DJ. Adenovirus-mediated flt-1 targeted proapoptotic Kashentseva E, Xiang J, Douglas JT, and Curiel DT. Advanced
gene therapy of human prostate cancer. Mol Ther 2004; 10: generation adenoviral vectors possess augmented gene transfer
1059–1070. efficiency based upon coxsackie adenovirus receptor-indepen-
92. Kaliberov SA, Kaliberova LN, and Buchsbaum DJ. Combined dent cellular entry capacity. Cancer Res 2000; 60: 6784–6787.
ionizing radiation and sKDR gene delivery for treatment of pros- 108. Lemoine NR. Tumor-selective transcriptional targeting for can-
tate carcinomas. Gene Ther 2005; 12: 407–417. cer gene therapy. Cancer Gene Ther 2000; 7: 1207–1208.
93. Kaliberov SA, Markert JM, Gillespie GY, Krendelchtchikova V, 109. Li H, Lindenmeyer F, Grenet C, Opolon P, Menashi S, Soria C,
Della Manna D, Sellers JC, Kaliberova LN, Black ME, and Yeh P, Perricaudet M, and Lu H. AdTIMP-2 inhibits tumor
Buchsbaum DJ. Mutation of Escherichia coli cytosine deaminase growth, angiogenesis, and metastasis, and prolongs survival in
significantly enhances molecular chemotherapy of human glioma. mice. Hum Gene Ther 2001; 12: 515–526.
Gene Ther 2007; 14: 1111–1119. 110. Li JH, Lax SA, Kim J, Klamut H, and Liu FF. The effects of
94. Kaplan JM, Armentano D, Sparer TE, Wynn SG, Peterson PA, combining ionizing radiation and adenoviral p53 therapy in
Wadsworth SC, Couture KK, Pennington SE, St George JA, nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys 1999;
Gooding LR, and Smith AE. Characterization of factors involved in 43: 607–616.
modulating persistence of transgene expression from recombinant 111. Li S, and Huang L. Nonviral gene therapy: Promises and chal-
adenovirus in the mouse lung. Hum Gene Ther 1997; 8: 45–56. lenges. Gene Ther 2000; 7: 31–34.
95. Kasano K, Blackwell JL, Douglas JT, Dmitriev I, Strong TV, 112. Li S, Xia X, Zhang X, and Suen J. Regression of tumors by IFN-
Reynolds P, Kropf DA, Carroll WR, Peter GE, Bucy RP, Curiel alpha electroporation gene therapy and analysis of the responsi-
DT, and Krasnykh V. Selective gene delivery to head and neck ble genes by cDNA array. Gene Ther 2002; 9: 390–397.
cancer via an integrin targeted adenoviral vector. Clin Cancer 113. Liau G, Su EJ, and Dixon KD. Clinical efforts to modulate angio-
Res 1999; 5: 2571–2579. genesis in the adult: gene therapy versus conventional approaches.
96. Kay MA, Glorioso JC, and Naldini L. Viral vectors for gene ther- Drug Discov Today 2001; 6: 689–697.
apy: The art of turning infectious agents into vehicles of therapeu- 114. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ,
tics. Nat Med 2001; 7: 33–40. Giovannone AJ, Lin JW, Chute DJ, Mischel PS, Cloughesy TF,
97. Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, and Roth MD. Dendritic cell vaccination in glioblastoma patients
Gore M, Ironside J, MacDougall RH, Heise C, Randlev B, Gillenwater induces systemic and intracranial T-cell responses modulated by
AM, Bruso P, Kaye SB, Hong WK, and Kirn DH. a controlled trial of the local central nervous system tumor microenvironment. Clin
intratumoral ONYX-015, a selectively-replicating adenovirus, in Cancer Res 2005; 11: 5515–5525.
combination with cisplatin and 5-fluorouracil in patients with recur- 115. Lin E, and Nemunaitis J. Oncolytic viral therapies. Cancer Gene
rent head and neck cancer. Nat Med 2000; 6: 879–885. Ther 2004; 11: 643–664.
98. Kievit E, Bershad E, Ng E, Sethna P, Dev I, Lawrence TS, and 116. Lin P, Buxton JA, Acheson A, Radziejewski C, Maisonpierre PC,
Rehemtulla A. Superiority of yeast over bacterial cytosine deami- Yancopoulos GD, Channon KM, Hale LP, Dewhirst MW, George
nase for enzyme/prodrug gene therapy in colon cancer xeno- SE, and Peters KG. Antiangiogenic gene therapy targeting the
grafts. Cancer Res 1999; 59: 1417–1421. endothelium-specific receptor tyrosine kinase Tie2. Proc Natl
99. Kievit E, Nyati MK, Ng E, Stegman LD, Parsels J, Ross BD, Acad Sci USA 1998; 95: 8829–8834.
Rehemtulla A, and Lawrence TS. Yeast cytosine deaminase 117. Lohr F, Hu K, Haroon Z, Samulski TV, Huang Q, Beaty J,
improves radiosensitization and bystander effect by 5-fluorocytosine Dewhirst MW, and Li CY. Combination treatment of murine
of human colorectal cancer xenografts. Cancer Res (Suppl) 2000; tumors by adenovirus-mediated local B7/IL12 immunotherapy
60: 6649–6655. and radiotherapy. Mol Ther 2000; 2: 195–203.
Donald J. Buchsbaum et al. 609

118. Lopez CA, Kimchi ET, Mauceri HJ, Park JO, Mehta N, Murphy 135. Mulvihill S, Warren R, Venook A, Adler A, Randlev B, Heise C,
KT, Beckett MA, Hellman S, Posner MC, Kufe DW, and and Kirn D. Safety and feasibility of injection with an E1B-
Weichselbaum RR. Chemoinducible gene therapy: a strategy to 55 kDa gene-deleted, replication-selective adenovirus (ONYX-
enhance doxorubicin antitumor activity. Mol Cancer Ther 2004; 015) into primary carcinomas of the pancreas: A phase I trial.
3: 1167–1175. Gene Ther 2001; 8: 308–315.
119. Ma H, Liu Y, Liu S, Kung HF, Sun X, Zheng D, and Xu R. 136. Mundt AJ, Vijayakumar S, Nemunaitis J, Sandler A, Schwartz H,
Recombinant adeno-associated virus-mediated TRAIL gene ther- Hanna N, Peabody T, Senzer N, Chu K, Rasmussen CS, Kessler
apy suppresses liver metastatic tumors. Int J Cancer 2005; 116: PD, Rasmussen HS, Warso M, Kufe DW, Gupta TD, and
314–321. Weichselbaum RR. A Phase I trial of TNFerade biologic in
120. MacKie RM, Stewart B, and Brown SM. Intralesional injection patients with soft tissue sarcoma in the extremities. Clin Cancer
of herpes simplex virus 1716 in metastatic melanoma. Lancet Res 2004; 10: 5747–5753.
2001; 357: 525–526. 137. Nakamori M, Fu X, Pettaway CA, and Zhang X. Potent antitumor
121. Mahan SD, Ireton GC, Stoddard BL, and Black ME. Alanine- activity after systemic delivery of a doubly fusogenic oncolytic
scanning mutagenesis reveals a cytosine deaminase mutant with herpes simplex virus against metastatic prostate cancer. Prostate
altered substrate preference. Biochemistry 2004; 43: 8957–8964. 2004; 60: 53–60.
122. Mahasreshti PJ, Navarro JG, Kataram M, Wang MH, Carey D, 138. Nanda D, Vogels R, Havenga M, Avezaat CJ, Bout A, and Smitt
Siegal GP, Barnes MN, Nettelbeck DM, Alvarez RD, Hemminki A, PS. Treatment of malignant gliomas with a replicating adenoviral
and Curiel DT. Adenovirus-mediated soluble FLT-1 gene therapy vector expressing herpes simplex virus-thymidine kinase. Cancer
for ovarian carcinoma. Clin Cancer Res 2001; 7: 2057–2066. Res 2001; 61: 8743–8750.
123. Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, 139. Nemunaitis J, Swisher SG, Timmons T, Connors D, Mack M,
Hunter WD, Palmer CA, Feigenbaum F, Tornatore C, Tufaro F, Doerksen L, Weill D, Wait J, Lawrence DD, Kemp BL, Fossella
and Martuza RL. Conditionally-replicating herpes simplex virus F, Glisson BS, Hong WK, Khuri FR, Kurie JM, Lee JJ, Lee JS,
mutant, G207 for the treatment of malignant glioma: Results of a Nguyen DM, Nesbitt JC, Perez-Soler R, Pisters KM, Putnam JB,
phase I trial. Gene Ther 2000; 7: 867–874. Richli WR, Shin DM, Walsh GL, Merritt J, and Roth J.
124. Martinet O, Ermekova V, Qiao JQ, Sauter B, Mandeli J, Chen L, Adenovirus-mediated p53 gene transfer in sequence with cispla-
and Chen S-H. Immunomodulatory gene therapy with interleukin tin to tumors of patients with non-small-cell lung cancer. J Clin
12 and 4–1BB ligand: long- term remission of liver metastases in Oncol 2000; 18: 609–622.
a mouse model. J Natl Cancer Inst 2000; 92: 931–936. 140. Nguyen M, Huan-Tu G, Gonzalez-Edick M, Rivera VM,
125. Maxwell IH, and Cripe TP. Transcriptional targeting. Tumor Clackson T, Jooss KU, and Harding TC. Rapamycin-regulated
Targeting 2000; 4: 189–209. control of antiangiogenic tumor therapy following rAAV-mediated
126. McIvor RS. Gene therapy of genetic diseases and cancer. Pediatr gene transfer. Mol Ther 2007; 15: 912–920.
Transplant 1999; 3 Suppl 1: 116–121. 141. Nielsen LL, and Maneval DC. P53 tumor suppressor gene ther-
127. Mezhir JJ, Advani SJ, Smith KD, Darga TE, Poon AP, Schmidt apy for cancer. Cancer Gene Ther 1998; 5: 52–63.
H, Posner MC, Roizman B, and Weichselbaum RR. Ionizing 142. Nienhuis AW, Dunbar CE, and Sorrentino BP. Genotoxicity of
radiation activates late herpes simplex virus 1 promoters via the retroviral integration in hematopoietic cells. Mol Ther 2006; 13:
p38 pathway in tumors treated with oncolytic viruses. Cancer 1031–1049.
Res 2005; 65: 9479–9484. 143. Niranjan A, Moriuchi S, Lunsford LD, Kondziolka D, Flickinger
128. Miller CR, Williams CR, Buchsbaum DJ, and Gillespie GY. JC, Fellows W, Rajendiran S, Tamura M, Cohen JB, and Glorioso
Intratumoral 5-fluorouracil produced by cytosine deaminase/5- JC. Effective treatment of experimental glioblastoma by HSV
fluorocytosine gene therapy is effective for experimental human vector-mediated TNFα and HSV-tk gene transfer in combination
glioblastomas. Cancer Res 2002; 62: 773–780. with radiosurgery and ganciclovir administration. Mol Ther 2000;
129. Minaguchi T, Mori T, Kanamori Y, Matsushima M, Yoshikawa 2: 114–120.
H, Taketani Y, and Nakamura Y. Growth suppression of human 144. Nishikawa M, and Huang L. Nonviral vectors in the new millen-
ovarian cancer cells by adenovirus-mediated transfer of the PTEN nium: Delivery barriers in gene transfer. Hum Gene Ther 2001;
gene. Cancer Res 1999; 59: 6063–6067. 12: 861–870.
130. Mineta T, Rabkin SD, Yazaki T, Hunter WD, and Martuza RL. 145. Oldfield EH, Ram Z, Culver KW, Blaese RM, DeVroom HL, and
Attenuated multi-mutated herpes simplex virus-1 for the treat- Anderson WF. Gene therapy for the treatment of brain tumors
ment of malignant gliomas. Nat Med 1995; 1: 938–943. using intra-tumoral transduction with the thymidine kinase gene
131. Molenkamp BG, van Leeuwen PA, Meijer S, Sluijter BJ, and intravenous ganciclovir. Hum Gene Ther 1993; 4: 39–69.
Wijnands PG, Baars A, van den Eertwegh AJ, Scheper RJ, and de 146. Pandha HS, Martin L-A, Rigg A, Hurst HC, Stamp GWH, Sikora
Gruijl TD. Intradermal CpG-B activates both plasmacytoid and K, and Lemoine NR. Genetic prodrug activation therapy for
myeloid dendritic cells in the sentinel lymph node of melanoma breast cancer: A phase I clinical trial of erbB-2-directed suicide
patients. Clin Cancer Res 2007; 13: 2961–2969. gene expression. J Clin Oncol 1999; 17: 2180–2189.
132. Moolten F. Tumor chemosensitivity conferred by inserted herpes 147. Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D,
thymidine kinase genes: Paradigm for a prospective cancer con- Nicoll J, Harland J, Mabbs R, and Brown M. The potential for
trol strategy. Cancer Res 1986; 46: 5276–5281. efficacy of the modified (ICP 34.5(-) ) herpes simplex virus
133. Mullen CA, Kilstrup M, and Blaese RM. Transfer of the bacterial HSV1716 following intratumoural injection into human malig-
gene for cytosine deaminase to mammalian cells confers lethal nant glioma: a proof of principle study. Gene Ther 2002; 9:
sensitivity to 5-fluorocytosine: A negative selection system. Proc 398–406.
Natl Acad Sci USA 1992; 89: 33–37. 148. Pataer A, Fang B, Yu R, Kagawa S, Hunt KK, McDonnell TJ, Roth
134. Multiple Adenoviral Vector Safety and Toxicity. Human Gene JA, and Swisher SG. Adenoviral bak overexpression mediates
Therapy – Special Issue 2002; 13: 1–175. caspase-dependent tumor killing. Cancer Res 2000; 60: 788–792.
610 Cancer gene therapy

149. Pataer A, Smythe WR, Yu R, Fang B, McDonnell T, Roth JA, and 164. Rödicker F, Stiewe T, Zimmermann S, and Pützer BM.
Swisher SG. Adenovirus-mediated bak gene transfer induces Therapeutic efficacy of E2F1 in pancreatic cancer correlates with
apoptosis in mesothelioma cell lines. J Thorac Cardiovasc Surg TP73 induction. Cancer Res 2001; 61: 7052–7055.
2001; 121: 61–67. 165. Rodriguez R, Schuur ER, Lim HY, Henderson GA, Simons JW, and
150. Pawliuk R, Bachelot T, Zurkiya O, Eriksson A, Cao Y, and Henderson DR. Prostate attenuated replication competent adeno-
Leboulch P. Continuous intravascular secretion of endostatin in virus (ARCA) CN706: A selective cytotoxic for prostate-specific
mice from transduced hematopoietic stem cells. Mol Ther 2002; antigen-positive prostate cancer cells. Cancer Res 1997; 57:
5: 345–351. 2559–2563.
151. Pederson LC, Buchsbaum DJ, Vickers SM, Kancharla SR, Mayo 166. Rogulski KR, Kim JH, Kim SH, and Freytag SO. Glioma cells
MS, Curiel DT, and Stackhouse MA. Molecular chemotherapy transduced with an Escherichia coli CD/HSV-1 TK fusion gene
combined with radiation therapy enhances killing of cholangio- exhibit enhanced metabolic suicide and radiosensitivity. Hum
carcinoma cells in vitro and in vivo. Cancer Res 1997; 57: Gene Ther 1997; 8: 73–85.
4325–4332. 167. Rosenberg SA, Aebersold P, Cornetta K, Kasid A, Morgan RA,
152. Pederson LC, Vickers SM, Buchsbaum DJ, Kancharla SR, Mayo Moen R, Karson EM, Lotze MT, Yang JC, and Topalian SL. Gene
MS, Curiel DT, and Stackhouse MA. Combining cytosine deami- transfer into humans – immunotherapy of patients with advanced
nase expression, 5-fluorocytosine exposure, and radiotherapy melanoma, using tumor-infiltrating lymphocytes modified by ret-
increases cytotoxicity to cholangiocarcinoma cells. J Gastrointest roviral gene transduction. N Engl J Med 1990; 323: 570–578.
Surg 1998; 2: 283–291. 168. Roth JA and Cristiano RJ. Gene therapy for cancer: What have
153. Phillips MI, and Tang YL. Genetic modification of stem cells for we done and where are we going?. J Natl Cancer Inst 1997; 89:
transplantation. Adv Drug Deliv Rev 2008; 60: 160–172. 21–39.
154. Piche A, Grim J, Rancourt C, Gomez-Navarro J, Reed JC, and 169. Roth JA, Molldrem J, and Smythe WR. The current status of
Curiel DT. Modulation of Bcl-2 protein levels by an intracellular cancer gene therapy trials. PPO Updates: Prin Prac Oncol 1999;
anti-Bcl-2 single-chain antibody increases drug-induced cytotox- 13: 1–15.
icity in the breast cancer cell line MCF-7. Cancer Res 1998; 58: 170. Roth JA, Swisher SG, Merritt JA, Lawrence DD, Kemp BL,
2134–2140. Carrasco CH, El-Naggar AK, Fossella FV, Glisson BS, Hong WK,
155. Pirollo KF, and Chang EH. Targeted delivery of small interfering Khurl FR, Kurie JM, Nesbitt JC, Pisters K, Putnam JB, Schrump
RNA: approaching effective cancer therapies. Cancer Res 2008; DS, Shin DM, and Walsh GL. Gene therapy for non-small cell
68: 1247–1250. lung cancer: A preliminary report of a phase I trial of adenoviral
156. Podsakoff GM. Lentiviral vectors approach the clinic but fall p53 gene replacement. Semin Oncol 1998; 25: 33–37.
back: National Institutes of Health Recombinant DNA Advisory 171. Saleh M, Jonas NK, Wiegmans A, and Stylli SS. The treatment of
Committee review of a first clinical protocol for use of a lentiviral established intracranial tumors by in situ retroviral IFN-gamma
vector. Mol Ther 2001; 4: 282–283. transfer. Gene Ther 2000; 7: 1715–1724.
157. Ponnazhagan S, Curiel DT, Shaw DR, Alvarez RD, and Siegal 172. Saleh M, Wiegmans A, Malone Q, Stylli SS, and Kaye AH.
GP. Adeno-associated virus for cancer gene therapy. Cancer Res Effect of in situ retroviral interleukin-4 transfer on established
2001; 61: 6313–6321. intracranial tumors. J Natl Cancer Inst 1999; 91: 438–445.
158. Post DE, and Van Meir EG. A novel hypoxia-inducible factor 173. Sandig V, Brand K, Herwig S, Lukas J, Bartek J, and Strauss M.
(HIF) activated oncolytic adenovirus for cancer therapy. Oncogene Adenovirally transferred p16INK4/CDKN2 and p53 genes coop-
2003; 22: 2065–2072. erate to induce apoptotic tumor cell death. Nat Med 1997; 3:
159. Rainov NG. A phase III clinical evaluation of herpes simplex 313–319.
virus type 1 thymidine kinase and ganciclovir gene therapy as an 174. Scappaticci FA, Smith R, Pathak A, Schloss D, Lum B, Cao Y,
adjuvant to surgical resection and radiation in adults with previ- Johnson F, Engleman EG, and Nolan GP. Combination angiosta-
ously untreated glioblastoma multiforme. Hum Gene Ther 2000; tin and endostatin gene transfer induces synergistic antiangio-
11: 2389–2401. genic activity in vitro and antitumor efficacy in leukemia and
160. Ramesh R, Saeki T, Templeton NS, Ji L, Stephens LC, Ito I, solid tumors in mice. Mol Ther 2001; 3: 186–196.
Wilson DR, Wu Z, Branch CD, Minna JD, and Roth JA. 175. Schuler G, Schuler-Thurner B, and Steinman RM. The use of
Successful treatment of primary and disseminated human lung dendritic cells in cancer immunotherapy. Curr Opin Immunol
cancers by systemic delivery of tumor suppressor genes using an 2003; 15: 138–147.
improved liposome vector. Mol Ther 2001; 3: 337–350. 176. Schuler M, Herrmann R, De Greve JLP, Stewart AK, Gatzemeier
161. Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley U, Stewart DJ, Laufman L, Gralla R, Kuball J, Buhl R, Heussel
D, Brennan D, Petty R, MacLean A, Harland J, McKie E, Mabbs CP, Kommoss F, Perruchoud AP, Shepherd FA, Fritz MA,
R, and Brown M. Toxicity evaluation of replication-competent Horowitz JA, Huber C, and Rochlitz C. Adenovirus-mediated
herpes simplex virus (ICP 34.5 null mutant 1716) in patients with wild-type p53 gene transfer in patients receiving chemotherapy
recurrent malignant glioma. Gene Ther 2000; 7: 859–866. for advanced non-small-cell lung cancer: Results of a multicenter
162. Rancourt C, Rogers BE, Sosnowski BA, Wang M, Piché A, phase II study. J Clin Oncol 2001; 19: 1750–1758.
Pierce GF, Alvarez RD, Siegal GP, Douglas JT, and Curiel DT. 177. Shimada H, Matsubara H, Shiratori T, Shimizu T, Miyazaki S,
Basic fibroblast growth factor enhancement of adenovirus-medi- Okazumi S, Nabeya Y, Shuto K, Hayashi H, Tanizawa T, Nakatani
ated delivery of the herpes simplex virus thymidine kinase gene Y, Nakasa H, Kitada M, and Ochiai T. Phase I/II adenoviral p53
results in augmented therapeutic benefit in a murine model of gene therapy for chemoradiation resistant advanced esophageal
ovarian cancer. Clin Cancer Res 1998; 4: 2455–2461. squamous cell carcinoma. Cancer Sci 2006; 97: 554–561.
163. Regulier E, Paul S, Marigliano M, Kintz J, Poitevin Y, Ledoux C, 178. Small EJ, Carducci MA, Burke JM, Rodriguez R, Fong L, van
Roecklin D, Cauet G, Calenda V, and Homann HE. Adenovirus- Ummersen L, Yu DC, Aimi J, Ando D, Working P, Kirn D, and
mediated delivery of antiangiogenic genes as an antitumor Wilding G. A phase I trial of intravenous CG7870, a replication-
approach. Cancer Gene Ther 2001; 8: 45–54. selective, prostate-specific antigen-targeted oncolytic adenovi-
Donald J. Buchsbaum et al. 611

rus, for the treatment of hormone-refractory, metastatic prostate 192. Toda M, Martuza RL, and Rabkin SD. Tumor growth inhibition
cancer. Mol Ther 2006; 14: 107–117. by intratumoral inoculation of defective herpes simplex virus
179. Springer CJ, and Niculescu-Duvaz I. Prodrug-activating systems vectors expressing granulocyte-macrophage colony-stimulating
in suicide gene therapy. J Clin Invest 2000; 105: 1161–1167. factor. Mol Ther 2000; 2: 324–329.
180. Stackhouse MA, Buchsbaum DJ, Grizzle WE, Bright SJ, Olsen CC, 193. Todo T, Feigenbaum F, Rabkin SD, Lakeman F, Newsome JT,
Kancharla S, Mayo MS, and Curiel DT. Radiosensitization medi- Johnson PA, Mitchell E, Belliveau D, Ostrove JM, and Martuza RL.
ated by a transfected anti-erbB-2 single-chain antibody in vitro and Viral shedding and biodistribution of G207, a mutimutated, con-
in vivo. Int J Radiat Oncol Biol Phys 1998; 42: 817–822. ditionally replicating herpes simplex virus type 1, after intracere-
181. Stackhouse MA, Pederson LC, Grizzle WE, Curiel DT, Gebert J, bral inoculation in Aotus. Mol Ther 2000; 2: 588–595.
Haack K, Vickers SM, Mayo MS, and Buchsbaum DJ. 194. Todo T, Martuza RL, Rabkin SD, and Johnson PA. Oncolytic her-
Fractionated radiation therapy in combination with adenoviral pes simplex virus vector with enhanced MHC class I presentation
delivery of the cytosine deaminase gene and 5-fluorocytosine and tumor cell killing. Proc Natl Acad Sci USA 2001; 98:
enhances cytotoxic and antitumor effects in human colorectal and 6396–6401.
cholangiocarcinoma models. Gene Ther 2000; 7: 1019–1026. 195. Toyoizumi T, Mick R, Abbas AE, Kang EH, Kaiser LR, and
182. Stegman LD, Rehemtulla A, Beattie B, Kievit E, Lawrence TS, Molnar-Kimber KL. Combined therapy with chemotherapeutic
Blasberg RG, Tjuvajev JG, and Ross BD. Noninvasive quantita- agents and herpes simplex virus type 1 ICP34.5 mutant (HSV-
tion of cytosine deaminase transgene expression in human tumor 1716) in human non-small cell lung cancer. Hum Gene Ther
xenografts with in vivo magnetic resonance spectroscopy. Proc 1999; 10: 3013–3029.
Natl Acad Sci USA 1999; 96: 9821–9826. 196. Trepel M, Grifman M, Weitzman MD, and Pasqualini R.
183. Sterman DH, Treat J, Litzky LA, Amin KM, Coonrod L, Molnar- Molecular adaptors for vascular-targeted adenoviral gene deliv-
Kimber K, Recio A, Knox L, Wilson JM, Albelda SM, and Kaiser ery. Hum Gene Ther 2000; 11: 1971–1981.
LR. Adenovirus-mediated herpes simplex virus thymidine kinase/ 197. Tsukuda K, Wiewrodt R, Molnar-Kimber K, Jovanovic VP, and
ganciclovir gene therapy in patients with localized malignancy: Amin KM. An E2F-responsive replication-selective adenovirus
Results of a phase I clinical trial in malignant mesothelioma. targeted to the defective cell cycle in cancer cells: potent antitu-
Hum Gene Ther 1998; 9: 1083–1092. moral efficacy but no toxicity to normal cell. Cancer Res 2002;
184. Streit M, Stephen AE, Hawighorst T, Matsuda K, Lange- 62: 3438–3447.
Asschenfeldt B, Brown LF, Vacanti JP, and Detmar M. Systemic 198. Valery CA, Seilhean D, Boyer O, Marro B, Hauw JJ, Kemeny JL,
Inhibition of Tumor Growth and Angiogenesis by Marsault C, Philippon J, and Klatzmann D. Long-term survival
Thrombospondin-2 Using Cell-based Antiangiogenic Gene after gene therapy for a recurrent glioblastoma. Neurology 2002;
Therapy. Cancer Res (Suppl) 2002; 62: 2004–2012. 58: 1109–1112.
185. Sun Y, Finger C, Alvarez-Vallina L, Cichutek K, and Buchholz 199. Van Roy M, Wielockx B, Baker A, and Libert C. The use of tissue
CJ. Chronic gene delivery of interferon-inducible protein 10 inhibitors of matrix metalloproteinases to increase the efficacy of
through replication-competent retrovirus vectors suppresses a tumor necrosis factor/interferon gamma antitumor therapy.
tumor growth. Cancer Gene Ther 2005; 12: 900–912. Cancer Gene Ther 2007; 14: 372–379.
186. Suzuki K, Fueyo J, Krasnykh V, Reynolds PN, Curiel DT, and 200. Vile RG, Russell SJ, and Lemoine NR. Cancer gene therapy:
Alemany R. A conditionally replicative adenovirus with enhanced Hard lessons and new courses. Gene Ther 2000; 7: 2–8.
infectivity shows improved oncolytic potency. Clin Cancer Res 201. Viret C, and Lindemann A. Tumor immunotherapy by vaccina-
2001; 7: 120–126. tion with cytokine gene transfected cells. Int Rev Immunol 1997;
187. Suzuki S, Tadakuma T, Asano T, and Hayakawa M. Coexpression 14: 193–212.
of the partial androgen receptor enhances the efficacy of prostate- 202. Walker JR, McGeagh KG, Sundaresan P, Jorgensen TJ, Rabkin
specific antigen promoter-driven suicide gene therapy for pros- SD, and Martuza RL. Local and systemic therapy of human pros-
tate cancer cells at low testosterone concentrations. Cancer Res tate adenocarcinoma with the conditionally replicating herpes
2001; 61: 1276–1279. simplex virus vector G207. Hum Gene Ther 1999; 10: 2237–2243.
188. Swisher SG, Roth JA, Komaki R, Gu J, Lee JJ, Hicks M, Ro JY, 203. Weinberg RA. How cancer arises. Sci Am 1996; 275: 62–70.
Hong WK, Merritt JA, Ahrar K, Atkinson NE, Correa AM, 204. Wen XY, Bai Y, and Stewart AK. Adenovirus-mediated human
Dolormente M, Dreiling L, El-Naggar AK, Fossella F, Francisco R, endostatin gene delivery demonstrates strain-specific antitumor
Glisson B, Grammer S, Herbst R, Huaringa A, Kemp B, Khuri FR, activity and acute dose-dependent toxicity in mice. Hum Gene
Kurie JM, Liao Z, McDonnell TJ, Morice R, Morello F, Munden R, Ther 2001; 12: 347–358.
Papadimitrakopoulou V, Pisters KM, Putnam JB, Jr., Sarabia AJ, 205. Wong RJ, Patel SG, Kim S, DeMatteo RP, Malhotra S, Bennett
Shelton T, Stevens C, Shin DM, Smythe WR, Vaporciyan AA, JJ, St-Louis M, Shah JP, Johnson PA, and Fong Y. Cytokine gene
Walsh GL, and Yin M. Induction of p53-regulated genes and tumor transfer enhances herpes oncolytic therapy in murine squamous
regression in lung cancer patients after intratumoral delivery of cell carcinoma. Hum Gene Ther 2001; 12: 253–265.
adenoviral p53 (INGN 201) and radiation therapy. Clin Cancer Res 206. Worgall S, Wolff G, Falck-Pedersen E, and Crystal RG. Innate
2003; 9: 93–101. immune mechanisms dominate elimination of adenoviral vectors
189. Tepper JE. Combined radiotherapy and chemotherapy in the following in vivo administration. Hum Gene Ther 1997; 8: 37–44.
treatment of gastrointestinal malignancies. Semin Oncol 1992; 207. Worth LL, Jia S-F, Zhou Z, Chen L, and Kleinerman ES.
19: 96–101. Intranasal therapy with an adenoviral vector containing the
190. Thomas CE, Ehrhardt A, and Kay MA. Progress and problems murine interleukin-12 gene eradicates osteosarcoma lung metas-
with the use of viral vectors for gene therapy. Nat Rev Genet tases. Clin Cancer Res 2000; 6: 3713–3718.
2003; 4: 346–358. 208. Wu J, Zhao W, Zhong L, Han Z, Li B, Ma W, Weigel-Kelley KA,
191. Thompson TC, and Rodriguez R. Gene therapy prolongs PSA Warrington KH, and Srivastava A. Self-complementary recombinant
doubling time in prostate cancer patients. Molecular Therapy adeno-associated viral vectors: packaging capacity and the role of
2007; 15: 442–443. rep proteins in vector purity. Hum Gene Ther 2007; 18: 171–182.
612 Cancer gene therapy

209. Xia ZJ, Chang JH, Zhang L, Jiang WQ, Guan ZZ, Liu JW, Zhang viral vector and its application for the mitigation of toxicity in
Y, Hu XH, Wu GH, Wang HQ, Chen ZC, Chen JC, Zhou QH, Lu suicide gene therapy of gastrointestinal cancers. Mol Ther 2001;
JW, Fan QX, Huang JJ, and Zheng X. [Phase III randomized 3: 385–394.
clinical trial of intratumoral injection of E1B gene-deleted adeno- 217. Yamini B, Yu X, Pytel P, Galanopoulos N, Rawlani V, Veerapong
virus (H101) combined with cisplatin-based chemotherapy in J, Bickenbach K, and Weichselbaum RR. Adenovirally delivered
treating squamous cell cancer of head and neck or esophagus]. tumor necrosis factor-alpha improves the antiglioma efficacy of
Ai Zheng 2004; 23: 1666–1670. concomitant radiation and temozolomide therapy. Clin Cancer
210. Xiang J, Gomez-Navarro J, Arafat W, Liu B, Barker SD, Alvarez Res 2007; 13: 6217–6223.
RD, Siegal GP, and Curiel DT. Pro-apoptotic treatment with an 218. Yoshikawa T, Niwa T, Mizuguchi H, Okada N, and Nakagawa S.
adenovirus encoding Bax enhances the effect of chemotherapy in Engineering of highly immunogenic long-lived DC vaccines by
ovarian cancer. J Gene Med 2000; 2: 97–106. antiapoptotic protein gene transfer to enhance cancer vaccine
211. Xie X, Zhao X, Liu Y, Zhang J, Matusik RJ, Slawin KM, and potency. Gene Ther 2008; 15: 1321–1329.
Spencer DM. Adenovirus-mediated tissue-targeted expression of 219. Young A, and Kerr DJ. Genetic and immunological therapy for
a caspase-9-based artificial death switch for the treatment of cancer. J R Soc Med 2000; 93: 10–14.
prostate cancer. Cancer Res 2001; 61: 6795–6804. 220. Yu DC, Chen Y, Dilley J, Li Y, Embry M, Zhang H, Nguyen N,
212. Xu F, Ma Q, and Sha H. Optimizing drug delivery for enhancing Amin P, Oh J, and Henderson DR. Antitumor synergy of CV787,
therapeutic efficacy of recombinant human endostatin in cancer a prostate cancer-specific adenovirus, and paclitaxel and docetaxel.
treatment. Crit Rev Ther Drug Carrier Syst 2007; 24: 445–492. Cancer Res 2001; 61: 517–525.
213. Xu L, Pirollo KF, Tang W-H, Rait A, and Chang EH. Transferrin- 221. Yu DC, Chen Y, Seng M, Dilley J, and Henderson DR. The addi-
liposome-mediated systemic p53 gene therapy in combination tion of adenovirus type 5 region E3 enables calydon virus 787 to
with radiation results in regression of human head and neck xeno- eliminate distant prostate tumor xenografts. Cancer Res 1999;
grafts. Hum Gene Ther 1999; 10: 2941–2952. 59: 4200–4203.
214. Xu M, Kumar D, Stass SA, and Mixson AJ. Gene therapy with 222. Zacchigna S, Zentilin L, Morini M, Dell’Eva R, Noonan DM,
p53 and a fragment of thrombospondin I inhibits human breast Albini A, and Giacca M. AAV-mediated gene transfer of tissue
cancer in vivo. Mol Genet Metab 1998; 63: 103–109. inhibitor of metalloproteinases-1 inhibits vascular tumor growth
215. Yamabe K, Shimizu S, Ito T, Yoshioka Y, Nomura M, Narita M, and angiogenesis in vivo. Cancer Gene Ther 2004; 11: 73–80.
Saito I, Kanegae Y, and Matsuda H. Cancer gene therapy using a 223. Zhou Z, Zwelling LA, Kawakami Y, An T, Kobayashi K, Herzog C,
pro-apoptotic gene, caspase-3. Gene Ther 1999; 6: 1952–1959. and Kleinerman ES. Adenovirus-mediated human topoisomerase
216. Yamamoto M, Alemany R, Adachi Y, Grizzle WE, and Curiel DT. IIα gene transfer increases the sensitivity of etoposide-resistant
Characterization of the cyclooxyygenase-2 promoter in an adeno- human breast cancer cells. Cancer Res 1999; 59: 4618–4624.
20 Regulatory process for approval of biologicals
for cancer therapy
ANTONIO J. GRILLO-LÓPEZ

Abstract The tenet ‘Above all do no harm’ is necessary the expectation of a therapeutic benefit that will make
and important but not sufficient [1]. Today’s physicians their efforts worthwhile. Likewise, Oncologists have
have a responsibility to actively participate in making traditionally been more willing to accept a higher level
new therapies available to the cancer patient. We know of adverse events in treating their patients than physi-
that cancer can be cured. However, biologicals and cians in other specialties. In cancer therapy primum non
all new anticancer agents must be efficiently and nocere really implies a very delicate weighing of risk
rapidly evaluated to find those that are most promising. versus benefit where the balance weighs somewhat
They must then be incorporated into combination more heavily on the benefits side while accepting more
regimens that provide the greatest opportunity for cures. risk than one would accept for drugs in other therapeutic
Our regulatory agencies and processes were originally areas. A patient may be denied the possibility of a cure
established with the goal of protecting the patient because of therapeutic nihilism or therapeutic conservatism.
from unscrupulous investigators. Now it appears as if One extreme is the nihilistic approach that says – we
they exist to protect the cancer and not the patient. have nothing that can cure you and therefore we will
Where did we go wrong? What is the track record of the ‘watch and wait’. This approach, originally devised by
regulatory agencies? How can regulatory obstacles be Portlock and Rosenberg [2, 3] for patients with early
overcome? How can regulatory challenges be turned stage Follicular Non-Hodgkin’s Lymphoma (NHL) is
into opportunities? What is the optimal process for the today obsolete as we develop potentially curative com-
development of new anti-cancer therapies? bination regimens for this disease [4]. At the other
extreme is the urge to persist in administering one che-
Keywords Biological therapy • targeted therapy motherapy regimen after the next to a patient who is
• antibodies • rituximab • FDA • EMEA • regulatory refractory and at the latter stages of the disease.
• IND • NDA • BLA • ODAC • accelerated approval Hippocrates said, first – to help, and then (when you can
• regular approval • clinical trials • endpoints no longer help) – to do no harm.
Physicians are bound by a sacred oath to care for their
patients, to offer them the best that modern science
provides for their disease, complications, and general
The Doctor–Patient Relationship: wellbeing [5]. This duty, this responsibility, is critical to
Duty and Responsibility the patient with an incurable illness such as cancer. It is
even more important when the potential for cure exists
As to diseases, make a habit of two things – to help, or at least to
do no harm. for a cancer that was previously held to be incurable.
Hippocrates When that potential exists for a new agent still under
investigation, time is of the essence. Developing that
Primum non nocere (above all do no harm) is an impor- agent then becomes a race where the losers (patients)
tant consideration for the physician caring for the cancer will perish because that curative therapy took too long
patient [1]. The extreme, inaction for fear of doing harm, to be evaluated and approved. The physician directly
is simply not acceptable particularly when challenged caring for the patient must seek the most effective
by a potentially curable illness. In cancer therapy all therapies even when investigational and provide his
new agents present benefits and risks that must be patient with access to those via participation in clinical
weighed with the patient’s best interest in mind. In the trials or referral to centers where those therapies are
face of a serious illness like cancer, patients are generally available. This duty, this responsibility extends beyond
willing to take greater risks and accept more toxicity in the physician directly caring for the patient. Physicians in

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 613
© Springer Science + Business Media B.V. 2009
614 Regulatory process for approval of biologicals for cancer therapy

the pharmaceutical industry must do everything within place in the therapeutic armamentarium for the target
their reach to ensure that new agents are developed and disease. Regulatory agencies have no important role in
submitted for approval in the most expeditious fashion. this. Pharmaceutical industry can be a partner but should
Physicians in our regulatory agencies [Food and Drug play a secondary role. Thus, investigators in academia
Administration (FDA), European Medicines Evaluation and in the community (including academic institutions,
Agency (EMEA) and others] must likewise understand cancer centers, cooperative groups, consortia, and others)
that they have similar duties and responsibilities to the have very important duties and responsibilities to the
patient. Being employed by a bureaucracy does not patient in the development and optimization of combi-
mean that your sacred oath is no longer valid. In fact, nation regimens.
physicians in regulatory agencies could be an important
catalyst in making new promising therapies available to
patients in a timely fashion. However, the opposite has
occurred and as a result, regulatory agencies and their
Development of Cancer
bureaucratic processes are today the most important Therapeutics
impediment to the timely development and availability The mistakes are all there, waiting to be made.
of new (some of them potentially curative) anticancer Chessmaster S.G Tartakower
therapies. The patient has great expectations of his
personal physician but may not understand the role that Progress towards more effective and less toxic therapies
physicians in the regulatory agencies are playing in defining and progress towards curative therapies depends on the
their destiny. Regulatory agencies need to recognize that very complex process of drug development and approval.
new anti-cancer agents are not truly available to all that It is only after approval that a new agent will be widely
can benefit from them until approved and marketed. available to the patients who will benefit from its use.
Many types of expanded access and ‘compassionate However, even after a new agent is approved it is still
use’ programs have been tried but these will never necessary to continue the clinical research process that
substitute for approval and marketing. The fact is that, will determine its optimal use within a combination
prior to marketing, anti-cancer therapeutics are available regimen. On the average, it takes about 12 years for a new
only to a select minority of patients that qualify for agent to proceed from discovery to approval (Fig. 1).
these programs while the majority are deprived of this Preclinical development accounts for the first 4 or 5
opportunity. Thus, regulatory agencies have a very years; and clinical development, regulatory review and
serious responsibility to expedite the approval of new approval take over 7 years. It is thought that regulatory
anti-cancer agents and make them broadly available. It time accounts for 4 to 5 of these 12 years (FDA imposed
must be made clear that the majority of physicians in the delays, clinical holds, excessive and unreasonable
regulatory agencies are good professionals and well requirements, waiting for meetings to be scheduled,
meaning individuals who work very hard for less than review time, waiting for FDA responses, etc.).
adequate salaries. They deserve respect and support. It The clinical development process begins prior to the
is a minority that, strategically placed, has created and first patient being treated. The Phase I protocols must be
enforced the obstacles that impede progress. written, a clinical plan defined, and all preclinical data
However, single agent approval although necessary submitted to the regulatory agencies before one can pro-
and important is not sufficient as the majority of effec- ceed. All of these documents are reviewed by the regu-
tive therapies (and specifically, curative therapies) are latory agency and a determination is made as to whether
not based on single agents but on combination regimens or not the Phase I (PI) clinical trials can be initiated.
[6–9]. The regulatory agency’s responsibility, as estab- Once a recommended dose for PII has been established
lished by law, is the review and approval of new anti- in PI, the PII trials can commence. Ideally this occurs
cancer agents (single agents). Pharmaceutical industry with some overlap with the PI studies. Both the PI and
is responsible for the discovery and development of new the PII trials should include, as early as possible, combi-
single agents and this is what the agency is required to nation regimens. These pilot combination studies are
regulate. Once the single agent is approved, it is the not necessary for the evaluation of the safety profile or
oncology community’s responsibility to find its optimal the activity of the single agent; however, they are impor-
use in combination with other agents. The oncology tant for two reasons: (a) getting an early start towards
community is then responsible for defining the role of treatment optimization within a combination regimen,
the new agent within a combination regimen and its and (b) evaluating possible combinations suitable for
Antonio J. Grillo-López 615

Figure 1. Timeline: New anticancer agents

PIII studies. It is unusual to find the optimal combina- the shortage of well qualified and experienced physi-
tion regimen this early in development. Following PII, cians to assume the role of project clinician (or of Chief
the PIII trials are then carried out with a view to regula- Medical Officer, CMO) in a pharmaceutical company. It
tory approvals. used to be that any physician considering such a role
The clinical development plan incorporates all activi- would unequivocally be given the message, by his peers
ties from PI to approval and beyond. It is usually written in academia, that this would be a black mark on his cur-
by a physician (project clinician) with support from other riculum vitae and that his career might not survive that
professionals. The extent and overall scope of this criti- experience. This has changed over time and today there are
cal road map depends heavily on projections of the many more qualified and experienced physicians, many
resources that will be available over the 6 to 10 years it with an academic background, in the pharmaceutical
will take to complete clinical development. Most new industry. However, over time, the need has also increased.
agents are developed by pharmaceutical companies. As an example, in San Diego, California there are over
A large and well established company is more likely to 400 bio-medical companies. They all need a CMO and
dedicate substantial resources than a small start-up bio- most also need one or more project clinicians depending
technology company that depends, at least initially, on on how many new agents they have in clinical trials.
venture capital. Regardless of available resources, the There are simply not enough physicians with clinical trials
clinical development plan must clearly define the path to and regulatory expertise to satisfy the need.
a regulatory approval. Given a new agent with reasonable Simple mistakes can have major impact in the lengthy
efficacy and tolerable toxicity, this is the main objective. and complex process of cancer drug development. As an
It is the objective not only for the pharmaceutical company example, one small company contracted with a contract
and its investors but also for the patient and all who have research organization (CRO) to have them create a data-
a role in the care of the patient. The road to therapeutic base, perform data entry, and carry out the statistical anal-
success in cancer is paved with many compounds that ysis for one of their clinical trials. Despite the company’s
failed to show the desired efficacy or were too toxic. One extraordinary efforts to make sure that the CRO’s database
must, however, wonder how many active compounds was identical to the company’s database for their other
were trampled under and became part of that pavement studies, one simple mistake was made. Due to this, the
due to simple mistakes, inappropriate development summary data tables for safety and efficacy cutting across
plans, technical problems in the conduct of clinical trials, all studies were very obviously incorrect. After extensive
companies going bankrupt, inadequate resources, misin- investigation (double checking, and painstaking review of
terpretation of clinical data, and/or any combination of source documents, data entry, programming, queries, dic-
the myriad problems and challenges that plague the clin- tionaries, coding conventions, quality control, and of all
ical phases of development. One important problem is the data), it was determined that the CRO had reversed the
616 Regulatory process for approval of biologicals for cancer therapy

coding conventions followed by the company for gender. The rationale is relatively simple. The intent is to
This one simple mistake cost the company a 2 months make the patient’s immune system an ally. Not to impair
delay (or $3.0 million for this particular company) in filing it but to find ways of working with and enhancing the
the regulatory dossier. At the other extreme (more serious immune system. This is readily understood in the case
mistakes) some new agents have been turned down by the of MAbs. They work either by utilizing the patient’s
FDA for incomplete or inadequate data collection. When immune system (complement, effector cells, etc.) to kill
this happens at the end of PIII after many years of devel- the tumor cell or by binding to the target antigen and
opment, the losses are measured in the hundreds of mil- transmitting an apoptotic signal. MAbs are also used
lions and more importantly in thousands of lives when an as per Ehrlich, as ‘magic bullets’. They are used to
effective product (with some curative potential) is lost to deliver to the tumor cell a payload consisting of a
the cancer patient. toxin, chemotherapeutic agent, or a radioisotope. Other
Some may marvel at how well we have performed in biological therapies, in the broad definition of the term
terms of cancer drug development over the past few (treatments that are or resemble natural body substances
decades given all the challenges, mistakes and prob- or that are made from natural body substances), have
lems. Such complaisance is unacceptable. I would prefer very specific molecular targets (targeted therapies).
to tackle the challenges and problems, avoid/preempt The end of the last millennium ushered in a new era in
the mistakes and find solutions that will enable us to cancer therapeutics. MAbs, led by rituximab, had arrived.
perform at peak efficiency as we head towards better Some other MAbs, as well as a variety of other targeted
and hopefully curative therapies. We owe that and more therapies, are now approved (Herceptin, Mylotarg,
to the patients that we serve. Campath, Zevalin, Bexxar, Erbitux, Avastin) and many
are under investigation. We witnessed the development
and approval of the first MAbs for the treatment of can-
cer, rituximab [12, 13]. Early in this new millennium, we
Biologicals and the Treatment witnessed the approval of the first radio immunotherapy
[14, 15]. Other firsts will soon follow: the first vaccine for
of Cancer cancer, the first antisense product, the first genomics-
derived therapeutic, and others. This will be the era of
Rationale for Developing Biological MAbs, of biologicals, of targeted therapies. Gone are the
Therapies days when chemotherapy was the only and, unfortunately,
According to the theory of aerodynamics, as may be readily dem- usually the last resource. These new therapeutics open the
onstrated through wind tunnel experiments, the bumblebee is door for new combination regimens and new multimo-
unable to fly. But the bumblebee, being ignorant of these scientific dality therapies with curative potential. We now have much
truths, goes ahead and flies anyway – and makes a little honey
every day!
more selective and specific therapeutics. We now have
Ross E. Hutchins agents with very different mechanisms of action that are
Back in the late 1980s and early 1990s, antibodies ideal for combination therapies. Importantly, never before,
(MAbs) were the bumblebees of cancer therapeutics. in the history of cancer therapy, have we been able to har-
Practically no one, after years of frustrating clinical ness, control, and direct the immune system as we can
experiences, believed that MAbs would ever fly. The today. The immune system can now become an important
1994 review by Dillman listed the many MAbs taken and effective partner in our efforts to cure cancer. It is
to clinical trials with disappointing results [10]. only the beginning. There is a bright future ahead for the
Frustration and disappointment were understandable. many cancer patients who will benefit from all the prom-
After all, it had been nearly 100 years since Paul ising advances that we envision today and that will
Ehrlich first spoke about MAbs and ‘magic bullets’ become reality in this new era.
[11]. And yet some of us knew, even then, that har-
nessing the power of MAbs for therapeutic applica- Rituximab as an Example
tions was a question of time [7]. Anticancer therapies,
The human mind treats a new idea the way the body treats a strange
such as radiotherapy and chemotherapy, had been the protein, it rejects it.
standard for too many years, as we sought additional P. B. Medawar
and better alternatives. They had served us well; how-
ever, their toxicity was tolerated, only because we The development of rituximab began at IDEC Pharma-
lacked other options. MAbs had arrived; it was just a ceutical Corporation’s (IDEC) laboratories in San Diego,
question of time! California in 1991. Mice were injected with human
Antonio J. Grillo-López 617

CD20 antigen and the anti-CD20 MAbs they produced was doing what it was intended to do. Significant reac-
were isolated and characterized. One of those, the tions (primarily fever and chills) were seen even in the
murine IDEC-2B8, was chosen for chimerization. The first patient treated. These were biologic manifestations
chimeric MAb (IDEC-C2B8) was favored in order to of MAb induced B-cell depletion. The total absence of
reduce the immunogenicity of the murine parent MAb, such reactions might indicate the MAb was not effec-
to utilize a human framework which allowed for com- tive. Two patients had objective responses, to a single
plement and effector cell binding, and to selectively infusion of MAb, which lasted for months [19]. The PI/
effect B-cell depletion. The technology necessary for II study was designed to define the pharmacokinetic
humanization was not yet fully developed at that time profile [20], confirm the dose to be used in a four infu-
and thus was not pursued. A vector was engineered that sion schedule [21] and to establish a response rate in
enabled high MAb yields when inserted in CHO cells patients with relapsed or refractory Non-Hodgkin’s
and used in a high volume fermentation process. The Lymphoma (NHL) [22, 23]. The PIII (pivotal trial) com-
appropriate preclinical pharmacology and toxicology pleted enrollment in 1 year, the data was collected and
studies were carried out including B-cell depletion studies analyzed, and the complete filing (including all preclini-
in monkeys. Mechanistic studies confirmed the desired cal and clinical studies) was submitted simultaneously
complement binding, complement dependent cytolysis to the US FDA (electronic and paper copy) and in
(CDC), effector cell binding and antibody dependent Europe to the EMEA in February 1997 [24–27]. The
cellular cytotoxicity (ADCC). The MAb was clearly clinical work including the design of the overall clinical
shown to induce apoptosis. All of this pre-clinical work plan; writing and implementing all of the protocol for
was carried out within a year at IDEC’s laboratories [16, the initial eight studies; establishing collaborations with
17] (Table 1). Synergism studies were completed in col- numerous academic institutions in North America and
laboration with scientists at UCLA [18]. All the pre- Europe; collecting, analyzing and interpreting the data;
clinical development work was performed at IDEC’s writing all reports, abstracts and publications; generating
laboratories in San Diego, California with no govern- the electronic dossier; filing, presenting, and defending
ment or NIH support. This MAb was ‘made in the USA’, the data with regulatory agencies in the US and Europe;
at a small biotechnology company, a start up funded by and many other activities critical to the ultimate success
venture capital, and with no taxpayer’s monies. Rituximab of this project were carried out by the Medical and
is an example of American entrepreneurship at its best. Regulatory Affairs Division at IDEC [7] (Table 2). The
IDEC-C2B8 was ready for clinical trials by late 1992. clinical phase of development was completed in 3 years
The Investigational New Drug application (IND) was (first patient enrolled in Phase I to last patient enrolled
submitted to the FDA who placed the project on a clinical in PIII), a new record in NHL as even the NCI’s coop-
hold due to their concerns about the effect of B-cell deple- erative groups will take at least 3 years to complete one
tion on immunoglobulin levels and immunocompetence. PIII study. It took 1 year (total time from last patient
Finally, after months of scientific discussions, the FDA enrolled in PIII to BLA filing) to complete the necessary
allowed the PI clinical trials to proceed (February 1993). observation time on the Phase III patients, and prepare
The desired B-cell depletion was observed. The MAb and file the regulatory dossier (same day filing of a US
BLA and an European dossier). Regulatory review and
approval, in the US, took 6 months plus a 3 months delay
Table 1. IDEC’s pre-clinical scientists responsible for the
imposed by the FDA while resolving manufacturing
development of rituximab (IDEC-C2B8)a
issues. Clinical/regulatory development time was a record
Name Responsibilities setting 4 years and 9 months as compared to the average
Nabil Hanna, Ph.D. VP, Pre-Clinical R&D – in charge of of over 7 years for cancer drugs in general (Fig. 2).
all preclinical development The five things that made this clinical development
Darrel Anderson, Ph.D. Isolation and characterization of the
murine parent antibody, IDEC-2B8
program successful were: an active agent, the right people,
Roland Newman, Ph.D. Gene identification and cloning good leadership, ironclad data and peer level relationship
experiments; antibody engineering with the FDA. Clearly rituximab was bound to be suc-
Mitchell Reff, Ph.D. Engineering of the chimeric antibody cessful. It worked exactly as it had been designed. IDEC
IDEC-C2B8 and vector engineering had the right people empowered by good leaders. The
Chris Burman, Ph.D. VP, Manufacturing – in charge of
antibody production company had a core group of professionals who had years
of experience in clinical development. They were not
a
Key scientists. Many others at IDEC contributed and are not inventing the wheel. IDEC was not a ‘virtual company’
listed for space reasons. outsourcing a lot of work. It was a small company and yet
618 Regulatory process for approval of biologicals for cancer therapy

Table 2. IDEC’s clinical scientists responsible for the development of rituximab (IDEC-C2B8)a
Name Responsibilities
Antonio J. Grillo-López, M.D. Chief Medical Officer and Senior VP Medical and Regulatory Affairs – in charge of all clinical
development and regulatory affairs
Brian K. Dallaire, Pharm. D. Senior Director, Clinical Operations – conduct of clinical trials including strategy, timelines, budgets
and resources
John Leonard, Ph.D. Senior Director, Project Planning and Regulatory Affairs – planning and timelines for all clinical
trials; communications with the regulatory agencies; filings and dossiers
Anne McClure, M.S. Director, Medical Writing – documentation (protocols, clinical trial reports) for each study,
regulatory filings (INDs, periodic reports, regulatory dossiers); publications
Jay Rosenberg, Ph.D. Head, Clinical Immunology Laboratory – analysis of all patient samples for bcl-2 (pcr), pharmacoki-
netics; and other specialized tests
David Shen, Ph.D. Senior Director, Biometrics – data entry, programming, queries, tracking, statistical planning and
strategy, data analysis, interaction with regulatory agency’s statisticians
Chester Varns Director, Clinical Trials Monitoring – protocol design, study implementation, clinical trial monitoring
and GCP, data collection and clean-up, interaction with clinical sites
Alice Wei Senior Director, Regulatory Affairs - planning and strategy for regulatory evens, communications
with regulatory agency’s staff; filings and dossiers
Christine White, M.D. Senior Director, Hematology and Oncology – safety officer; interactions with clinicians and their
staff at study sites; publications and presentations

a
Many others at IDEC Pharmaceuticals, as well as investigators and staff at investigational sites, made important contributions and
are not listed due to space constraints.

Figure 2. Rituxan: Clinical development


timeline

able to handle the work load for this program mostly inter- ever contested by reviewers, auditors, peers, or editors.
nally. Virtual companies generate virtual data. That was not The FDA contested only one patient, a patient in the
the case at IDEC. The clinical program was subject to Phase III study that had been classified as inevaluable. The
careful prospective planning, meticulous study imple- result of the joint review by he FDA and the clinical group
mentation and conduct, rigorous adherence to GCP and at IDEC was that the patient’s classification was revised to
GLP requirements, strict auditing procedures, precise sta- evaluable, response was a CR, and duration was long.
tistical methodology, and painstaking attention to correct The FDA had actually added a CR and increased the
response and duration adjudication. The clinical data, the overall response rate (not their original intent). Lastly, a
cornerstone critical to the approval of any new agent, was peer level relationship with the FDA is of the utmost
ironclad. Not a single patient’s response or duration was importance. Establishing a relationship that is amicable,
Antonio J. Grillo-López 619

collegial, professional and based on mutual respect is the progression free survival (PFS) at 5 years] [30, 31].
key to success. It requires openness, timeliness, and the These studies led to the GELA study, led by Bertrand
most professional and scientific approach. In such a situ- Coiffier, which showed that the R + CHOP combination
ation the FDA staff can be your allies and partners. Ideally almost quadrupled the event free survival of CHOP [32].
you end up with what amounts to a joint development This study established R + CHOP as the gold standard
team as happened with the rituximab CALA (Table 3). for the treatment of DLBCL. Rituximab had reached its
On a personal note, there was one new idea in the curative potential in combination with CHOP. No other
development of rituximab that was rejected by several agent, alone or in combination, has ever achieved a sig-
prominent lymphoma experts. I knew that rituximab’s nificant improvement over CHOP alone.
ultimate contribution to the care of the lymphoma
patient would be determined by its contribution within a
curative combination regimen such as CHOP in the
treatment of Diffuse Large Cell B-Cell Lymphoma The USA’s Food and Drug
(DLBCL). Thus, one of the first clinical trials I designed
was the Phase II pilot study of rituximab plus CHOP
Administration
Want of foresight, unwillingness to act, lack of clear thinking, con-
(R + CHOP) [28]. The definitive goal was to treat patients
fusion of counsel – these are the features which constitute the endless
in a disease where CHOP was known to be curative repetition of history.
(about 40% cure rate). Synergism between rituximab Sir Winston Churchill
and doxorubicin had already been shown [29]. However,
the toxicity of the combination and its impact on the The FDA was created in 1938 by an act of congress
cure rate of CHOP were unknown. Therefore, we chose (FDAC Act) and charged originally with protecting the
to perform the first study of R + CHOP in patients with consumer by evaluating the safety of new products [33].
Low Grade NHL. Several prominent lymphoma experts The role of the FDA has evolved over time. The act was
rejected this new idea of the simultaneous administra- revised in 1962 and thereafter the FDA required that,
tion of rituximab and CHOP as they might reject a foreign in addition to safety, efficacy be established in two
protein. Their objection was that ‘antibodies have lim- adequate and well-controlled clinical trials. In 1997,
ited efficacy and should be used only after chemother- the FDA’s Modernization Act (FDAMA) was approved
apy, to treat minimal residual disease’. They argued for and it allowed the FDA to accept one clinical trial and
sequential rather than simultaneous administration. My other supportive studies as evidence of efficacy [34].
new idea, that the synergism shown in cell lines would However, in practice the FDA generally continues to
lead to enhanced efficacy in clinical trials and that we operate as if efficacy should be shown in ‘two adequate
would miss that opportunity if we gave the antibody in and well controlled clinical trials’ and as if the only
sequence rather than simultaneously with chemotherapy, acceptable endpoint is overall survival. One unwritten
was not considered valid. One investigator did accept rule is that short of the ‘two adequate and well con-
my new idea as valid (did not reject it as a foreign protein) trolled clinical trials’ the one Phase III study would have
and enthusiastically participated in this study. to reach high statistical significance for its primary end-
The study revealed a 100% response rate (7+ years point at the p = 0.01 level (rather than 0.05 in the case of
median TTP). Side effects were similar to those of CHOP two studies). The presence of good supportive trials is
and rituximab with no additive toxicity. We conducted a of no consequence in this situation. Any application
similar study in DLBCL and found a 94% ORR [82% needs to contain ‘two adequate and well controlled clinical
trials’ that show significance at the p = 0.05 level or one
Table 3. Rituximab’s CALA Team (1996–97) that that shows significance at the p = 0.01 level.
Responsibility FDA IDEC The acts of Congress that created and, over the years,
Medical Bernard Parker, M.D. Antonio have modified the FDA and its mandates have been
Grillo-López, M.D. interpreted by the agency in their own manner. These
Susan Jerian, M.D. – interpretations are often issued in the form of guidelines
Patricia Keegan, M.D. – which are not always followed by the FDA itself. All of
Regulatory – Alice Wei this has evolved into a bureaucratic process that is com-
– Augusta Cerny
Product Reviewer Mark Brunswick, Ph.D. – plex, costly and time consuming. Each subsequent law,
Statistical Jawahar Tiwari, Ph.D. David Shen, Ph.D. each subsequent guideline is one more band aid on a
Technical Support Robin Jones Ken Fite gaping wound that in reality can only be addressed by
CALA Coordinator Michael Fauntleroy – major reconstructive surgery. A few within the agency
620 Regulatory process for approval of biologicals for cancer therapy

contribute to this bureaucracy. The majority try to do dar days of the committee’s recommendation. However,
their best to work within this flawed system. Leadership the FDA requests the committee’s advice via a series of
is a major problem. The agency has for long periods of questions that are delivered to committee members usu-
time operated under interim leaders. The commissioner ally the day before the meeting [38, 39]. The list does not
of the FDA is appointed by the President and must be usually include the key question – should this product be
sanctioned by Congress. This is a highly political, drawn approved; or, more appropriately, will this new agent
out process for a job that few want. In the absence of true benefit the patients who may receive it. The questions
leadership, the agency falls back on the more primitive usually bring up technical issues regarding study con-
instincts of self protection. Any publicity is bound to be duct, data collection, choice of endpoints, or statistical
negative and any misstep can affect their Congress analysis. The FDA’s desire for the committee to provide
approved budget. Any action taken by the FDA will be a certain answer or recommendation is many times trans-
subject to intense scrutiny and criticism. Adverse events parent, equivalent to a jury being asked – do you find this
occurring post approval will be assumed to have hap- guilty person guilty. Sometimes a question is asked in a
pened because of the FDA’s negligence in scrutinizing negative way that could serve to lead the committee to
safety data. In this environment it is easier to say no or respond negatively. Many times questions are asked that
not to act at all. Nevertheless, FDA reform is a buzzword require an in-depth understanding of regulatory law at
that one hears mostly from politicians prior to elections. which committee members are not experts.
After that, it seems like no one dares tackle this subject. One could argue that the committee is set up to fail.
The members are academicians with a wealth of aca-
demic knowledge. There is not a single community
Oncologist on the committee. In the US, the majority of
The Oncologic Drugs Advisory cancer patients are treated in the community, not at aca-
Committee of the FDA demic centers. Members have experience mostly with
Phase II carried out in their own institution and Phase III
Life is short, Art is long, Opportunity is fleeting,
Experience is deceitful, Judgment is difficult.
trials conducted by the cooperative groups. Not one com-
Hippocrates mittee member has ever been responsible for taking a
new agent all the way through clinical development
The US Congress has enacted legislation authorizing (Phase I to III) and to an approval. Not one committee
the establishment of committees of experts to advise member has ever had to produce, negotiate, file and
government agencies in discharging responsibilities. defend a regulatory dossier submitted to the FDA and had
The US Food and Drug Administration (FDA), in turn, that product approved. The one member of the committee
created the Oncologic Drugs Advisory Committee with this kind of experience is the Industry Representative
(ODAC) whose structure and function is defined in the who has a voice but not a vote. Committee members are
Code of Federal Regulations (CFR), in the charter pub- not trained on their role at ODAC or on regulatory law.
lished by the FDA, and other documents [35–37]. There is information on an FDA website but no active
According to the charter, the committee is expected to training is provided. Members are not allowed to meet in
“review and evaluate data concerning the safety and the absence of an FDA representative. Why not? This
effectiveness of marketed and investigational human would allow for an exchange of experiences and ideas
drug products for use in the treatment of cancer and that could only have positive results. ODAC’s recom-
make appropriate recommendations to the Commissioner mendations are just that, the agency can accept them or
of Food and Drugs”. not. They should probably be mandatory. After all, the
The FDA requests advice from the committee members agency brought the agent in question to the committee
(members) on a variety of matters, including various because they felt that en expert recommendation was
aspects of clinical investigations and applications for required. Why should they then decide not to follow that
marketing approval of drug products. They receive sum- recommendation? Members do not see all of the applica-
mary information about the applications and copies of tions that are filed with the FDA. The agency selectively
the FDA’s review of the application documents. Based presents those where there are issues that require the
on this information, the committee may recommend committee’s expertise. How do we know that there were
approval or disapproval of a drug’s marketing applica- no similar issues with the other applications? Why can’t
tion. The FDA generally follows the committee’s recom- the committee learn from the discussion of those other
mendation, but is not bound to do so. The FDA must applications? What precedents are set in the approval or
notify affected persons of their decision within 90 calen- denial of applications never seen by the committee?
Antonio J. Grillo-López 621

Wouldn’t it be better for the committee to review all The BLA is filed with the FDA upon the completion of
applications? For these and many other reasons judgment Phase III after the necessary patient observation period.
is difficult. And yet, the FDA makes it look like their Completing all of the documentation necessary for the
questions are reasonably addressed by the committee as if BLA will take an average of 6 to 8 months from last
judgment were simple. Each committee member should patient observation date (last patient last visit) or, depend-
ask – am I being manipulated by the FDA? Actually, ing on the primary endpoint, from the median patient’s
judgment could be simple as will be shown below. last visit. The FDA then has a certain period of time to
review and approve the application. In the case of an
application for accelerated approval that might be 6
months. Nevertheless, the FDA can ‘stop the clock’ at any
Regulatory Review and time or occasionally even ‘restart the clock’. The agency
Approval Process can also request that the new agent be presented to ODAC.
There are risks and costs to a program of action. But they are far
Again, a long and drawn out process that favors inaction
less than the long-range risks and costs of comfortable inaction. over action. There are numerous possibilities for improve-
John F. Kennedy ment. An action oriented process is sorely needed.
A different way of looking at the regulatory review
The current process for preclinical development entails and approval process is by considering its overall place
meeting all regulatory requirements (for pharmacology, in the larger scheme leading to treatment optimization.
toxicology, and manufacturing), filing reports on all the It is through combination therapies that patients get the
data on an ongoing basis and having periodic meetings most benefit. The optimal role of any new agent within
with the agency. At the end of this process following a combination regimen takes many years to define.
another meeting with the FDA, the IND is filed. The Nitrogen mustard was available and used effectively as
first clinical trial can begin after a mandatory 30 days a single agent (despite its toxicity) for over 20 years
wait provided that the agency does not impose a clinical before its role in the MOPP regimen was defined and
hold. During clinical development periodic meetings found to constitute a curative therapy for Hodgkin’s
are held with the FDA, data on all studies is filed (clinical Disease (Fig. 3). Many patients benefited from Nitrogen
study reports), study conduct is subject to GCP and end Mustard during those 20 years. Thus, one could envi-
of Phase II or pre-licensing application (BLA or NDA) sion a process that begins with discovery, followed by
meeting is scheduled. At this meeting all plans for the preclinical development, clinical development, regula-
content of the application (BLA) are presented and tory review and approval, and reaching completion with
discussed with particular emphasis on the protocol, treatment optimization. The FDA’s role is modest when
endpoints, and statistical analysis. The company may viewed from this perspective. Their responsibility should
also submit a Special Protocol Assessment (SPA) where only be that of ensuring the safety of new anticancer
the FDA reviews the Phase III protocol and an agree- agents and thus limited to the review and approval of
ment is reached as to its validity for approval purposes data generated during clinical development. Everything
(given the appropriate results). Phase III is then imple- else they regulate today is really the responsibility of the
mented and conducted meeting all GCP requirements company developing the new agent (discovery to approval)
and with the appropriate audits during and at the end of or of the oncology community (approval to completion
the study. Nowadays, most Phase III studies will include of treatment optimization) (Fig. 4).
provisions for a Data Safety Monitoring Board (DSMB)
or a Data Safety and Efficacy Monitoring Board
(DSEMB), a mechanism for histopathological review Accelerated Versus Regular Approvals
of biopsies and a mechanism for third party clinical Take calculated risks. That is quite different from being rash.
review and classification of responders or of radiological George S. Patton
exams. Procedures for blinding and for reporting are
carefully defined. Interim analyses require additional The two mechanisms for the approval of new anticancer
rigorously documented processes. agents of approval are (1) Regular Approval and (2)
The cost of all of this has risen exponentially over the Accelerated Approval. Requirements for Regular
past few years. It is said that the cost of bringing one Approval include evidence of clinical benefit as mea-
new drug to market exceeds a billion dollars and in a sured by endpoints reflecting improved quality or quan-
Phase III solid tumor study the per-patient, fully allo- tity of life or the use of established surrogate endpoints
cated, costs can exceed $100,000.00. for clinical benefit such as survival, disease free survival,
622 Regulatory process for approval of biologicals for cancer therapy

Figure 3. Nitrogen mustard

Figure 4. New anticancer agents:


Optimization

and improvement in tumor-related symptoms. In con- vided by these studies, and approval can be withdrawn
trast, Accelerated Approval requirements allow the use depending on their results.
of surrogate endpoints reasonably likely to predict clini- Accelerated approval was instituted because Regular
cal benefit (for example, durable response) provided Approvals took too long and there was a desire to bring
there is “substantial evidence” of efficacy from well- new agents earlier to the patients who needed them.
controlled clinical trials [40, 41]. This mechanism is Clearly there is an element of risk as Accelerated
specifically for situations involving “serious and life- approval occurs earlier when data is not as mature as for
threatening” illness where patients’ diseases are unre- a Regular approval. Regular Approval, however, signi-
sponsive or patients are intolerant of available therapies fies that large numbers of patients may have to go
(i.e. medical need). The new therapy must provide an untreated because an effective new agent is not avail-
advantage over existing therapies. Importantly, an addi- able. Accelerated Approval implies a calculated risk.
tional requirement is the implementation of post- This risk is minimized when safety is acceptable and
approval, confirmatory studies. The FDA’s approval is when mature data will be available in a reasonable
contingent on subsequent confirmation of efficacy pro- period of time.
Antonio J. Grillo-López 623

The FDA, as will be seen in 8.0 below, has a less than happened only exceptionally. There is some risk but
desirable track record with Accelerated Approvals. They this is the risk that the Accelerated Approval mandate
are bound by their own regulations to accept this mech- recognized and intended be taken. Most Accelerated
anism and yet they oppose it and have impeded its use. Approvals will make useful new agents available
A recent example is the case of satraplatin. This is an earlier. A minority will later fail to have a confirmed
oral platinum compound that was presented at the advantage and will need to be withdrawn. The FDA
ODAC meeting of July 2007. Satraplatin has been eval- is fully authorized to withdraw approvals in such cases.
uated in a large Phase III clinical trial (randomized, (c) The FDA could ask the question of ODAC and
double blind) for the indication of androgen indepen- thus shift the responsibility and the possible negative
dent Prostate Cancer in patients who have failed at least fallout from the agency to the committee, and this is
one prior chemotherapy regimen [42]. The company what they chose to do.
applied for and obtained an SPA (reviewed and approved 3. What response was sought? The FDA must have
by the FDA). They then applied for an Accelerated thought that there was some merit to this new
Approval based on the protocol and SPA defined agent; otherwise they would have denied the applica-
endpoint, progression free survival (PFS), for an interim tion outright. However, it was much less risky for
analysis. The median PFS was 9.7 weeks for the control them to wait until there was confirmatory OS data.
arm (prednisone) and 11.1 weeks for the satraplatin + They wanted the committee’s support and wanted
prednisone arm. This 15% increase in PFS is a modest the committee to be responsible for their decision to
but statistically significant improvement in PFS and wait. The ODAC was manipulated by the FDA to
valuable to the patient that has already failed at least one achieve their desired end result.
chemotherapy regimen. The FDA asked ODAC several
Unfortunately, all of this means that satraplatin will not
questions, some were endpoint related technical issues
be available for approximately another 2 years even if
and the last was whether or not they should wait until
all goes well with the re-submitted application (based
completion of the study and availability of OS data for
on OS data). This does not hurt the FDA, the agency
consideration of the application. At the end of the day,
doesn’t seem to care and doesn’t want to take the risk.
the committee voted only on this last question agreeing
This is not understood by committee members, they
unanimously that the FDA should postpone any deci-
have been manipulated before. Most of the media mis-
sion on the application pending completion of the Phase
interprets the situation. The public is biased against
III study and availability of OS data [43]. I believe that
pharmaceutical industry because of high drug prices and
the consequences of this decision are not well under-
any setback for a company is not viewed negatively. It is
stood by the committee, the media or the public. The
only the patients who could benefit from satraplatin who
situation is as follows:
will have to wait and suffer. It is difficult to understand
1. The question was totally inappropriate. The commit- how the availability of an oral platinum, the first of its
tee is not there to rule on whether an Accelerated or kind, could be anything other than a positive outcome.
a Regular Approval should be considered. This is a The oncology community should have the opportunity
regulatory issue to be decided by the FDA. The to evaluate satraplatin in different combinations and
application was for an Accelerated Approval and that find the optimal treatment regimen.
is what the FDA should have asked the committee –
is there a medical benefit to patients and should this
drug be granted Accelerated Approval. This question Clinical Trial Endpoints
needed to be addressed independently of the avail-
We will never have all the facts to make a perfect judgment, but
ability, at some later point, of OS data.
with the aid of basic experience we must leap bravely into the
2. Why was the question asked? The FDA had several future.
options regarding the application: (a) They could Russell R. McIntyre
have denied it and yet leave the door open for
re-submission when OS data was available. This option The agency has developed guidelines for the conduct of
presented relatively little risk for the FDA. (b) They clinical trials and the endpoints to be measured in deter-
could have approved; but then, they would have to mining efficacy. They have reviewed their performance
face the uncertainty as to whether the OS data would regarding endpoints for approval of new anti-cancer
support approval. This option is feared by the FDA agents in 2003 [44] and more recently they have issued
because of the negative publicity it generates if the new guidelines [45]. The bottom-line is that the FDA con-
data does not support approval. And yet, this has tinues to favor the most conservative approach. They favor
624 Regulatory process for approval of biologicals for cancer therapy

OS as the ‘most reliable…, precise and easy to measure identified) and occur during the study before any other
…, preferred endpoint’ and state that it should be ‘evalu- therapy is administered. In the case of OS, the effect is
ated in randomized controlled studies’ [45]. It is obvious survival duration. The cause is the biologic activity of
that they do not favor Accelerated Approval because it the anti-cancer agent being studied as influenced by
allows for single arm trials with endpoints other than OS. subsequent therapies, supportive care, etc. The effect, as
Nevertheless, one may still be able to negotiate and come modified by all of these factors, ends when death occurs.
to an agreement with the FDA to utilize TTP or PFS as Death occurs after the study has ended and patients may
primary endpoints. In fact, some new agents have even have gone on to other studies with experimental drugs
been approved with RR (response rate) as the primary and/or received additional conventional chemotherapy.
endpoint. What the FDA does not realize is that OS is Endpoints such as TTP and PFS are much better end-
plagued by multiple factors that introduce bias when this points for clinical trials than OS. In fact, the FDA has
endpoint is utilized. They claim that (in the case of OS) approved more new anti-cancer agents in the past 15
‘bias is not a factor in endpoint measurement’. years based on these than based on OS. Choosing the
Obviously, prolongation of survival when cure is not perfect endpoint is not easy but ‘with the aid of basic
possible is a most desirable objective for the individual experience we must leap bravely into the future’ and the
patient as well as for those around him. Even so, this is future lies with the shorter timeframes of TTP and PFS.
not a strong argument for OS as an endpoint because
survival as a goal for the patient is an entirely different
issue than survival as an endpoint for clinical trials.
Using OS as an endpoint has major drawbacks. These The FDA’s Track Record
studies require more patients, more time, more resources, Statistics is a tool, it is meant to serve, not to enslave [9].
cost much more, and delay the availability of useful
anti-cancer agents. Using OS as an endpoint prolongs In the past 15 years, since Accelerated Approval regula-
clinical trials, delays cancer drug approval and is not in tions have existed, the FDA has granted approval for
the better interests of all patients, current and future. around 30 anticancer indications [46–48]. If one counts
Shouldn’t we be trying “to help or at least to do no harm?” only the first indication for each new agent, there have
That is not achieved by using OS as an endpoint. been a total of 23 new agents that have been granted
OS feels comfortable to the FDA as it appears to Accelerated Approval. However, only 12 of these 23
decrease the risks that they take whenever they approve agents were approved based on response as the sole
a new agent. It only requires two time-points: date on endpoint and based on a single arm trial. This is the sub-
study and date of death. Death is easily confirmed via a set that truly represents the intent of “Accelerated
death certificate. OS is tantalizingly deceptive as it gives Approval”. In the case of the other 11 agents there were
the impression of being so firm, simple, well defined multiple indications, and randomized trials were avail-
and definitive that little judgment might be required in able for some, or the endpoints included some measure
its interpretation whereas other endpoints might appear of response duration. Twelve agents in 15 years, less
to be more problematic [34]. Actually, TTP and PFS are than one agent per year, is hardly a record of which to be
better endpoints even though they do require the perfor- proud and clearly indicative of the FDA’s bias against
mance of CTs, bone scans, or MRIs. These endpoints Accelerated Approval. Accelerated approval has been
are not subject to the multiple factors affecting OS, par- made more and more difficult since it became available
ticularly subsequent therapies, which are unrelated to in 1992. It appears as if the intent is to dissuade appli-
the experimental therapy under evaluation and make OS cants from utilizing this mechanism. Pharmaceutical
a poor endpoint even for randomized and controlled companies have become increasingly reluctant to use
clinical trials. Studies utilizing OS as the primary end- this mechanism. The FDA’s track record to date repre-
point require more patients, more time, more resources, sents a failure to capitalize on an opportunity to bring
cost much more, and delay the availability of useful many more promising new agents early-on to the patient.
anti-cancer agents. Using OS as an endpoint prolongs They do use their statistics to make it look otherwise.
clinical trials, delays cancer drug approval and is not in But then, the FDA does use statistics in different ways
the better interests of the cancer patient. Instead of OS such as a tool in their decision making. However, they
we should use endpoints with clear cause and effect. In do go to the extreme of expecting that statistics will sub-
the case of TTP, the cause is the biologic activity of the stitute for common sense and good judgment [9]. Some
anti-cancer agent being studied, and the effect is lack of believe that Statistics is precise, an exact, science. They
progression, an effect that ends when progression occurs. would have you believe that an analysis is correct if the
Both the onset and the end of the effect are easily p-value is less than 0.05 or better yet, less than 0.01 and
measured (on-study date to date when progression is that truth lies in a favorable hazard ratio, a significant
Antonio J. Grillo-López 625

log-rank test, a pair of Kaplan Meier (KM) curves that drug development and clinical cancer treatment devel-
do not meet, or a median PFS projection from such opment are two related but distinct objectives. It is criti-
curves. The truth is that statistics are fallible and often cally important to understand this distinction. The FDA
inaccurate. A p-value of 0.0298 (or any p-value with is intended to regulate drug development and not treat-
four decimal places) is a joke in spite of the apparent ment development.
accuracy conferred by the four decimal places. A p-value
is worth only as much as the data from which it is
derived. There is truth in the saying that Statistics is the Drug Development Versus Treatment
science of producing reliable fact from unreliable figures. Development
Clinical data is never 100% accurate, relevant, or spe-
cific. Patients and patient populations are remarkably Clinical cancer drug development may be defined as the
heterogeneous biological entities that change and evolve process required for the development of a new antican-
constantly. It is impossible to reliably quantify an end- cer agent (single agent). It is usually carried out by a
point that depends on multiple and variable factors. pharmaceutical company, is tightly regulated and influ-
Patient populations even in the most carefully stratified, enced by the FDA, and has drug approval and marketing
randomized and balanced clinical trials will be hetero- as its goal. Clinical cancer drug development consists of
geneous. You may think that prognostic factors are bal- a lengthy series of activities that require expert profes-
anced, but, as an example, two arms balanced on the sional staff and complex and costly equipment, systems
number of prior therapies can be quite different depending and procedures.
on the nature of the therapies and the patient’s response. Clinical cancer treatment development may be
How can you stratify or balance for immune system defined as the process required for the development of a
responsiveness or for bone marrow reserves when we new anticancer treatment (combination or multimodal-
can’t even measure those very accurately? Some are ity). It is usually carried out by academic institutions,
concerned when two curves on a Kaplan Meier graph consortia, or cooperative groups, is loosely regulated
for PFS come together at the tail end. The only way that and not significantly influenced by the FDA, and has the
two KM curves will not come together at some point [in development of new combinations or multimodality
a TTP, PFS or OS graph] is when one of the two thera- treatments as its goal (Fig. 5).
pies confers immortality. A median PFS, from a Kaplan The FDA’s responsibility, as established by law, is
Meier graph, will be just a “projected or estimated PFS” the review and approval of new anti-cancer agents.
until all events prior to that median have occurred or Pharmaceutical industry is responsible for cancer drug
there are no remaining temporarily censored patients development and this is what the agency is required to
before that median. Only then will that median be fixed, regulate. Clinical cancer drug development should con-
before that there is every probability that it will change. sist of the appropriate Phase I and Phase II clinical trials
The shape of the curve also changes over time. And, (sponsored by a pharmaceutical company) to determine
likewise, it will be fixed only when all events affecting the safety and clinical activity of a new anti-cancer
the curve have occurred. Few clinicians truly under- agent as monotherapy. Development should be col-
stand Kaplan Meier graphs, their intricacies or short- lapsed to the shortest possible timeframe and allow for
comings. The FDA errs in the opposite direction. They early termination of ineffective agents. This would also
expect too much of statistics. allow for the expeditious development of the truly
worthwhile drugs and decrease overall development
time and cost (Fig. 6).
The responsibility for cancer treatment development
Is there a Simpler Approach to should be squarely on the shoulders of the oncology
community. It is the oncology community (including
the Development of New academic institutions, cancer centers, cooperative groups,
Therapies? A Proposal consortia, and others) that, in collaboration with pharma-
ceutical industry, should take a new anti-cancer agent
I don’t know the key to success, but the key to failure is trying to
please everybody.
through the appropriate Phase II and Phase III clinical
Bill Cosby trials to find its optimal use in combination with other
agents. It is the oncology community that should be
A simpler approach would be to approve new anti-cancer responsible for defining the role of a new agent within a
agents based on reasonable safety and efficacy in Phase treatment regimen and its place in the therapeutic arma-
II clinical trials [6]. This would reduce average clinical mentarium. This task is complex and requires multiple
development time from 7 to 3 years. Clinical cancer clinical trials of different combinations. The Phase III
626 Regulatory process for approval of biologicals for cancer therapy

Figure 5. Cancer treatment development

Figure 6. Cancer drug approval: FDA

studies require large numbers of patients and take a as from the regulatory viewpoint. These are: (a) agents
long time to complete. It can take decades before the with significant activity as single agents, (b) agents with
ultimate use of a new anti-cancer agent, within combi- some activity and/or synergy with other agents, and (c)
nation regimens, can be elucidated (Fig. 4). Nevertheless, agents with no significant clinical activity as single
during this time, patients can benefit from treatment agents but with significant synergism with other agents.
with the new agent even though its optimal use in Agents with significant activity as single agents
combination is still in the process of being defined. have a response rate that compares favorably with that
This process, cancer treatment development, should of other available agents (when used as monotherapy).
not be regulated by the FDA. They should be approved on the basis of Phase II trials
with data from historical controls. The entire applica-
tion need not include more than 300 to 350 patients.
Kinds of Useful Anticancer Agents Some measurement of response duration, such as TTP
It is practical to classify clinically useful anti-cancer or PFS, would be useful but should not be an absolute
agents in three categories from the development as well requirement. After all, these new anti-cancer agents
Antonio J. Grillo-López 627

are most frequently combined with other agents rather It is hard to please everybody but this does seem like
than used as monotherapy. It is the response duration a reasonable and simple approach.
of the combination that is of interest. However, the
optimal combination may take years to define. In the
meantime, these agents could be approved and made
available to patients who may benefit from them. What
Conclusions
We cannot avoid meeting great issues. All that we can determine
is the downside of such an approach? At the extreme,
for ourselves is whether we shall meet them well or ill.
the FDA may approve a drug that later shows toxic Theodore Roosevelt
effects not seen in the original experience included in
the application, and/or the response rate may turn There is a regulatory process in place for the approval of
out to be lower than originally determined, and/or biologicals for cancer therapy and, whether we like it or
no additive effect is seen when used in combination not, it must be followed if new anticancer agents are to
regimens. It is unlikely that all of these will be be developed and approved. The process is unduly com-
observed. If this extreme situation does occur, then plex and the requirements many times unnecessary or
approval can be withdrawn or, alternatively, the drug overly rigorous. The FDA’s interpretation of their man-
will die on its own as it will not be prescribed (a sort of date has gone above and beyond what was originally
market induced apoptosis). There are risks but they are envisioned; but then, they themselves interpret the laws
small relative to the potential benefit of having an by which they must abide. Additionally the current pro-
active agent approved earlier rather than much later. cess significantly delays the availability of new agents
Agents with some activity and/or synergy with other to the patients who can benefit from them. This must
agents have a response rate lower than that of other avail- change. The proposal discussed in section 9.0 above
able agents (when used as monotherapy). They may or (approval based on reasonable safety and efficacy in
may not have synergism with other drugs or combina- Phase II clinical trials) is a possibility that should be
tions. Their approval requires a risk benefit determina- seriously considered. It would serve to reduce average
tion. The FDA will not approve such agents unless they clinical development time from 7 to 3 years (Fig. 6)
show superiority or non-inferiority to a standard regimen making the new agent available to patients earlier. Also,
in a Phase III trial. Thus, in most instances, such an agent it would make the new agent available to the oncology
will live or die based on its activity within a combination community earlier for the broad combination studies
that will probably not be the optimal combination. These that would eventually lead to the identification of the
agents should be approved based on Phase II trials and agent’s role within the optimal treatment regimen (Fig. 5).
the oncology community allowed to conduct the multiple Every person interested in the wellbeing of the cancer
studies that will eventually show how they may best be patient must help effect this change. These are great
utilized in combination regimens. As above, there are issues and no one can avoid meeting them.
risks, but not much to lose in approving such an agent.
Agents with no significant clinical activity as single
agents but with significant synergism with other agents Curing Cancer
are inactive as monotherapy but exhibit significant syn-
Destiny is not a matter of chance, it is a matter of choice; it is not a
ergy with other drugs or combinations. In this case, thing to be waited for, it is a thing to be achieved.
there is no choice but to carry out controlled, random- William Jennings Bryan
ized Phase III trials to show the synergistic effect.
Two of these three kinds of useful anti-cancer agents We know that we can cure cancer. Rituximab is an example
can be approved based on reasonable safety and efficacy of a biological that has significantly increased the
in Phase II clinical trials. They would be available to cure rate in an important cancer (DLBCL). Every year
patients earlier, development costs and drug prices should there are in excess of 15,000 NHL patients in the US,
decrease and the oncology community could proceed with another 15,000 in Europe, and a total of over 50,000
combination studies earlier so that new curative regimens worldwide who are potentially curable if treated with R
might be identified. A criticism might be that Pharmaceutical + CHOP. This also means that every year delay in getting
Industry will benefit from these earlier approvals. This rituximab to the market cost over 50,000 lives. We need
should be tempered by the additional risk and cost that more efficient and faster processes for the approval of
early approvals entail including the occasional approval cancer therapies. In 2004 there were over 150 new anti-
withdrawal or failure in the marketplace. All in all, it cancer agents in clinical trials, including 59 biologicals.
seems like a win–win situation for the patient. Of those, 13 were in Phase III. Of the 59 biologicals only
628 Regulatory process for approval of biologicals for cancer therapy

5 have been approved. We need to do better than this. need to act and show they are truly ‘concerned for the
The destiny of the cancer patient is a matter of choice welfare and dignity and decency and innate integrity of
and the choice must be to act faster, decrease bureau- life for every individual’.
cracy and delays, approve earlier and make promising
new agents available to those who need them. Just like
rituximab there may be new agents in development that References
can extend lives or be curative. They must be evaluated 1. Smith CM (2005). Origin and uses of primum non nocere – above
earlier with a view to their optimal role in combination all, do no harm! J Clin Pharmacol 45(4):371–377.
regimens. Regulatory review and approval processes 2. Portlock CS and Rosenberg S (1979). No initial therapy for stage
should help and not impede this ultimate objective. III and IV non-Hodgkin’s lymphomas of favorable histologic
types. Ann Intern Med, 90:10–13.
3. Horning S and Rosenberg S (1984). The natural history of initially
The Patient’s Plight untreated low-grade non-Hodgkin’s lymphomas. N Engl J Med
311(r):1471–1475.
Were we directed from Washington when to sow, and when to reap, 4. Grillo-López AJ (2001). The important role of monoclonal antibodies
we should soon want bread. in the treatment of non-Hodgkin’s lymphomas. Onc Spec 2:700–705.
Thomas Jefferson 5. World Medical Association International Code of Medical Ethics
(2006). Modern version of the Hippocratic oath. http://www.wma.
The patient is at the receiving end of drug development. net/e/policy/c8.htm
The system is such that it takes a long time for effective 6. Grillo-López AJ (2005). The ODAC chronicles: Part III. The
drugs to go from discovery to being broadly available to FDA’s philosophy and process for cancer drug evaluation and
approval (Editorial). Expert Rev Anticancer Ther 5(1):1–5.
patients. It is the government that imposes the delays,
7. Grillo-López AJ (2007). Curing cancer: A historical perspective on
hurdles and restrictions. The patient has to wait and yet, the development of rituximab. Book chapter in: Rituximab-Mediated
the patient does not have the luxury of time. FDA reform Molecular Signaling and Interaction with Chemotherapeutic Drugs
must stop being a buzz word heard only prior to elec- (B Bonavida, Ed.), Transworld Research Network, Kerala, India,
tions. It must become reality. pp. 1–7.
8. Grillo-López AJ (2001). Curative therapies for NHL: A question of
time! (Editorial) in Monoclonal antibodies in the treatment of hema-
What does the Future Hold? tologic malignancies – A special issue of Curr Pharm Bio 2(4):1–2.
9. Grillo-López AJ (2004). The ODAC chronicles: Part II. Statistics
I believe that the essence of government lies with unceasing con- and clinical medicine in the USA – the triumph of science over art?
cern for the welfare and dignity and decency and innate integrity of (Editorial). Expert Rev Anticancer Ther 4(6):941–944.
life for every individual. 10. Dillman RO (1994). Antibodies as cytotoxic therapy. J Clin Oncol
Lyndon B. Johnson 12(7):1497–1515.
11. Ehrlich P (1957). The Collected Papers of Paul Ehrlich, Vol II (IF
Today’s physicians have a responsibility to actively par- Himmelweite, M Marquardt, and H Dale, Eds.), Pergamon Press,
ticipate in making new therapies available to the cancer London, England, pp. 550–557.
patient. We know that cancer can be cured. However, 12. Grillo-López AJ, Dallaire BK, Varns CL, et al. (2000). Rituximab:
The first monoclonal antibody approved for the treatment of lym-
biologicals and all new anticancer agents must be effi- phoma. Curr Pharm Biotechnol 1(1):1–9.
ciently and rapidly evaluated to find those that are most 13. Grillo-López, AJ (2003). Rituximab: The first decade (1993–2003).
promising. They must then be incorporated into combi- Expert Rev Anticancer Ther 3(6):767–779.
nation regimens that provide the greatest opportunity 14. Grillo-López AJ (2002). Zevalin: The first radioimmunotherapy
approved for the treatment of lymphoma. Expert Rev Anticancer
for cures. Physicians in the pharmaceutical industry
Ther 2(5):485–493.
must do everything within their reach to ensure that new 15. Grillo-López AJ (2005). 90Y-ibritumomab tiuxetan: Rationale for
agents are developed and submitted for approval in the patient selection in the treatment of indolent non-Hodgkin’s lym-
most expeditious fashion. Physicians in our regulatory phoma. Semin Oncol 32(1, Pt. 2):44–49.
agencies (FDA, EMEA, and others) must likewise 16. Reff ME, Carner K, Chambers KS, et al. (1994). Depletion of B
cells in vivo by a chimeric mouse human monoclonal antibody to
understand that they have similar duties and responsi-
CD20. Blood 83(2):435–445.
bilities to the patient. Being employed by a bureaucracy 17. Anderson DR, Grillo-López A, Varns C, et al. (1997). Targeting
does not mean that your sacred oath is no longer valid. cytotoxic immunotherapy: Targeted anti-cancer therapy using
In fact, physicians in regulatory agencies could be an rituximab, a chimaeric anti-CD20 antibody (IDEC-C2B8) in the
important catalyst in making new promising therapies treatment of non-Hodgkin’s B-cell lymphoma. Biochem Soc Trans
25:705–708.
available to patients in a timely fashion. Government 18. Demidem A, Lam T, Alas S, et al. (1997). Chimeric anti-CD20
must come to understand that they can correct the issues (IDEC-C2B8) monoclonal antibody sensitizes a B-cell lymphoma
that we face given the current regulatory process for the cell line to cell killing by cytotoxic drugs. Cancer Biother Radiopharm
approval of biologicals and other cancer therapies. They 12(3):177–185.
Antonio J. Grillo-López 629

19. Maloney DG, Liles TM, Czerwinski DK, et al. (1994). Phase I patients with diffuse large B-cell lymphoma. N Engl J Med
clinical trial using escalating single-dose infusion of chimeric anti- 346(4):235–242.
CD20 monoclonal antibody (IDEC-C2B8) in patients with recurrent 33. FDA History. http://www.fda.gov/oc/history/default.htm
B-cell lymphoma. Blood 84(8):2457–2466. 34. Grillo-López AJ (2004). The ODAC chronicles – Part I: My first ODAC
20. Berinstein NL, Grillo-López AJ, Bence-Bruckler I, et al. (1998). experience (Editorial). Expert Rev Anticancer Ther 4(5):579–582.
Association of serum rituximab (IDEC-C2B8) concentration and 35. Establishment of standing technical advisory committees for
anti-tumor response in the treatment of recurrent low-grade or fol- Human Prescription Drugs. Code of Federal Regulations. Title 21,
licular non-Hodgkin’s lymphoma. Ann Oncol 9:995–1001. Volume 1 [Revised as of April 1, 2003], Part 14, Subpart I, Section
21. Grillo-López AJ (2000). Rituximab: An insider’s historical per- 14.160.
spective. Semin Oncol 27(6, 12 Suppl):9–16. 36. Committee Charter: Oncologic Drugs Advisory Committee. http://
22. Maloney DG, Grillo-López AJ, Bodkin DJ, et al. (1997). IDEC- www.FDA.gov.
C2B8: Results of a Phase I multiple-dose trial in patients with relapsed 37. Guidance for Industry Advisory Committees: Implementing Section
non-Hodgkin’s lymphoma. J Clin Oncol 15(10):3266–3274. 120 of the Food and Drug Administration Modernization Act of 1997.
23. Maloney DG, Grillo-López AJ, Bodkin D, et al. (1997). IDEC- http://www.fda.gov/cder/guidance/index.htm
C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in 38. Grillo-López AJ (2005). The ODAC chronicles: Part VIa. ODAC’s
patients with relapsed low-grade non-Hodgkin’s lymphoma. Blood structure and function (Editorial). Expert Rev Anticancer Ther
90(6):2188–2195. 5(4):573–577.
24. McLaughlin P, Grillo-López AJ, Link BK, et al. (1998). Chimeric 39. Grillo-López AJ (92005). The ODAC chronicles: Part VIb. ODAC’s
anti-CD20 monoclonal antibody therapy for relapsed indolent structure and function (Editorial). Expert Rev Anticancer Ther
lymphoma: Half of patients respond to a 4-dose treatment pro- 5(5):753–756.
gram. J Clin Oncol 16:2825–2833. 40. New Drug, Antibiotic, and Biological Drug Product Regulations;
25. McLaughlin P, Hagemeister FB, and Grillo-López AJ (1999). Accelerated Approval, 57 Fed. Reg. 58,942 (1992) [codified as
Rituximab in indolent lymphoma: The single-agent pivotal trial. amended at 21 CFR 314.500-.560, 601.40-.46 (1999)].
Semin Oncol 26(5, 14 Suppl):79–87. 41. Subpart E – Accelerated approval of biological products for serious
26. Grillo-López AJ, Varns C, Shen D, et al. (1999). Overview of the clini- or life-threatening illnesses. 57 Fed. Reg. 58959 (1992) (Cite
cal development of rituximab: First monoclonal antibody approved for 21CFR601.40).
the treatment of lymphoma. Semin Oncol 26(5, 14 Suppl):66–73. 42. Briefing documents for the ODAC meeting of July 24, 2007. http://
27. Wei A, Shen D, Kim D, et al. (2003). The Electronic Solution to www.fda.gov/ohrms/dockets/ac/07/briefing/2007-4309b1-00-in-
the FDA Submission – Notes From the Field. Appl Clin Trials dex.htm
12(10):60–64. 43. US panel: FDA needs more data on GPC cancer drug (2007).
28. Czuczman M, Grillo-López AJ, Saleh M, et al. (1995). Phase II Reuters News Agency 24 July 2007. http://www.reuters.com/article/
clinical trial of IDEC-C2B8/CHOP combination therapy in low governmentFilingsNews/idUSN2437083920070724
grade lymphoma: Preliminary results. Proc Am Soc Clin Oncol 44. Johnson JR, Williams G, Pazdur R (2003). Endpoints and United
14:401 (#1261). States Food and Drug Administration approval of oncology drugs.
29. Demidem A, Lam T, Alas S, et al. (1997). Chimeric anti-CD20 J Clin Oncol 21:1404–1411.
(IDEC-C2B8) monoclonal antibody sensitizes a B cell lymphoma cell 45. Guidance for industry: Clinical trial endpoints for the approval
line to cell killing by cytotoxic drugs. Cancer Biother Radiopharm of cancer drugs and biologics (2007). http://www.fda.gov/cder/
12(3):177–185. guidance/7478fnl.pdf
30. Vose JM, Link BK, Grossboard ML, et al. (2001). Phase II Study 46. FDA Approval Statistics (2007). FDA/Center for Drug Evaluation
of Rituximab in combination with CHOP chemotherapy in patients and Research Last Updated: 08/20/2007 07:47:37. Originator:
with previously untreated, aggressive non-Hodgkin’s lymphoma. OTCOM/DLIS. HTML by SH. http://www.accessdata.fda.gov/
J Clin Oncol 19:389–397. scripts/cder/onctools/statistics.cfm
31. Vose JM, Link BK, Grossbard ML, et al. (2005). Long-term update 47. Dagher R, Johnson J, Williams G, et al. (2004). Accelerated Approval
of a phase II study of rituximab in combination with CHOP che- of oncology products: A decade of experience. J Natl Cancer Inst
motherapy in patients with previously untreated, aggressive non- 96:1500–1509.
Hodgkin’s lymphoma. Leuk Lymphoma 46(11):1569–1573. 48. Grillo-López AJ (2005). The ODAC chronicles: Part IV. Hurdles
32. Coiffier B, Lepage E, Briere J, et al. (2002). CHOP chemother- pre- and post-accelerated approval (Editorial). Expert Rev Anticancer
apy plus rituximab compared with CHOP alone in elderly Ther 5(2):197–200.
21.1 Cancer biotherapy: 2009 disease-related
activity
ROBERT K. OLDHAM AND ROBERT O. DILLMAN

In the last decade, we have witnessed a burgeoning number we now use. As new approaches in biotherapy are tested,
of biologicals under clinical investigation, either singly scientists and clinicians must be constantly aware that
or in combination with other biologicals or chemothera- our current histological anatomic classification system
peutic agents. Biotherapies have demonstrated efficacy is largely artificial and that clues to the clinical activity
against certain malignancies and are approved for gen- of biotherapy may allow us to reclassify neoplasms
eral use in medicine. It is expected that their assimilation according to biological features and behavior patterns.
into our standard anticancer armamentarium will con- Indeed, recent advances in the molecular analysis of
tinue to broaden in the next decade, leading to the domi- tumors by gene expression profiling and proteomics
nance of biotherapy in cancer treatment soon after the have begun to lead to a new paradigm for the classifica-
year 2010. This chapter will summarize disease-related tion of malignancies based on the molecular signature
activity for selected biotherapies as we move into the of an individual’s tumor derived from the global assess-
second decade of the new millennium. ment of thousands of genes. By examining the clustering
Historically, cancers have been classified by histo- of gene expression, new categories of malignancy have
pathological features, based on the pathologist’s ability begun to emerge that go beyond traditional distinctions
to discern similarities and differences among cancers by based on histology alone [6].
scrutinizing tissue sections under the microscope. Over Gene profiling and microarray analysis have recently
100 types of cancers have been so categorized. This been shown, by retrospective studies, to correlate with
pathology-based classification scheme, upon which the prognosis. Current efforts are focused on using this kind
previous standard modalities of cancer treatment – surgery, of genetic information to design clinical trials in breast,
radiation therapy, and chemotherapy – were structured, colon and lung cancer. The hypothesis in breast cancer
has contributed to major advances in the treatment of is that a certain profile might predict for a better prognosis
selected malignancies. using hormone therapy alone vs. adding chemotherapy
As cancer biology and cellular genetics, including for those individuals having a gene profile consistent
cancer susceptibility genes become better understood, with more aggressive disease. Once such a hypothesis is
we are learning that the unregulated growth of tissues, validated through current prospective clinical trials, the
the activation of oncogenes, mutations, the persistence adjuvant therapy of breast cancer will be much improved,
of cells that normally become senescent, and matura- given that we can focus chemotherapy on the most likely
tional aberrations may segregate in patterns not amena- subgroup to benefit [2, 3, 4, 7, 13].
ble to simplistic histopathological classification. Early Such considerations transcend all aspects of classical
evidence of such conflicts in categorization is provided developmental therapeutics, in which surgery, radiation
by the presence of similar antigens on cancer cells aris- therapy, and chemotherapy evolved along disease-specific
ing from very different tissues. For example, antibodies and anatomic lines. Thus, although phase I toxicity trials
have been described that react very clearly with breast are conducted across histological boundaries, phase II and
cancer, colon cancer, and certain other adenocarcino- III studies progress within a fairly rigid disease-specific
mas. As such, these antibodies cross histopathological format. As a consequence, antitumor activities in lung
and anatomic borders. Similarly, there may be common cancer, breast cancer, colon cancer, etc. are discussed
reactivities among tumors of lymphoid origin, of and debated. While such a testing paradigm may have
squamous differentiation, and of mesenchymal deriva- served oncologists well for protocol construction and for
tion. A better understanding of oncogene activation, regulatory oversight, this mindset of classical pathology
genomics and of normal and aberrant regulation of oriented phase II–III studies may actually inhibit the
growth and differentiation may provide for a dynamic development of biotherapy [8]. Furthermore, the devel-
classification scheme rather than a static descriptive one opment of biological therapies and molecularly targeted

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 631
© Springer Science + Business Media B.V. 2009
632 Cancer biotherapy: 2009 disease-related activity

anticancer agents poses special challenges to the design of these activities briefly with references into 2009 and is
clinical trials to include models that assess the effect of the for quick reference only. The reader is encouraged to
agent on its putative target and the validation of new sur- refer to specific, subject-oriented chapters and refer-
rogate endpoints [12]. The traditional objectives of phase ences for detailed information on specific methods of
I and II studies (maximally tolerated dose and response cancer biotherapy.
rate, respectively) developed for cytotoxic agents are less
relevant to biological and targeted therapies where a
“biologically effective dose” may prevent tumor growth
and induce stabilization of disease that is of important References
clinical benefit but is not reflected by a radiographically 1. Avner BP, Liao SK, Avner B, DeCell K, Oldham RK. Therapeutic
measurable objective response. With these new modali- murine monoclonal antibodies developed for individual cancer
patients. J Biol Response Mod 1989; 8:25–36.
ties, we must be open to the possibility that the biologi-
2. Brenton JD, Carey, LA, Ahmed AA, et al. Molecular classification
cal activity of these agents and approaches may sort and molecular forecasting of breast cancer: Ready for clinical
unpredictably and may require a more individualistic application? J Clin Oncol 2005; 23(29):7350–7360.
orientation. It is abundantly clear that monoclonal anti- 3. Cheang MCU, Voduc D, Bajdik C, et al. Basal-like breast cancer
bodies, proteomics, and gene expression profiling tech- defined by five biomarkers has superior prognostic value than triple-
negative phenotype. Clin Cancer Res 2008; 14(5):1368–1376.
nology can allow for the precise description of an
4. Cronin M, Sangli C, Liu M-L, et al. Analytical validation of the
individual patient’s cancer using panels of antibodies, oncotype DX genomic diagnostic test for recurrence prognosis and
mass spectroscopy signatures, and microarrays that rep- therapeutic response prediction in node-negative, estrogen receptor-
resent all of the expressed genes in the human genome positive breast cancer. Clin Chem 2007; 53(6):1084–1091.
[1, 5, 10, 11]. The use of the laboratory to develop 5. Lakhani SR, Ashworth A. Microarray and histopathological analysis
of tumours: the future and the past? Nat Rev Cancer 2001;
patient-specific therapies may allow scientists and clini- 1:151–157.
cians to approach the cancer problem from a completely 6. Liotta L, Petricoin E. Molecular profiling of human cancer. Nat
different and more individualized perspective [9]. Rev Genet 2000; 1:48–56.
Many biotherapies are supportive or ancillary to the 7. Loi S, Sotiriou C, Buyse M, et al. Molecular forecasting of breast
anti-tumor activity of standard approaches such as che- cancer: Time to move forward with clinical testing. J Clin Oncol
2006; 24(4):721–722.
motherapy. Use of colony-stimulating factors, erythro- 8. Oldham RK. Biologicals and biological response modifiers: design
poietin, blood transfusions, marrow transplantation, etc. of clinical trials. J Biol Response Mod 1985; 4:117–128.
has broad application in oncology. These techniques 9. Oldham RK. Custom-tailored drug immunoconjugates in cancer
were described in earlier chapters. therapy. Mol Biother 1991; 3:148–162.
10. Oldham RK, Lewis M, Orr DW et al. Adriamycin custom-tailored
While recognizing the limitations of organ-specific
immunoconjugates in the treatment of human malignancies. Mol
categorization and the likely reclassification of tumors Biother 1988; 1:103–113.
based on either gene expression clusters or the expres- 11. Orr D, Oldham R, Lewis M et al. Phase I trial of mitomycin C
sion of common molecular therapeutic targets, it remains immunoconjugates cocktails in human malignancies. Mol Biother
a standard and useful exercise to describe the clinical 1989; 1:229–240.
12. Tan AR, Swain SM. Novel agents: clinical trial design. Semin
activities in biotherapy within such a historical frame-
Oncol 2001; 28:148–153.
work. Therefore, in this chapter, a brief summary of the 13. van ‘t Veer LJ, Mongyue D, van de Vijver MJ, et al. Expression
activity of various forms of biotherapy will be provided profiling predicts outcome in breast cancer. Breast Cancer Res
by cancer type. This chapter is designed to describe 2003; 5:57–58.
21.2 Biological therapy of melanoma
ROBERT K. OLDHAM

Melanoma is among the human solid tumors that best melanoma has been carefully documented [92]. Indeed,
exemplify the application of biological therapy to the approximately 5% of patients with disseminated melanoma
treatment of cancer. In the first decade of the twenty-first do not have an identifiable primary lesion, suggesting that
century, more than 4 million people will be diagnosed the cutaneous disease may have spontaneously regressed
with malignant melanoma, and the incidence continues to before the growth of metastases is detectable [94].
increase in developed countries worldwide [9]. Although Paradoxically, partial regression of primary melanoma
primary cutaneous melanoma is highly curable, with 85% lesions has been reported by some observers to be associated
of patients enjoying long-term survival after surgical with a worse prognosis; however, this is likely due to the
excision, disseminated melanoma was amenable only to difficulty that this feature adds to the assessment of risk in
palliation with radiation therapy and chemotherapy [41]. the pathological specimen. This phenomenon has recently
Although short term responses to chemotherapy are not been shown to involve cytolytic T-lymphocytes [37, 71].
uncommon, particularly in skin, lung and subcutaneous Second, paraneoplastic depigmentation events, such as the
lesions, significant long-term benefit in visceral disease is development of vitiligo and “halo” formation around
infrequent, with 5-year survival remaining less than 5% primary skin melanomas and nevi are thought to indicate
for disseminated disease. Because melanoma frequently an immune mediated reaction against the pigmented cells
affects young individuals, half of the patients who die of in these lesions, and the development of vitiligo in
melanoma had a life expectancy of greater than 25 years, malignant melanoma during biotherapy carries an improved
and the productive years of life lost exceed those of any prognosis [24]. The corollary of this clinical observation
other solid tumor [121]. may have therapeutic implications: if the immune
recognition of melanoma can lead to the destruction of
normal melanocytes, immunization with melanocyte
antigens may lead to the rejection of melanomas [7]. Third,
Melanoma and the Immune the presence of tumor infiltrating lymphocytes in primary
System melanoma lesions indicates a host immune response,
and the degree of lymphocyte invasion is of prognostic
Melanoma has long been known to be a tumor with
significance [15, 25]. Fourth, there are patients whose
unusual behavior in certain patients. Spontaneous, com-
melanoma may remain dormant for many years, suggesting
plete regression occurs in one patient in 1,000–10,000
innate immune surveillance and control of micrometastatic
with lesser degrees of temporary spontaneous partial
disease, who subsequently relapse and die from dissemi-
regression in as high as 0.5% of patients. Such observa-
nated melanoma 10 or more years after the diagnosis of the
tions indicate that the body has some inherent capacity
primary lesion [108, 110]. Finally, the incidence of mela-
to induce regression of this disease in selected instances.
noma is higher and the prognosis worse among immuno-
Despite considerable investigation, the mechanisms of
suppressed kidney transplant recipients, providing
such spontaneous regressions remain unclear, and formal
additional support for the role of immune surveillance in
proof of the involvement of an innate or adaptive anti-
the evolution of malignant melanoma [23, 34].
melanoma immune response is still lacking.
Several features of malignant melanoma suggest that the
in vivo biological responses that play a role in the progres-
sion and regression of this form of cancer involve the Biotherapy of Malignant
immune system. For this reason, melanoma is an attractive
and well-examined model for the therapeutic use of bio-
Melanoma
logical agents, particularly those designed to enhance or Early observations indicated that the immune system
manipulate the host immune response. First, the infrequent could be harnessed therapeutically to treat malignant mel-
spontaneous regression of cutaneous and disseminated anoma. Intratumoral injection of bacterial extracts such as

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 633
© Springer Science + Business Media B.V. 2009
634 Biological therapy of melanoma

Bacille Calmette-Guerin (BCG) or Corynebacterium studied in melanoma. Up to 90% of melanoma nodules


parvum frequently induced the regression of the injected injected intralesionally with BCG will demonstrate
melanoma and occasionally lead to tumor regression at regression in immunocompetent patients. In 15–20% of
sites of lymphatic drainage and rarely distant metastases such patients, associated distant shrinkage of nodules
[90]. The natural history of malignant melanoma and these may be seen [77]. Tumor regression by the local applica-
early therapeutic observations provide a compelling ratio- tion of BCG has also been successfully applied to the
nale for the systematic evaluation of biological therapy in treatment of superficial bladder cancer, and the mecha-
malignant melanoma. In this light, many agents have now nism of tumor regression has been investigated in this
been evaluated to enhance or mimic the host immune model. The local application of BCG sensitizes host
response against melanoma. These agents have included T-cells to peptides in the extracts, and the presentation of
effector cells such as lymphocytes derived from resected these peptides by host antigen presenting cells within the
tumors or the blood of melanoma patients; antibodies injected tumor bed triggers local T-cell cytokine produc-
directed at various epitopes on the melanoma cell; vac- tion that can activate host cells that can directly recog-
cines composed of natural or synthetic components of the nize and kill tumor cells [98, 99, 108]. Preparations of
melanoma cell or derived from mimic “anti-idiotype” cellular constituents, such as cell-wall skeleton, may also
antibodies; non-specific stimulators of the immune system serve as effective adjuvants [123]. Systemic effects have
such as BCG and the cytokines. Until recently, only the been demonstrated by the emergence of antimelanoma
cytokines have consistently shown clinical, anti-tumor antibodies and by lymphocytic infiltration of regressing
activity in melanoma, and two of these cytokines, inter- noninjected lesions [77].
feron alpha and interleukin-2, have emerged as useful in Vaccines derived from allogeneic melanoma cells have
clinical practice for the treatment of patients with malig- long been of interest. In one early report, an enriched
nant melanoma [10, 53, 62]. tumor cell vaccine was infused intra-lymphatically, pro-
Although durable responses are rare, cytotoxic therapy viding proof of principle for the concept of inducing
for melanoma leads to tumor regression in a substantial host antitumor immunity with antigens from allogeneic
number of patients, and the activity of single agents and melanoma cells by demonstrating that the effects of
chemotherapy combinations is well established. As a such intra-lymphatic immunotherapy were not limited
result, treatment regimens that combine chemotherapy to regional lymph nodes and that systemic response
and biological therapy agents, usually cytokines, are were seen [1]. These initially encouraging results have
referred to as biochemotherapy [46, 96, 109, 119]. lead to intense study of this strategy of immunomodula-
Monoclonal antibodies that influence the immune tion for at least the last quarter century, yet these efforts
response are currently in clinical trials. Chief among have not significantly improved the survival of mela-
these are anti-CTLA-4 antibodies, which have been noma patients [78]. However, immunizing melanoma
promising in early-phase clinical trials with the induc- patients with a pure Gm2 ganglioside, known to be on
tion of both partial and complete responses in patients melanoma cell membranes, has induced circulating IgG
with advanced melanoma. Toxicities have been substantial and IgM antibodies. The induction of such antibodies
with bowel inflammation and severe colitis as well as has been reported to be accompanied by prolonged
iritis being seen in many of these patients. Vitiligo also survival [69, 75, 80].
is a side effect of this form of biological therapy and While definitive proof of the success of vaccines
may predict a good response to the monoclonal antibody derived from allogeneic melanoma cells is lacking, at
therapy [6, 27, 33, 42, 91, 107]. least one allogeneic cell lysate vaccine (Melacine) has
The gene therapies for malignant melanoma are new undergone rigorous analysis and was approved in 1999
topics of interest; in these strategies, a therapeutic gene is for the treatment of disseminated melanoma in Canada
introduced into an effector cell or a gene encoding a strong based on phase III results demonstrating superior quality
antigen is introduced directly into the melanoma cell. of life during active therapy for disseminated melanoma
Clinical trials with gene therapies are now underway. compared to a four-drug chemotherapy combination
[39]. The two therapies achieved similar efficacy results.
Survival data was also compared from the Phase III
Immunostimulation study with a comprehensive meta-analysis of therapies
for Stage IV melanoma showing that the median sur-
and Vaccines vival of 11 months among evaluable patients receiving
Attempts to augment native immunologic defenses using this allogeneic vaccine was better than that achieved by
various non-specific immunostimulators, particularly other available therapies [59]. Further, a significantly
Bacillus Calmette-Guerin (BCG), have been intensively longer median survival of 18.2 months was observed in
Robert K. Oldham 635

patients who were clinical responders to Melacine therapy. has yielded a response rate of approximately 20% [51,
Melacine has not been approved by the FDA for use in 81]. On occasion, these responses have been complete,
the U.S. as of 2008. but activity in visceral disease has been infrequent.
More recent studies using genetically engineered Perhaps as many as 30% of patients achieving a com-
vaccines have been performed. Details can be found in plete response enjoy an enduring remission. Studies
the chapter on vaccines and the chapter on gene therapy. using alpha-interferon in the adjuvant surgical setting
This work was prompted by previous studies using par- demonstrated a reduced recurrence rate with substantial
tially purified tumor vaccines (Table 1). doses of interferon. This study lead to FDA approval of
More recent studies have used highly purified or interferon for the treatment of melanoma. Beta and gamma
genetically engineered vaccines with a variety of anti- interferon were also evaluated in melanoma with both
gens and adjuvants which are immunogenic in humans now approved for non-cancer indications. A suggested
(Tables 2 and 3). potentiation by cimetidine of human leukocyte inter-
Clinical trials with genetically engineered vaccines feron activity in malignant melanoma [11, 47] has not
are ongoing and while there is evidence of stimulation been confirmed by later studies using natural or recom-
of cytotoxic T cells, enhancement of antibody titers and binant alpha-interferon [51, 68].
augmentation of DTH responses, the anti-tumor activi- There has been great interest in exploring the benefits
ties of these vaccines remain to be confirmed. of adjuvant IFN for high risk, surgically excised mela-
noma. Based on the findings in metastatic disease, it
was hoped that IFN would be most active when the
tumor burden was low and would increase the overall
Interferon survival of patients at high risk of relapse after surgery
In phase I trials with alpha-interferon, an occasional for melanoma. Multi-institutional, randomized con-
melanoma patient experienced objective tumor regression. trolled trials (RCTs) have been conducted in the United
Subsequent phase II studies using partially purified States and Europe in an attempt to demonstrate a benefit
human leukocyte or lymphoblastoid alpha-interferon to adjuvant IFN. The inter-group United States trials,
showed response rates as high as 12% [43, 95]. The com- lead by the Eastern Cooperative Oncology Group
posite experience using recombinant alpha-interferon (ECOG), E1684, E1690, and E1694, have received the

Table 1. Tumor antigen vaccine trials


Stage of disease No. of patients Vaccine preparation Adjuvant Controls Results Ref.
Resected II 25 Autologous soluble BCG Historical Improved survival over 162
membrane extract historical growth
Disseminated 56 Specific TAA Freund’s complete None Regression seen in 137
approximately 25%
of patients
Disseminated 13 Polyvalent TAA None None CR 1 of 13 PR 1 of 13 36
Resected stage II 94 Polyvalent melanomatumor 40 pts – Alum Historical Improved survival over 38
antigen vaccine 17 pts – Cytoxan historical controls

Table 2. Adjuvants: effects on immunogenicity


Mechanisms of action
Approach Depot effect Macrophage CD4+ T cell and B cell CTL
Conjugate vaccines + − ++ −
Recombinant vector vaccines
BCG + + + ++
Vaccinia + − + ++
Immunological adjuvants
Alum or oil (squalene) + − − −
BCG or BCG CWS + + + −
Endotoxin (lipid A) − ++ + −
Liposomes + − − +
MDP derivatives − + + +
636 Biological therapy of melanoma

Table 3. Clinical trials with immunogenic antigens to patients with stage III disease that had involved
Antigen Vaccine No. of patients responding Ref. regional lymph nodes [55]. In addition, the dose and
duration of therapy differ and among studies and this
GM2 BCG 50/58 37
GM2 KLH 30/30 131, 217 issue remains contentious. Data from the United States
GD2 KLH 4/6 131, 217 intergroup studies suggest that higher doses of IFN in the
Mage 1 – Ongoing trial 361 adjuvant setting result in greater effectiveness. A meta-
Mage 3 – Ongoing trial 106 analysis of ten randomized controlled trials was recently
Tyrosinase – Ongoing trial 29
conducted and concluded that there was a clear benefit
to adjuvant IFN for prolonging disease free survival but
most attention and scrutiny in North America. E1684 the benefits were less clear in terms of overall survival
enrolled 287 patients and randomly assigned patients to [125]. In this meta-analysis, high dose versus low dose
observation versus 1 year of high dose IFN for 1 year IFN was compared, and there was no statistically signifi-
(“high dose” defined here as at least 10 million units per cant difference between high dose and low dose IFN treat-
square meter) versus observation. Disease free and ment. However, a systematic review of eight randomized
overall survival was both improved by IFN treatment controlled trials of systemic adjuvant interferon as
[55], and long-term follow-up data from this study was monotherapy versus no treatment (and thus excluded
confirmatory [55, 56]. In follow-up, E1690 accrued 642 E1694 from analysis) for high risk melanoma concluded
patients to observation or treatment with high dose IFN that there was no clear benefit of IFN therapy on overall
or low dose IFN. Although median survival for high survival and that heterogeneity between the trials made
dose IFN was essentially identical to the median sur- meta-analysis inappropriate [64]. Thus, despite fairly
vival for high dose IFN on E1684, overall survival was rigorous and intensive study, controversy remains regard-
not improved by treatment with high dose IFN (or low ing the benefits of IFN in the adjuvant setting [56].
dose IFN) [54]. The lack of benefit for IFN in E1690 Studies of interferon alpha in metastatic melanoma
appears to derive from an unexplained improvement in patients have shown response rates ranging from 0–30%
the median survival of the observation group in E1690 with an overall average of 16% [15, 19, 20, 32, 35, 52,
compared to E1684. A significant number of patients on 63, 112, 117]. As such, about one in six patients with
observation on E1690 received IFN at the time of recur- metastatic melanoma will benefit from interferon alpha
rence, which may confound interpretation of the results therapy. However, it is notable that a small percentage of
of IFN treatment. Finally, E1694, enrolled 774 patients the total group responding to this treatment will achieve
to treatment with high dose IFN or a vaccine, and dem- a complete response, and occasionally, long remissions
onstrated that the high dose IFN significantly prolonged have been observed. A review of 11 phase II studies of
both disease free and overall survival [54]. The vaccine single agent IFN conducted in the 1980s and comprising
treatment arm of E1694 had a similar survival compared 315 patients described an overall response rate of 15%
to the control arm of E1690, indicating that the improve- and a median survival of 8 months [61]. The dose, route
ment in disease free and overall survival resulted from and schedule of IFN administration differed among the
the treatment with IFN rather than any detrimental effect studies, but intramuscular injections given three times
of the vaccine treatment. weekly were usually used, and doses of 10 million units
Other large studies have explored the role of adjuvant per square meter or higher (up to 50 million units per
IFN in melanoma. For example, among the more square meter) were used in all cases. Complete remis-
recently reported, the AIM High Study randomized 654 sions were observed in 0–4% of patients. It is difficult to
patients to treatment with low dose IFN for 2 years or extrapolate the optimal dose and frequency of adminis-
observation. No significant effect was observed on dis- tration of interferon alpha from the available data; a very
ease free or overall survival [12]. Several other impor- wide range of doses and frequencies have been evaluated
tant studies, also using low dose IFN, have also been in the reported clinical trials (Tables 4 and 5). It is also
performed and failed to convincingly demonstrate an not clear that a strong dose–response relationship exists
overall survival benefit for adjuvant therapy for mela- for interferon alpha in malignant melanoma.
noma [18]. Definition of the high risk population has It is important to compare these responses to those
differed among randomized studies, primarily depend- observed for single agent chemotherapy (also in the
ing on the inclusion of patients with intermediate or 15–20% range). Higher response rates were seen with
deep primary lesions or involved regional lymph nodes. drug combinations and the best three drug combinations
In the E1684 study that initially demonstrated a benefit to report responses in the 30–40% range and generally
adjuvant IFN, the benefits of adjuvant IFN were limited include cisplatin and dacarbazine. However, even the
Robert K. Oldham 637

Table 4. Clinical trials with recombinant interferon activity for the two drugs [3, 36] while a third showed
Study Dose (MU) route/ Evaluable CR/PR no improvement over dacarbazine alone [120].
schedule patients (%) A very encouraging randomized phase II trial has dem-
Creagen [56] 12/M2 i.m. 3 times 30 20 onstrated that bevacizumab (Avastin) has shown activity
per. week in metastatic melanoma when compared in a randomized
Creagen [58] 50/M2 i.m. 3 times 31 23 phase II trial with interferon. A large randomized phase
per week III study is in progress to compare bevacizumab plus
Coates [49] 20/M2 i.v. daily × 5 15 0
interferon with interferon alone.
Hersey [134] 50/M2 i.m. 3 times 18 11
per week
Legha [178] 3–36 i.m. daily or 3 62 8
times per week
Interleukin 2/lymphokine-
activated Killer (LAK) Cells
Table 5. Biochemotherapy treatment programs
Interleukin-2/lymphokine activated killer (LAK) cells
Study Evaluable patients CR/PR (%)
have been reported to provide a response rate of 50% for
Interferon + DTIC patients with advanced metastatic melanoma [100, 124]
Kirkwood [159] 23 4
(Table 6). Most of these responses have been partial and
Gundersen [124] 15 20
Mulder [220] 31 35 of only several months duration, although occasional
Thompson [354] 86 21 patients do experience complete responses. Subsequent
Sertoli [322] 72 26 studies with interleukin 2/LAK cells and studies using
Interferon + platinum/combinations interleukin 2 in combination with interferon continue to
Hamblin [130] 12 83
Richards [286] 74 57
demonstrate that melanoma is a neoplasm sensitive to
Legha [179] 30 56 these kinds of biological approaches [11, 28, 31, 58,
Pyrhonen [271] 45 53 106, 114]. Overall, response rates have been in the
Richner [288] 20 35 20–30% range. Responses continue to be most frequent
in patients with skin, lymph node, and lung metastasis
and less frequent with abdominal and bony metastasis.
best chemotherapy combinations have failed to demon- The propensity of melanoma to metastasize to the brain
strate a survival advantage, and the duration of response is has been a limiting factor, since many of these patients
usually less than 6 months [60]. A recent meta-analysis have responded peripherally only to relapse and die
explored the net benefit of IFN therapy in metastatic with central nervous metastases. The major challenge
malignant melanoma by compiling all randomized trials from these studies using interleukin-2-based biothera-
that compared a treatment regimen with IFN to a non- pies is to determine why one patient will respond with a
IFN containing treatment regimen. Eleven studies were complete remission and another patient will not respond
identified (five unpublished) that collectively comprised at all. Is this related to the state of immunological acti-
1,164 patients. The meta-analysis showed that the regi- vation inherent to the patient or the state achieved by
mens including IFN-alpha improved response rates and these biotherapies or does it somehow relate to the sen-
overall survival compared with regimens without IFN- sitivity of the melanoma cells to biotherapy?
alpha with odds ratios of 0.65 (for response) and 0.69 If it is possible to summarize all the available data
(for survival). In melanoma, the response rate for the into a single response rate by combining a very heterog-
IFN containing regimens was 24% compared with 17% enous group of clinical trials, IL-2 would appear to ben-
(range, 5–30%) for those without IFN [45]. Taken together, efit about one melanoma patient out of every five treated
these data indicate that IFN is an active agent and an with an overall response rate of 18% (Table 7). Complete
important component of treatment regimens for meta- responses with long-term remissions, rare in chemother-
static melanoma. The combination of interferon alpha apy treated patients, are occasionally seen with IL-2.
with chemotherapy met with early encouragement. Many studies have shown a higher percentage of com-
Several phase II studies showed the combination of dac- plete responders with high dose IV bolus IL-2, but no
arbazine and alpha interferon to induce responses in difference in survival compared to lower dose regimens.
25–30% of patients with metastatic melanoma [15, 36, Similar to the situation with alpha interferon, the opti-
93, 113]. In randomized studies comparing the combi- mal dose and schedule of administration have not been
nation to single agent dacarbazine, two showed superior determined, and a clear dose–response relationship has
638 Biological therapy of melanoma

not been demonstrated. IL-2 has been used alone or in technologically demanding and very expensive [65, 66,
combination with lymphokine-activated killer (“LAK”) 83]. To date, the results are not clearly superior to the
cells. The addition of LAK cells, while of historical use of interleukin 2 alone or interleukin with peripheral
importance in the original description of IL-2 therapy blood cells (LAK) activated in vitro. On the other hand,
for advanced malignancies, provides, at best, marginal these cells are exquisitely more specific for the mela-
improvement over IL-2 alone [104, 105]. Good preclinical noma cells. Clearly, this approach validates the belief of
models support the combination of IL-2 and interferon many investigators that specific T-cell reactivity occurs
alpha, but studies, both phase II and randomized trials, in advanced cancer and demonstrates the feasibility of
showed no benefit for this combination in metastatic applying such approaches as a clinical biotherapy [29,
melanoma [115]. 66, 82, 107].
Of historical interest is the combination of chemotherapy Rosenberg has pioneered the use of TIL cells plus
and IL-2. Although the clinical trials combining single interleukin-2 in the treatment of cancer utilizing IL-2/
agent dacarbazine with IL-2 did not demonstrate a ben- TIL with and without cyclophosphamide and in various
efit for combining these modalities (Table 8) [29, 38, 40, schedules and doses in several clinical trials. In sum-
118], cisplatin based regimens were more promising mary, approximately 86 patients were treated; 28 of
[2, 8, 26, 50, 63, 97]. Four trials with cisplatin and IL-2 whom had had prior IL-2 and 58 had no prior IL-2.
have shown overall response rates in excess of 50%, and A 34% response rate was seen for the total group of
all of these also included alpha interferon. Randomized patients with the same response rates in the both the IL-2
studies have not confirmed these data [48]. pretreated patients and the IL-2 naive patients. Both the
response rate and duration of response were equivalent
in the two groups and some of the remissions were of
very long duration.
Interleukin 2/T-cells 111
In labeled TIL cells were given to selected patients
As further evidence for the interleukin-2 mediated effect and with nuclear imaging the presence of these labeled
on melanoma, Rosenberg et al. reported a 50% response cells in tumor tissue was verified. Thus, both based on
rate using T-cells isolated from melanoma nodules and the in vitro studies showing enhanced killing by T cells
grown to large numbers in vitro, another example of compared to LAK cells and in studies using TIL
adoptive immunotherapy of cancer [101, 105]. These cells clinically, where a higher response rate was seen,
tumor-infiltrating lymphocytes (TIL) appear to have a the evidence is accumulating that these more specific T
higher level of specific activity and, in large part, cells are more powerful when used with interleukin-2 in
are MHC-restricted T cells specifically reactive with the treatment of advanced melanoma. It’s of particular
the individual’s melanoma from whom the T cells were interest to note that patients resistant to interleukin-2
derived. Concomitant studies by Dillman and Oldham respond to interleukin-2 plus TIL cells, indicating a role
[30, 65] have confirmed the feasibility of this approach for the cells alone as a therapeutic approach in treating
using tumor-derived activated cells (TDAC) although advanced melanoma. Curiously, no one has performed the
with much lower response rates. This approach of growing kind of T cell dose response studies normally done with
T cells from the tumor and/or draining lymph nodes is drugs in patients with advanced cancer. The major question

Table 8. The activity of combined IL-2 + chemotherapy in malignant melanoma


First author Year Chemotherapy Other therapy No. of patients CR + PR
Papadopoulos [244] 1990 DTIC 30 33% (10)
Flaherty [138] 1990 DTIC 32 22% (7)
Dillman [102] 1990 DTIC 27 26% (7)
Stoter [435] 1991 DTIC 25 24% (6)
Fiedler [131] 1992 DTIC 16 13% (2)
Blair 1991 CDDP, DTIC 28 43% (12)
Demchak [93] 1991 CDDP 27 37% (10)
Hamblin 1991 CDDP, DTIC IFN 12 83% (10)
Richards [372] 1992 CDDP, BCNU IFN, TAM 74 57% (40)
Legha [244] 1992 CDDP, DTIC, VLB IFN 30 56% (17)
Khayat [212] 1993 CDDP IFN 39 54% (21)
Atkins [13] 1994 CDDP, DTIC TAM 38 42% (16)
Robert K. Oldham 639

that remains is what would be the effect of giving who on May 22, 1989, received an infusion of tumor
repeated massive doses of activated T cells to patients infiltrating lymphocytes (TILs) marked with the neomy-
with advanced melanoma? What would be the influence cin phosphotransferase gene (a gene which carries neo-
of dose, schedule, type of T cell, etc.? These studies mycin resistance in bacteria). While this gene transfer
need to be done before conclusions on the efficacy of T cell was not designed for the therapeutic enhancement of
therapy are drawn. TIL activity, the ten melanoma patients who consented
Studies conducted by Oldham and co-workers reported to this gene “therapy” demonstrated that foreign genes
a series of patients with various types of cancer treated could be effectively and safely transferred into human
with a combination of interleukin-2 and tumor-derived cells and administered to patients without apparent
activated cells (TDAC) [65–67, 72, 73, 82, 84, 85, 87, adverse effects on the patient nor risk to the caregivers
88]. These tumor-derived T cells are similar to the TIL or public [102]. This study also confirmed that the engi-
cells described by Rosenberg and in these studies some neered T cells trafficked to the melanoma nodules.
366 tumor biopsies were tested with the T cells grown These results allowed Rosenberg and colleagues to pro-
successfully in 75% of the cases. Sixty-three patients ceed to alter the TILs with therapeutic intent. On January
were treated with a minimum of at least 1010 TDAC by 29, 1991, the first patient treated on a true gene therapy
intravenous infusion and partial responses were seen in protocol was an advanced melanoma patient who
eight patients, including the dramatic complete regres- received an infusion of TILs that contained the gene for
sion of scalp nodules in a patient with renal cancer. The tumor necrosis factor (TNF). Several metastatic mela-
cells were well tolerated, responses were seen in patients noma and other advanced cancer patients were treated
with renal cancer and melanoma, most of whom were in this early study, and responses have been observed
previously resistant to interleukin-2 and all of whom had [101]. The rationale for this strategy is to provide high
bulky, advanced cancer [66]. Taken together, the results local concentrations of TNF at tumor sites. TNF is quite
of T-cell therapy indicate that these cells are active in toxic when systemically administered, but presumably
advanced cancer. The observation that they are active in the TILs provide sufficient local concentrations of TNF
patients previously resistant to Interleukin-2 demon- for an antitumor effect while sparing systemic toxicity.
strates that the cells alone have consistent activity in a Another gene therapy approach initially described by
minority of patients with advanced cancer. Nabel and colleagues is to introduce the HLA B-7 gene
Over the last few years, Rosenberg and a few other directly into cutaneous melanomas of non-HLA B-7
investigators in Europe have begun to look at ways to patients with metastatic disease [79]. The concept was
manipulate the immune system prior to infusing the to induce a cytotoxic immune response against a foreign
activated T-cells. Very aggressive therapy such as com- major histocompatibility complex (MHC) class I antigen,
binations of high-dose chemotherapy in the setting of an leading to the recognition of the injected cutaneous
autologous transplant plus whole body radiation therapy melanoma cell as foreign and ultimately generating a
followed by the infusion of autologous T-cells and systemic anti-melanoma immune response. In the initial
Interleukin-2, has resulted in a greater than 75% study, the five patients treated locally injected with the
response rate in the first few patients treated with this HLA B7 gene all showed evidence of safe and success-
approach at the NCI. Many of these patients had bulky, ful gene transfer, cytotoxic T-lymphocytes were shown
Interleukin-2 resistant disease, and the induction of to invade the lesions, and one patient showed tumor
complete responses in such patients is very encouraging regression not only of the lesion that had the HLA B7
[70, 103]. gene delivery but also of distant cutaneous and pulmo-
nary metastases [79]. The initial success of this strategy
has lead to a number studies with the HLA B7 gene
(commercialized as “Allovectin 7”), now most com-
Gene therapy monly administered intramuscularly as naked DNA.
A new category of biological therapy has become recog- Further details of this line of investigation may be found
nized, utilizing gene engineered cells. By definition, in the chapters on gene therapy and vaccines elsewhere
gene therapy is the introduction of genetic material into in this book. Other genes under evaluation include gran-
cells for therapeutic purposes. The gene therapy of cancer ulocyte macrophage colony stimulating factor (GM-CSF)
has focused on advanced melanoma and renal cancer and B2-microglobin, whose gene products will induce an
models for the development of this new therapeutic antitumor immune response when inactivated autologous
approach. The first patient treated on an approved gene tumor cells are reinfused into the patient. The cytokine
therapy protocol was a patient with malignant melanoma gene therapy concept has been expanded in other clinical
640 Biological therapy of melanoma

trials that will evaluate the feasibility of genes for IL-2, antigen GD3, appears to have induced clinical responses
gamma interferon, TNF, and IL-4 transduced into autol- (four partial and two minor responses among 21 patients)
ogous tumor cells or fibroblasts to produce active cytok- using unconjugated antibody [122]. These studies are in
ines effective in the treatment of melanoma and other contrast to previous studies using other anti-melanoma
cancers [22]. To date, the clinical benefit and activity of antibodies. MAb R24 provoked a clear inflammatory
gene therapy for melanoma remains to be fully delin- reaction with increased number of mast cells with evi-
eated [5, 17, 49, 74, 76]. dence of their degranulation, an influx of polymorpho-
nuclear cells, complement deposition, particularly C3,
C5, and C9, and infiltration with T3+/T8+/Ia+ lympho-
cytes. MAb R24 has been studied in conjunction with
Antibodies other biological modalities such as IL-2 [4], interferon-
Relatively specific visualization of tumor nodules with alfa-2a, with total lymphoid irradiation (which did not
radiolabeled antibody has been reported [13] and anti- interdict the human anti-murine antibody response as
body given intravenously can subsequently be demon- had been hoped) and by novel routes of administration
strated on the melanoma cell surface by histopathologic (e.g., isolated limb perfusion) [16]. Cheung et al. have
techniques [111]. However, only minor clinical activity studied another IgG3 anti-ganglioside MAb, 3F8, in a
has been observed with either unconjugated antibody or phase I trial [14]. Two of nine patients with metastatic
with preparations of radiolabeled, drug-conjugated, or malignant melanoma achieved partial responses, one
toxin-conjugated anti-melanoma monoclonal antibodies. lasting 22 weeks and the other continuing at 56 weeks.
The 9.2.27 antimelanoma monoclonal antibody rec- The most prominent toxicities consisted of severe pain,
ognizes a melanoma-associated antigen (a 250-kD hypertension, and focal urticaria; a decrease in serum
chondroitin sulfate proteoglycan core glycoprotein) that complement activity was observed when dosages
is found on 90% of melanoma cells and relatively few equaled or exceeded 20 mg/m2. Houghton reported that
non-melanoma cells [86]. Oldham et al. reported the the combination of MAb R24 and MAb 3F8 yielded one
selective targeting of this antibody to biopsied mela- complete remission among 13 patients with melanoma.
noma nodules in eight patients [86]. The 9.2.27 antibody Goodman et al. observed no anti-tumor activity among
does not activate complement, poorly activates ADCC, eight patients given MAb MG-21, another antibody rec-
but may have application because its target antigen ognizing a GD3 surface antigen commonly displayed
modulates very little. Their studies suggest that quanti- by human melanoma cells [44, 45]. In an intriguing
ties on the order of 0.5 to several grams of antibody are report, Livingston and associates reported one complete
necessary for saturation of target antigens in vivo. response among 13 patients with metastatic malignant
Schroff et al. have observed that an interrupted infu- melanoma who received murine IgG2a MAb ME-36.1,
sional schedule appears to compromise the in vivo local- which recognizes the melanoma-associated ganglio-
ization of antibody to tumor, presumably by the sides GD2 and GD3. This patient’s posttreatment B
development of human anti-murine antibodies (HAMA), lymphocytes, when stimulated with polyclonal goat
which impede antibody localization and accelerate anti-idiotypic ME-36.1, were found to synthesize human
clearance [111]. Goodman et al. later administered antibodies preferentially and specifically recognizing
MAb96.5 and MAb48.7 to four patients and MAb96.5 GD2, suggesting to the researchers that the induction of
alone to a fifth patient. MAb96.5 is an IgG2a immuno- human anti-melanoma ganglioside antibodies provoked
globulin that recognizes p97, a transferrin-like cell sur- by ME-36.1 may have mediated the observed clinical
face glycoprotein of 97 kD [44, 45]. MAb48.7 is an response. These observations that murine monoclonal
IgG1 immunoglobulin recognizing a melanoma-associ- antibodies can induce complement activation, active
ated cell surface proteoglycan. Although the infusion of antibody-dependent cellular cytotoxicity, and generate
antibody was well tolerated, there were no objective human anti-melanoma antibodies may support a further
remissions nor histopathologic changes found in biop- role for the investigation of unconjugated antibody in
sied tissue, although impressive antibody binding to malignant melanoma.
melanoma cells was observed. Perhaps the lack of activity A murine monoclonal anti-melanoma antibody-ricin
reflected the lack of in vitro activation of human com- A chain immunotoxin (XOMAZYME-MEL) was stud-
plement and very modest ADCC. ied in several clinical trials, since the initial report of its
Studies by Vadhan-Raj et al. [122] are of interest in use in 22 patients in early 1987 [113]. The monoclonal
that the IgG3 anti-GD3 mouse monoclonal antibody antibody moiety of this immunotoxin is an IgG2A anti-
R24, directed against the sialoganglioside membrane body recognizing melanoma-associated proteoglycan
Robert K. Oldham 641

membrane antigens of 220 kD and greater than 500 kD. 6. Beck KE, Blansfield JA, Tran, KQ et al. Enterocolitis in patients with
One patient demonstrated complete disappearance of cancer after antibody blockade of cytotoxic T-lymphocytes-associated
antigen 4. J Clin Oncol May 20, 2006; 24(15):2283–2289.
detectable melanoma (a retrocardiac pulmonary metasta- 7. Berd D, Maguire HC, Jr., Schuchter LM et al. Autologous hapten-
sis) over an 8-month period following a single infusion modified melanoma vaccine as postsurgical adjuvant treatment
of immunotoxin, with maintenance of a complete remis- after resection of nodal metastases. J Clin Oncol 1997; 15:
sion for greater than 2 years until relapse occurred in the 2359–2370.
8. Blair S, Flaherty L, Valdivieso M. Comparison of high dose inter-
brain. However, no major responses were seen in the
leukin 2 with combined chemotherapy and low dose IL2 in meta-
remaining 21 patients. The treatment was accompanied static malignant melanoma. Proc Am Soc Clin Oncol 2003; 3:294
by a transient reduction of serum albumin with associated (abstract).
fluid retention and weight gain, alopecia, fever, fatigue, 9. Borden EC. Introduction. In: Borden EC, ed. Melanoma: biologi-
and malaise, with mild allergic reactions occurring in cally targeted therapeutics. Totowa, NJ: Humana, 2002.
10. Borgstrom S, von Eyben FE, Flodgren P, Axelsson B, Sjogren HO.
three patients. In a phase II trial with this immunotoxin,
Human leukocyte interferon and cimetidine for metastatic mela-
three partial responses were observed among 43 patients, noma. N Engl J Med 1982; 307:1080–1081.
these responses lasting for 10+, 13+, and 15+ months 11. Budd GT, Osgood B, Barna B et al. Phase I clinical trial of inter-
[87, 116]. Cyclophosphamide at a dose of 1 g/m2 admin- leukin 2 and alpha-interferon: toxicity and immunologic effects.
istered following the infusion of immunotoxin has not Cancer Res 1989; 49:6432–6436.
12. Cameron DA, Cornbleet MC, Mackie RM et al. Adjuvant inter-
inhibited the generation of host immune responses feron alpha 2b in high risk melanoma – the Scottish study. Br J
directed against the immunotoxin preparation. Cancer 2001; 84:1146–1149.
Given the studies by various investigators using unla- 13. Carrasquillo JA, Abrams PG, Schroff RW et al. Effect of antibody
beled antibodies, which have been marginally useful in dose on the imaging and biodistribution of indium- 111 9.2.27
the treatment of melanoma, several investigators have anti-melanoma monoclonal antibody. J Nucl Med 1988; 29:
39–47.
pursued studies using antibody in combination with 14. Cheung NK, Lazarus H, Miraldi FD et al. Ganglioside GD2 spe-
cytokines [4, 21] with no evidence of activity over and cific monoclonal antibody 3F8: a phase I study in patients with
above using the cytokines alone. Similarly, early trials neuroblastoma and malignant melanoma. J Clin Oncol 1987;
with radiolabeled antibodies for therapy of melanoma 5:1430–1440.
15. Coates A, Rallings M, Hersey P, Swanson C. Phase-II study of
have not proven useful [57].
recombinant alpha 2-interferon in advanced malignant melanoma.
Since antibodies can induce inflammatory infiltrates J Interferon Res 1986; 6:1–4.
and activities around melanoma nodules, further work is 16. Coit D, Houghton A, Corden-Cardo C. Isolation limb perfusion
ongoing with regard to using these antibodies to produce with monoclonal antibody R24 in patients with malignant lym-
inflammatory effects and combining these effects with phoma. Proc Am Soc Clin Oncol 1988; 7:248 (abstract).
17. Cooper, LJN. Long live T cells. Blood January 2005; 105(1):8–9.
cytokines and chemotherapy. Engineered subfragments
18. Creagan ET, Dalton RJ, Ahmann DL et al. Randomized, surgical
of antibody as well as humanized antibodies are currently adjuvant clinical trial of recombinant interferon alfa-2a in selected
in clinical trials and it is felt some of these improved mol- patients with malignant melanoma. J Clin Oncol 1995; 13:
ecules may improve the delivery of immunotoxins and/or 2776–2783.
radionuclides to melanoma deposits [89]. 19. Creagan ET, Ahmann DL, Green SJ et al. Phase II study of low-
dose recombinant leukocyte A interferon in disseminated malig-
nant melanoma. J Clin Oncol 1984; 2:1002–1005.
20. Creagan ET, Ahmann DL, Green SJ et al. Phase II study of recom-
References binant leukocyte A interferon (rIFN-alpha A) in disseminated
malignant melanoma. Cancer 1984; 54:2844–2849.
1. Ahn SS, Irie RF, Weisenburger TH et al. Humoral immune response to 21. Creekmore S, Urba W, Koop W. Phase IB/II trial of R24 antibody
intralymphatic immunotherapy for disseminated melanoma: correlation and interleukin-2 (IL2) in melanoma. Proc Am Soc Clin Oncol
with clinical response. Surgery 1982; 92:362–367. 1992; 1886:345 (abstract).
2. Atkins MB, O’Boyle KR, Sosman JA et al. Multiinstitutional phase II 22. Culver KW, Blaese RM. Gene therapy for cancer. Trends Genet
trial of intensive combination chemoimmunotherapy for metastatic 1994; 10:174–178.
melanoma. J Clin Oncol 1994; 12:1553–1560. 23. Dantal J, Hourmant M, Cantarovich D et al. Effect of long-term
3. Bajetta E, Di Leo A, Zampino MG et al. Multicenter randomized immunosuppression in kidney-graft recipients on cancer inci-
trial of dacarbazine alone or in combination with two different doses dence: randomised comparison of two cyclosporin regimens.
and schedules of interferon alfa-2a in the treatment of advanced Lancet 1998; 351:623–628.
melanoma. J Clin Oncol 1994; 12:806–811. 24. Das D, Ata UR. A novel method for eliminating the melanin pig-
4. Bajorin DF, Chapman PB, Wong G et al. Phase I evaluation of a ments from melanoma cells undergoing cytogenetic analysis in cases
combination of monoclonal antibody R24 and interleukin 2 of uveal melanoma. Cancer Genet Cytogenet 2001; 127:181–183.
in patients with metastatic melanoma. Cancer Res 1990; 50: 25. Day CL, Jr., Lew RA, Mihm MC, Jr. et al. A multivariate analysis
7490–7495. of prognostic factors for melanoma patients with lesions greater
5. Baum BJ, Cotrim AP. Gene therapy: some history, applications, than or equal to 3.65 mm in thickness. The importance of revealing
problems, and prospects. Toxicol Path 2008; 26(1):97–103. alternative Cox models. Ann Surg 1982; 195:44–49.
642 Biological therapy of melanoma

26. Demchak PA, Mier JW, Robert NJ, O’Brien K, Gould JA, Atkins cell carcinoma: an overview of randomized trials. J Immunother
MB. Interleukin-2 and high-dose cisplatin in patients with meta- 1999; 22:145–154.
static melanoma: a pilot study. J Clin Oncol 1991; 9:1821–1830. 47. Hill NO, Pardue A, Khan A et al. Interferon and cimetidine for
27. Dillman RO. Monoclonal antibodies for treating cancer. Ann malignant melanoma. N Engl J Med 1983; 308:286.
Intern Med 1989; 111:592–603. 48. Ives NJ, Stowe RL, Lorigan P et al. Chemotherapy compared with
28. Dillman RO, Church C, Oldham RK, West WH, Schwartzberg L, biochemotherapy for the treatment of metastatic melanoma: a
Birch R. Inpatient continuous-infusion interleukin-2 in 788 meta-analysis of 18 trials involving 2,621 patients. J Clin Oncol
patients with cancer. The National Biotherapy Study Group expe- December 1, 2007; 24(34):5426–5434.
rience. Cancer 1993; 71:2358–2370. 49. Johnson LA, Heemskerk B, Powell DJ et al. Gene transfer of
29. Dillman RO, Oldham RK, Barth NM, Birch R, Arnold J, West tumor-reactive TCR confers both high avidity and tumor reactivity
WH. Recombinant interleukin-2 and adoptive immunotherapy to nonreactive peripheral blood mononuclear cells and tumor-infil-
alternated with dacarbazine therapy in melanoma: a National trating lymphocytes. J Immunol 2006; 177:6548–6559.
Biotherapy Study Group trial. J Natl Cancer Inst 1990; 82: 50. Khayat D, Borel C, Tourani JM et al. Sequential chemoimmuno-
1345–1349. therapy with cisplatin, interleukin-2, and interferon alfa-2a for
30. Dillman RO, Oldham RK, Barth NM et al. Continuous interleukin-2 metastatic melanoma. J Clin Oncol 1993; 11:2173–2180.
and tumor-infiltrating lymphocytes as treatment of advanced mela- 51. Kirkwood JM, Ernstoff M. Potential applications of the interferons
noma. A national biotherapy study group trial. Cancer 1991; 68:1–8. in oncology: lessons drawn from studies of human melanoma.
31. Dillman RO, Oldham RK, Tauer KW et al. Continuous interleu- Semin Oncol 1986; 13:48–56.
kin-2 and lymphokine-activated killer cells for advanced cancer: a 52. Kirkwood JM, Harris JE, Vera R et al. A randomized study of low
National Biotherapy Study Group trial. J Clin Oncol 1991; and high doses of leukocyte alpha-interferon in metastatic renal
9:1233–1240. cell carcinoma: the American Cancer Society collaborative trial.
32. Dorval T, Palangie T, Jouve M et al. Clinical phase II trial of Cancer Res 1985; 45:863–871.
recombinant DNA interferon (interferon alpha 2b) in patients with 53. Kirkwood JM, Ibrahim JG, Sondak VK et al. High- and low-dose
metastatic malignant melanoma. Cancer 1986; 58:215–218. interferon alfa-2b in high-risk melanoma: first analysis of intergroup
33. Dranoff G. CTLA-4 blockade: unveiling immune regulation. J trial E1690/S9111/C9190. J Clin Oncol 2000; 18:2444–2458.
Clin Oncol February 1, 2005; 23(4):662–664. 54. Kirkwood JM, Ibrahim JG, Sosman JA et al. High-dose interferon
34. Dreno B, Mansat E, Legoux B, Litoux P. Skin cancers in transplant alfa-2b significantly prolongs relapse-free and overall survival
patients. Nephrol Dial Transplant 1998; 13:1374–1379. compared with the GM2-KLH/QS-21 vaccine in patients with
35. Elsasser-Beile U, Drews H. Interferon in the treatment of malig- resected stage IIB-III melanoma: results of intergroup trial E1694/
nant melanoma. Results of clinical studies. Fortschr Med 1987; S9512/C509801. J Clin Oncol 2001; 19:2370–2380.
105:401–403, 42. 55. Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden
36. Falkson CI, Falkson G, Falkson HC. Improved results with the EC, Blum RH. Interferon alfa-2b adjuvant therapy of high-risk
addition of interferon alfa-2b to dacarbazine in the treatment of resected cutaneous melanoma: the Eastern Cooperative Oncology
patients with metastatic malignant melanoma. J Clin Oncol 1991; Group Trial EST 1684. J Clin Oncol 1996; 14:7–17.
9:1403–1408. 56. Kirkwood JM. Building upon the standard of care in adjuvant
37. Ferradini L, Mackensen A, Genevee C et al. Analysis of T cell therapy of high-risk melanoma. J Clin Oncol December 1, 2005;
receptor variability in tumor-infiltrating lymphocytes from a 23(34):8559–8563.
human regressive melanoma. Evidence for in situ T cell clonal 57. Larson SM, Carrasquillo JA, Krohn KA et al. Localization of
expansion. J Clin Invest 1993; 91:1183–1190. 131I-labeled p97-specific Fab fragments in human melanoma as a
38. Fiedler W, Jasmin C, De Mulder PH et al. A phase II study of basis for radiotherapy. J Clin Invest 1983; 72:2101–2114.
sequential recombinant interleukin-2 followed by dacarbazine in 58. Lee KH, Talpaz M, Rothberg JM et al. Concomitant administration
metastatic melanoma. Eur J Cancer 1992; 28:443–446. of recombinant human interleukin-2 and recombinant interferon
39. Fintor L. Melanoma vaccine momentum spurs interest, invest- alpha-2A in cancer patients: a phase I study. J Clin Oncol 1989;
ment. J Natl Cancer Inst 2000; 92:1205–1207. 7:1726–1732.
40. Flaherty LE, Redman BG, Chabot GG et al. A phase I-II study of 59. Lee ML, Tomsu K, Von Eschen KB. Duration of survival for
dacarbazine in combination with outpatient interleukin-2 in meta- disseminated malignant melanoma: results of a meta-analysis.
static malignant melanoma. Cancer 1990; 65:2471–2477. Melanoma Res 2000; 10:81–92.
41. Greenlee RT, Hill-Harmon MB, Murray T, Thun M. Cancer statis- 60. Legha SS. Current therapy for malignant melanoma. Semin Oncol
tics, 2001. CA Cancer J Clin 2001; 51:15–36. 1989; 16:34–44.
42. Gogas H, Ioannovich J, Dafni U et al. Prognotstic significance of 61. Legha SS. The role of interferon alfa in the treatment of metastatic
autoimmunity during treatment of melanoma with interferon. melanoma. Semin Oncol 1997; 24:S24–S31.
N Engl J Med February 16, 2006; 354(7):709–718. 62. Legha SS, Papadopoulos NE, Plager C et al. Clinical evaluation
43. Goldberg RM, Ayoob M, Silgals R, Ahlgren JD, Neefe JR. Phase of recombinant interferon alfa-2a (Roferon-A) in metastatic mela-
II trial of lymphoblastoid interferon in metastatic malignant mela- noma using two different schedules. J Clin Oncol 1987;
noma. Cancer Treat Rep 1985; 69:813–816. 5:1240–1246.
44. Goodman GE, Beaumier P, Hellstrom I, Fernyhough B, Hellstrom 63. Legha SS, Ring S, Eton O, Bedikian A, Plager C, Papadopoulos N.
KE. Pilot trial of murine monoclonal antibodies in patients with Development and results of biochemotherapy in metastatic mela-
advanced melanoma. J Clin Oncol 1985; 3:340–352. noma: the University of Texas M.D. Anderson Cancer Center
45. Goodman GE, Hellstrom I, Brodzinsky L. Phase I trial of monoclonal experience. Cancer J Sci Am 1997; 3 Suppl 1:S9–S15.
antibody MG-21 directed against a mlelanoma-associated GD3 gan- 64. Lens MB, Dawes M, Goodacre T, Newton-Bishop JA. Elective
glioside antigen. Proc Am Soc Clin Oncol 1987; 6:209 (abstract). lymph node dissection in patients with melanoma: systematic
46. Hernberg M, Pyrhonen S, Muhonen T. Regimens with or without review and meta-analysis of randomized controlled trials. Arch
interferon-alpha as treatment for metastatic melanoma and renal Surg 2002; 137:458–461.
Robert K. Oldham 643

65. Lewko WM, Good RW, Bowman D, Smith TL, Oldham RK. 84. Oldham RK. Therapy with interleukin-2 and tumor-derived acti-
Growth of tumor derived activated T-cells for the treatment of can- vated lymphocytes. Immunol Ser 1994; 61:251–271.
cer. Cancer Biother 1994; 9:211–224. 85. Oldham RK, Dillman RO, Yannelli JR et al. Continuous infusion
66. Lewko WM, Hall PB, Oldham RK. Growth of tumor-derived acti- interleukin-2 and tumor-derived activated cells as treatment of
vated T cells for the treatment of advanced cancer. Cancer Biother advanced solid tumors: a National Biotherapy Study Group Trial.
Radiopharm 2000; 15:357–366. Mol Biother 1991; 3:68–73.
67. Lewko WM, Smith TL, Bowman DJ, Good RW, Oldham RK. 86. Oldham RK, Foon KA, Morgan AC et al. Monoclonal antibody
Interleukin-15 and the growth of tumor derived activated T-cells. therapy of malignant melanoma: in vivo localization in cutane-
Cancer Biother 1995; 10:13–20. ous metastasis after intravenous administration. J Clin Oncol
68. Lipton A, Harvey HA, Simmonds MA. Lack of enhanced activity 1984; 2:1235–1244.
of systemic interferon by cimetidine in malignant melanoma. Proc 87. Oldham RK, Lewko WM, Good RW, Sharp E. Cancer biotherapy
Am Soc Clin Oncol 1984; 3:56 (abstract). with interferon, interleukin-2 and tumor-derived activated cells
69. Livingston PO, Ritter G, Srivastava P et al. Characterization of (TDAC). In Vivo 1994; 8:653–663.
IgG and IgM antibodies induced in melanoma patients by immuni- 88. Oldham RK, Maleckar JR, Friddell CS, Lewko WM, West WH,
zation with purified GM2 ganglioside. Cancer Res 1989; Yannelli JR. Tumor-derived activated cells: preliminary labora-
49:7045–7050. tory and clinical results. Clin Chem 1989; 35:1576–1580.
70. Lizee G, Radvanyi LG, Overwijk WW et al. Improving antitumor 89. Oldham RK, Dillman RO. Monelonal antibodies in Cancer
immune responses by circumventing immunoregularity cells and Therapy: 25 years of Progress JCO 2008; 26(11):1774–1777.
mechanisms. Clin Canc Res August 13, 2006; 12:4794–4803. 90. Paterson AH, Willans DJ, Jerry LM, Hanson J, McPherson TA.
71. Mackenson A, Cercelain G, Viels S. Direct evidence to support the Adjuvant BCG immunotherapy for malignant melanoma. Can
immunosurveillance concept in a regressive melanoma. J Clin Med Assoc J 1984; 131:744–748.
Invest 1994; 93:1402. 91. Peggs KS, Quezada SA, Korman AJ et al. Principles and use of
72. Maleckar JR, Friddell CS, Lewko WM, Yannelli JR, West WH, anti-CTLA4 antibody in human cancer immunotherapy. Curr
Oldham RK. Tumor-derived activated cells: culture conditions and Opin Immun April 2006; 18(2):206–213.
characterization. Immunol Ser 1989; 48:159–173. 92. Prehn RT. The paradoxical association of regression with a poor
73. Maleckar JR, Friddell CS, Sferruzza A et al. Activation and expan- prognosis in melanoma contrasted with a good prognosis in kera-
sion of tumor-derived activated cells for therapeutic use. J Natl toacanthoma. Cancer Res 1996; 56:937–940.
Cancer Inst 1989; 81:1655–1660. 93. Pyrhonen S, Hahka-Kemppinen M, Muhonen T. A promising
74. Michalek J, Buchler T, Hajek R. T lymphocyte therapy of cancer. interferon plus four-drug chemotherapy regimen for metastatic
Physiol Res 2004; 33:463–469. melanoma. J Clin Oncol 1992; 10:1919–1926.
75. Mitchell MS, Abrams J, Thompson JA et al. Randomized trial of 94. Reintgen DS, McCarty KS, Jr., Cox E, Seigler HF. Malignant
an allogeneic melanoma lysate vaccine with low-dose interferon melanoma in the American black. Curr Surg 1983; 40:215–217.
alfa-2b compared with high-dose interferon alfa-2b for resected 95. Retsas S, Priestman TJ, Newton KA, Westbury G. Evaluation of
stage III cutaneous melanoma. J Clin Oncol May 20, 2007; human lymphoblastoid interferon in advanced malignant mela-
25(15):2078–2085. noma. Cancer 1983; 51:273–276.
76. Morgan RA, Dudley ME, Yu YYL et al. High efficiency TCR gene 96. Ribas A, Hanson DC, Noe DA et al. Tremelimumab (CP-
transfer into primary human lymphocytes affords avid recognition 675,206), a cytotoxic T lymphocyte associated antigen 4 blocking
of melanoma tumor antigen glycoprotein 100 and does not alter monoclonal antibody in clinical development for patients with
the recognition of autologous melanoma antigens. J Immunol cancer. Oncologist July 1, 2007; 12(7):873–883.
2003; 171:3287–3295. 97. Richards JM, Mehta N, Ramming K, Skosey P. Sequential
77. Morton DL. Active immunotherapy against cancer: present status. chemoimmunotherapy in the treatment of metastatic melanoma.
Semin Oncol 1986; 13:180–185. J Clin Oncol 1992; 10:1338–1343.
78. Morton DL, Mozzillo N, Thompson JF et al. An internation, ran- 98. Robinson E, Bartal A, Cohen Y, Haasz R, Mekori T. Treatment of
domized, phase III trial of bacillus Calmete-Guerin (BCG) plus lung cancer by radiotherapy, chemotherapy, and methanol extrac-
allogeneic melanoma vaccine (MCV) or placebo after complete tion residue of BCG (MER): clinical and immunological studies.
resection of melanoma metastatic to regional or distant sites. J Clin Cancer 1977; 40:1052–1059.
Oncol 2007 ASCO Annual Meeting Proceedings June 20, 2007 99. Roeslin N, Lang JM, Morand G, Wihlm JM, Witz JP. Regional
supplement; 25(185):8508. immunotherapy in resectable squamous cell lung carcinoma.
79. Nabel GJ, Nabel EG, Yang ZY et al. Direct gene transfer with Analysis of a randomized study. Cancer Immunol Immunother
DNA-liposome complexes in melanoma: expression, biologic 1982; 13:174–175.
activity, and lack of toxicity in humans. Proc Natl Acad Sci U S A 100. Rosenberg SA. The adoptive immunotherapy of cancer using the
1993; 90:11307–11311. transfer of activated lymphoid cells and interleukin-2. Semin
80. Neidhart JA, Murphy SG, Hennick LA, Wise HA. Active specific Oncol 1986; 13:200–206.
immunotherapy of stage IV renal carcinoma with aggregated 101. Rosenberg SA. Karnofsky Memorial Lecture. The immunother-
tumor antigen adjuvant. Cancer 1980; 46:1128–1134. apy and gene therapy of cancer. J Clin Oncol 1992; 10:180–199.
81. Oldham RK. Biologicals for cancer treatment: interferons. Hosp 102. Rosenberg SA, Aebersold P, Cornetta K et al. Gene transfer into
Pract 1985; 20:72–91. humans – immunotherapy of patients with advanced melanoma,
82. Oldham RK, Maleckar, JR, Wart WH & Yanelli JR IK2 and Cellular using tumor-infiltrating lymphocytes modified by retroviral gene
Therapy In: Cytokines in Hemapoirsis Oncology and AIDS Eds transduction. N Engl J Med 1990; 323:570–578.
Freevel CR et al Springer-Verlag, Berlin pp 661–671 1990. 103. Rosenberg SA, Dudley, ME. Cancer regression in patients with
83. Oldham RK. Cancer cures: by the people, for the people, at what metastatic melanoma after the transfer of autologous antitumor
cost? Mol Biother 1990; 2:2–3. lymphocytes. PNAS1 10.1073; September 20, 2004.
644 Biological therapy of melanoma

104. Rosenberg SA, Lotze MT, Yang JC. Prospective randomized trial 115. Sparano JA, Fisher RI, Sunderland M et al. Randomized phase III
of high-dose interleukin-2 alone or in conjunction with lym- trial of treatment with high-dose interleukin-2 either alone or in
phokine-activated killer cells for the treatment of patients with combination with interferon alfa-2a in patients with advanced
advanced cancer. J Natl Cancer Inst 1993; 85:622. melanoma. J Clin Oncol 1993; 11:1969–1977.
105. Rosenberg SA, Lotze MT, Yang JC et al. Experience with the use 116. Spitler LE, del Rio M, Khentigan A et al. Therapy of patients with
of high-dose interleukin-2 in the treatment of 652 cancer patients. malignant melanoma using a monoclonal antimelanoma antibody-
Ann Surg 1989; 210:474–484. ricin A chain immunotoxin. Cancer Res 1987; 47:1717–1723.
106. Rosenberg SA, Lotze MT, Yang JC et al. Combination therapy 117. Steiner A, Wolf C, Pehamberger H. Comparison of the effects of
with interleukin-2 and alpha-interferon for the treatment of three different treatment regimens of recombinant interferons
patients with advanced cancer. J Clin Oncol 1989; 7:1863–1874. (r-IFN alpha, r-IFN gamma, and r-IFN alpha + cimetidine) in dis-
107. Rosenberg SA, Packard BS, Aibersold PM et al. Use of tumor- seminated malignant melanoma. J Cancer Res Clin Oncol 1987;
infiltrating lymphocytes and interleukin-2 in the immunotherapy 113:459–465.
of patients with metastatic melanoma. A preliminary report. N 118. Stoter G, Aamdal S, Rodenhuis S et al. Sequential administration
Engl J Med 1988; 319:1676–1680. of recombinant human interleukin-2 and dacarbazine in meta-
108. Ruotsalainen TM, Halme M, Tamminen K et al. Concomitant static melanoma: a multicenter phase II study. J Clin Oncol 1991;
chemotherapy and IFN-alpha for small cell lung cancer: a ran- 9:1687–1691.
domized multicenter phase III study. J Interferon Cytokine Res 119. Tahini AA, Kirkwood JM, Gooding WE et al. Durable complete
1999; 19:253–259. responses with high-dose bolus interleukin-2 in patients with meta-
109. Sanderson K, Scotland R, Lee P et al. Autoimmunity in a phase I static melanoma who have experienced progression after biochemo-
trial of a fully human anti-cytotoxic T-lymphocyte antigen-4 therapy. J Clin Oncol September 1, 2007; 25(25):3802–3807.
monoclonal antibody with multiple melanoma peptides and mon- 120. Thomson DB, Adena M, McLeod GR et al. Interferon-alpha 2a does
tanide ISA 51 for patients with resected stages III and IV mela- not improve response or survival when combined with dacarbazine
noma. J Clin Oncol February 1, 2005; 23(4):741–750. in metastatic malignant melanoma: results of a multi-institutional
110. Schmid-Wendtner MH, Baumert J, Schmidt M et al. Late metas- Australian randomized trial. Melanoma Res 1993; 3:133–138.
tases of cutaneous melanoma: an analysis of 31 patients. J Am 121. Tsao H, Rogers GS, Sober AJ. An estimate of the annual direct
Acad Dermatol 2000; 43:605–609. cost of treating cutaneous melanoma. J Am Acad Dermatol 1998;
111. Schroff RW, Woodhouse CS, Foon KA et al. Intratumor localiza- 38:669–680.
tion of monoclonal antibody in patients with melanoma treated 122. Vadhan-Raj S, Cordon-Cardo C, Carswell E et al. Phase I trial of
with antibody to a 250,000-dalton melanoma-associated antigen. a mouse monoclonal antibody against GD3 ganglioside in
J Natl Cancer Inst 1985; 74:299–306. patients with melanoma: induction of inflammatory responses at
112. Sertoli MR, Bernengo MG, Ardizzoni A et al. Phase II trial of tumor sites. J Clin Oncol 1988; 6:1636–1648.
recombinant alpha-2b interferon in the treatment of metastatic 123. Vosika GJ. Clinical immunotherapy trials of bacterial compo-
skin melanoma. Oncology 1989; 46:96–98. nents derived from mycobacteria and nocardia. J Biol Resp
113. Sertoli MR, Quierolo P, and Bajetta E. Dacarbazine (DTIC) with or Modif 1983; 2:321–342.
without recombinant interferon alpha 2a at different dosages in the 124. West WH, Tauer KW, Yannelli JR et al. Constant-infusion recom-
treatment of stage IV melanoma patient: preliminary results of a binant interleukin-2 in adoptive immunotherapy of advanced
randomized trial. Proc Am Soc Clin Oncol 1992; 11:345 (abstract). cancer. N Engl J Med 1987; 316:898–905.
114. Sosman JA, Hank JA, Sondel PM. In vivo activation of lym- 125. Wheatley K, Hancock B, Gore M, Suciu S. Eggermont AMM.
phokine-activated killer activity with interleukin-2: prospects for Interferon-α as adjuvant therapy for melanoma: a meta-analysis of the
combination therapies. Semin Oncol 1990; 17:22–30. randomised trials. Proc Am Soc Clin Oncol 2001; 20:1394 (abstract).
21.3 Biological therapy of genitourinary cancer
ROBERT K. OLDHAM

Kidney Cancer kidney cancer, these statistics illustrate the large potential
patient population that can benefit from an active biologi-
Overview cal and targeted therapy of cancer.
Recent progress in targeted therapy for renal cancer
Kidney cancer remains one of the best examples of a has lead to the identification of several agents, which
disease that responds to the immune system and is ame- target either vascular endothelial growth factor or a
nable to intervention with biological therapy. Cancer of tyrosine kinase enzyme. These molecular findings have
the kidney and renal pelvis was estimated to account for lead to the development of what is now being called
51,200 new cancer cases and cause 12,900 cancer deaths “targeted therapy” of kidney cancer and many other
in the United States in 2007. In the United States, kidney types of cancer [15].
cancer is the 12th leading cause of cancer death, com-
prising 3% of all malignancies in men and 2% in women.
The 5-year relative cancer free survival has improved Cytotoxic Chemotherapy is Ineffective
over the last 2 decades, from 52% in the years 1974–
Kidney cancer remains one of few modern examples of a
1976 to the current rate of 61% in the years 1989–2007,
malignant disease for which currently available cytotoxic
causing 2% of all cancer deaths in this country [32].
chemotherapy agents are essentially inactive. An exhaus-
Internationally, kidney cancer has the highest incidence
tive review of 143 published or nationally presented sin-
in North America and northern Europe, the lowest inci-
gle agent chemotherapy trials concluded that no agent
dence in India, China, Japan, and South America, and
achieved significant activity in the treatment of renal cell
accounts for 2% of all cancers worldwide [43, 56, 79].
carcinoma. This review included studies reported from
The incidence of kidney cancer has increased by
1975–1995 involving almost 4,000 patients treated with
approximately 2% per year over the past 2 decades. The
80 different single agents, showing a 4% overall response
greatest rise in incidence is in patients with localized dis-
rate [2]. Newer cytotoxic agents have had similarly disap-
ease (3.7% per year] and may be ascribed in large part to
pointing results in renal cell carcinoma. A review of che-
the increase in abdominal imaging in the population at
motherapy trials reported from 1990–1998 included 33
large. However, the incidence of regionally advanced
chemotherapy agents given to 1,347 patients on 51 phase
and distantly metastatic kidney cancer has also increased
II clinical trials lead to the conclusion that no chemother-
over this time period, indicating that factors in addition
apy has produced response rates that justify use as a sin-
to the increase in abdominal imaging may be contribut-
gle agent, although synergy may exist for certain
ing to the rising incidence of this disease [38].
combinations that warrant further study [63]. Responses
Currently, approximately 25% of patients with kidney
to chemotherapy in renal cell carcinoma must be evalu-
cancer present with disseminated disease while an addi-
ated in light of the low but finite level of background
tional one third to one half of patients without dissemina-
noise created by spontaneous regression of metastases.
tion at initial diagnosis develop metastases [22, 38]. Thus,
The lack of an active cytotoxic agent for this disease pro-
more than half of all patients with kidney cancer will
vides further rationale for developing and optimizing bio-
present with or develop disease for which systemic anti-
logical and targeted therapy for kidney cancer.
cancer therapy is needed. Five year survivals of over 90%
have recently been reported for patients with small, inci-
dentally detected kidney cancers [100]. However, relapse Kidney Cancer is Responsive
rates of 50% or higher in patients with larger or more
to the Immune System
extensive tumors provide a strong rationale for the devel-
opment of a systemic adjuvant post-surgical therapy for Several clinical observations similar to those described
patients with resectable kidney cancer. Since traditional for malignant melanoma indicate a role for the immune
forms of cytotoxic chemotherapy remain ineffective for system to influence the natural history and treatment of

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 645
© Springer Science + Business Media B.V. 2009
646 Biological therapy of genitourinary cancer

kidney cancer. First, patients with established metastatic authors reported a 7% response rate to medroxyproges-
kidney cancer may have long disease or treatment free terone in the subset of patients who remained on treatment
intervals, irrespective of the treatment received, sug- for at least 6 months [57]. Taken together, the results of
gesting a degree of innate immune control. In a large, these studies demonstrate the potential role of the innate
retrospective, single institution analysis, 5-year survival immune system in mediating the spontaneous regression
was observed in 4.5% of 670 patients with advanced of established metastases from renal cancer.
kidney cancer treated on one of several cytokine, che- Based on these clinical observations, renal cell carci-
motherapy, or hormonal therapy trials from 1975–1996, noma, like melanoma, appeared to constitute an ideal
including 9 of 294 patients (3%) treated with chemo- human tumor system for biotherapy. Early attempts to
therapy or hormonal therapy agents subsequently dem- harness the immune system in the treatment of kidney
onstrated to lack activity in this disease [61]. Similarly, cancer are of historical interest and employed a variety
patients may relapse many years, even decades after a of active specific immunization techniques. Tykka, in
nephrectomy with curative intent for renal cell carci- 1974, reported a 17% complete response rate of pulmo-
noma, indicating a potential role for immune surveillance nary metastatic lesions in patients receiving a monthly
to control micrometastatic kidney cancer effectively for vaccine composed of homogenized tumor plus either
many years [99]. Second, immunosuppression increases tuberculin or Candida every 4 weeks [101]. Subsequently,
the risk of the development of kidney cancer, as seen in three separate trials using either tumor homogenate plus
the increased risk of the development of renal cell carci- tuberculin, enzymatically digested tumor tissue plus
noma in kidney allograft recipients [46]. Third, immune Corynebacterium parvum, or whole tumor cells plus
stimulation with cytokine therapy can induce tumor attached dimethyldioctadecyl ammonium bromide
regression and prolong survival in patients with meta- (DDA) as vaccine have all reported responses in 15–25%
static kidney cancer and will be described in detail in of patients [54, 66, 70]. Furthermore, immune RNA has
this chapter. Fourth, spontaneous regression of metastatic been extracted from lymphocytes of guinea pigs specifi-
kidney cancer has been carefully documented [102]. cally immunized with renal tumor cells and coincubated
Indeed, the response rates for observation, placebo, and with autologous lymphocytes; these reinfused lympho-
“placebo-equivalent” therapy in phase II and III trials of cytes induced a response in three of six patients [96].
metastatic renal cell carcinoma have been as high as 7%. Thymosin fraction V, a partially purified extract of calf
The most notable example of a clinical study showing thymus glands, has induced objective responses in
a high rate of apparently spontaneous regression of meta- approximately 15% of advanced renal cancer patients.
static kidney cancer was a phase II trial was performed on These early studies provided the impetus for developing
73 patients with metastatic renal cell carcinoma who were biological therapies for renal cell carcinoma.
identified prospectively and treated with observation until Important laboratory observations provide evidence
evidence of progression. Five patients (7%) achieved a of an immunologic response to RCC and have been
partial or complete remission on observation and 12% reviewed elsewhere in this book. Several lines of evi-
were progression free for at least 1 year [68]. Recently dence indicate that cytotoxic T-lymphocytes (CTLs)
reported phase III trials in metastatic renal cell cancer play a central role in mediating the regression of RCC.
also demonstrated a spontaneous remission rate of greater First, lymphocytic infiltrates within renal tumors are
than 5%. A randomized trial comparing interferon gamma comprised primarily of CD3+ T-lymphocytes, and
to placebo in 197 patients with advanced renal cell carci- molecular analysis of these lymphocytic infiltrates has
noma showed a 7% response rate in the placebo group demonstrated clonally expanded populations of T-cell
that was higher than the response rate achieved in the receptor alpha and beta restricted T-cells that are gener-
treatment group [35]. Another recent phase III trial has ally not found in peripheral blood, normal kidney or
demonstrated modest rates of regression with what is lymph nodes [47, 72]. Second, tumor-specific cytotoxic
likely to be “placebo-equivalent” treatment in compari- T-cell lines from tumor infiltrating lymphocytes have
son to interferon alpha. Medroxyprogesterone acetate has been developed from patients with RCC that are capable
been evaluated for single agent activity in renal cell car- of recognizing and lysing autologous tumor cells [4].
cinoma in a number of trials, is generally considered to be Third, specific T-cell defined tumor associated antigens
inactive, and may be thought of as a “placebo-equivalent” have been identified from patients with RCC, including
in this setting [29]. A trial comparing interferon-alpha to HER-2/neu, PRAME, and RAGE [65]. Taken together,
medroxyprogesterone demonstrated the superiority of these correlative laboratory observations demonstrate
interferon alpha, but a 3% overall objective response rate the central role of the CTL in mediating the immune
was observed in the medroxyprogesterone arm, and the response to RCC.
Robert K. Oldham 647

Cytokine Therapy months or less for whom cytokine therapy may be less
beneficial [60, 109]. Another important variable in the
The treatment of MRCC with the cytokines interferon- outcome of patients with metastatic kidney cancer treated
alpha and interleukin-2 results in objective responses in with cytokine therapy is the presence of the primary kid-
about 15% of patients. A recent retrospective analysis of a ney tumor [26, 27, 58].
large, single institution patient database provides an exam- It is also becoming increasingly evident that not all his-
ple of the benefits that derive from treatment of MRCC tological types of kidney cancer are equally capable of
[60]. This review of 670 patients with advanced RCC ana- response to immune modulation. It is important to recog-
lyzed the outcomes of patients treated with cytokine nize that conventional (clear cell) renal cell carcinoma and
therapy versus chemotherapy or hormonal therapy in clin- its histological variants (conventional renal cell carcinomas
ical trials from 1976–1996. About 60% of patients were may show a mixture of cytoplasmic features, including
treated with cytokine therapy with interferon-alpha, inter- variants that have formerly been classified as “granular
leukin-2, or interferon-alpha plus interleukin-2 versus and cell” and “sarcomatoid” renal cell carcinomas) represent a
about 40% were treated with chemotherapy or hormonal specific clinico-pathological disease entity characterized
therapy. A highly significant and clinically meaningful by typical findings on light microscopy and loss of the von
survival advantage was demonstrated for cytokine therapy Hippel-Lindau tumor suppressor gene. This disease entity,
versus chemotherapy or hormonal therapy, with median now classified as conventional renal cell carcinoma, com-
survival times of 13 versus 6 months (p < 0.001, 95% prises approximately 60% of all epithelial tumors arising
confidence intervals of 12–15 months versus 5–8 months). from the kidney [78]. It is clear that the other histological
The survival advantage for cytokine therapy extended to types of tumors of renal epithelial origin (e.g. papillary,
all prognostic groups, with median survivals of 27 versus chromophobe, oncocytoma, and collecting duct tumors)
15 months for the favorable subset of patients, although represent distinct clinico-pathological diseases with dif-
patients with the poorest prognosis survived less than 6 ferent clinical behavior, but it is not clear whether other
months even with treatment. This important analysis dem- histological types of kidney cancer are capable of respond-
onstrated that in each risk group, there was an approxi- ing to immune manipulation [69].
mate doubling of the median overall survival when Recognition of known prognostic indicators as deter-
patients were treated with cytokine therapy compared minants of eligibility and stratification is a critical com-
with chemotherapy or hormonal therapy. ponent in the design of clinical trials and the evaluation
Trials of cytokine therapy in metastatic renal cell carci- of their results. Furthermore, because of the major con-
noma, have demonstrated wide variations in response tribution of prognostic factors on the outcome of patients
rate, including some trials that failed to demonstrate with metastatic kidney cancer (MRCC) irrespective of
responses to high dose interleukin-2 [1] while others have treatment, randomized trials are required to demonstrate
demonstrated responses as high as 33% [86]. From these the relative merits of a particular therapy. The overall
observations, several conclusions can be drawn. First, response rates observed with cytokine therapy for kid-
advanced kidney cancer has a variable natural history ney cancer have been relatively low, and the natural his-
determined at least in part by the innate immune response tory of patients with MRCC varies widely independent
to renal cell carcinoma. Second, pre-treatment patient and of treatment. As a result, the impact of treatment with
tumor characteristics have a large impact on the outcome these cytokines on survival has been controversial and
of a given treatment or observation strategy. Third, a mea- can only be directly assessed in phase III clinical trials.
sure of caution must be exercised when evaluating the Until recently, phase III trial data have been lacking, but
results of clinical trials in renal cell carcinoma, particu- several important phase III studies have recently been
larly when the results indicate marginal activity of a par- completed and reported that shed light on the impact of
ticular treatment. Several recent reports have identified cytokine therapy on the survival of patients with MRCC,
some of the important prognostic variables that help to the role of “cytoreductive” nephrectomy prior to cytokine
predict clinical outcomes in kidney cancer, including therapy in with metastases at initial presentation, and the
TNM stage, tumor grade, performance status, and certain incremental benefits that may be achieved with intensive
indicators of disease burden (lactate dehydrogenase level, therapy with high dose interleukin-2 [32, 45, 92].
presence of anemia, and presence of hypercalcemia). It is
possible to identify a favorable subset of patients with
advanced kidney cancer whose median survival exceeds Interferon-alpha
2 years with cytokine therapy and an unfavorable subset Interferon-alpha has produced objective responses in
of patients with a median survival of approximately 6 10–20% of patients with MRCC. An earlier review of
648 Biological therapy of genitourinary cancer

the data from multiple phase II studies showed a response evidence of disease progression, resulting in 6 responses
rate of 12% in 1,042 patients treated with a variety of (response rate, 15%; 95% confidence interval, 5.7–30%).
doses and schedules of interferon, and concluded that The ability of interferon-alpha to prolong survival has
longer survival could not be demonstrated in patients now been evaluated in several randomized trials in com-
with MRCC treated with interferon-alpha [9, 105]. The parison to a non-cytokine containing control arm [32, 48,
mean time to response may be as long as 3–4 months. It 57, 73, 97]. The results are presented in Table 1. The two
can be argued that an optimal dose and schedule for smaller studies showed no survival benefit for treatment
alpha-interferon has never been defined for renal cell with interferon-alpha, but included few patients and
carcinoma. The anti-tumor activity of interferon can be allowed crossover in one case. The two larger studies
attributed to its ability to modulate cellular immunity, to both demonstrated a statistically significant improve-
its direct (non-immunologically mediated) anti-prolifer- ment in survival for the interferon-containing arm at
ative or pro-apoptotic activity, or to an effect on tumor doses of 10–18 MU three times weekly. Because the
angiogenesis [52] It is not clear that all of these proper- addition of vinblastine has been shown not to improve
ties of interferon can be evoked optimally and simulta- survival over interferon alone, the improvement in sur-
neously from a single dose and schedule of interferon in vival of interferon plus vinblastine over vinblastine alone
vivo. Some data appear to demonstrate a dose–response can be attributed to the interferon. Taken together, the
relationship for interferon, suggesting that the clinical results of the four randomized, prospective trials com-
responses induced by interferon are likely to be due to paring cytokine therapy versus non-cytokine therapy in
its known anti-proliferative activity rather than to immu- several hundred patients, demonstrated an objective
nomodulation. However, evidence for a dose–response response rate of 16% versus 2% and average median sur-
relationship for interferon has not been consistently vival of approximately 12 versus 8 months. These results
observed among investigators. While some toxicities also closely resemble the results of the large, retrospec-
from interferon do appear to be dose-related, not all tive single-institution analysis of cytokine versus non-
antitumor responses appear to be dose-related. From his cytokine therapy described above in which median
extensive review of the literature relating to the treat- survivals were 13 versus 6 months [32].
ment of renal cell carcinoma with interferon, Muss [64] A randomized trial involving interferon alpha was
concluded that toxicity from interferon was dose-related conducted through the Southwestern Oncology Group
and that the highest therapeutic index for interferon and was designed to answer the important question of
occurred at doses ranging from 5–10 million units/m2. whether nephrectomy in the setting of known metastatic
However, it is interesting to note that some studies have disease is of clinical benefit [27]. Almost 350 patients
demonstrated responses using very low doses of inter- with metastatic kidney cancer were randomly assigned
feron. For example, a single-arm phase II trial conducted to receive interferon alpha immediately upon enroll-
by the Southwest Oncology Group [63] accrued 40 fully ment into the trial or following a nephrectomy.
evaluable, previously untreated patients with advanced Interestingly, the objective response rate to interferon
renal cell carcinoma treated with alpha-interferon 1 million alpha was unexpectedly low and not different between
units subcutaneously daily continually until there was the two arms of the study (less than 4%), yet the median

Table 1. Randomized trials comparing interferon alpha with non-cytokine treatment


Treatment N Response (%) Median survival (months) P value Publication
IFN 30 6 7 NA Steineck 1990(97)
MPA 30 3 7
IFN + VBL 44 20 16 0.19 Kriegmair 1995(48)
MPA 45 0 10
IFN + VBL 79 16 17 0.0049 Pyrhonen 1999(73)
VBL 81 2 10
IFN 167 16 8.5 0.011 MRC 1999(57)
MPA 168 2 6
Total
IFN +/− 320 16 11.5
VBL or MPA 324 2 7.7

IFN interferon-alpha, MPA medroxyprogesterone acetate, VBL vinblastine


Robert K. Oldham 649

survival was significantly improved (by 3 months) in 27% overall response rate is similar to that achieved
the patients who underwent nephrectomy before immu- with interleukin-2 and lymphokine-activated killer
notherapy, independent of other prognostic factors. (LAK) cell therapy and to higher doses of single agent
More rigorous assessment of objective response was interleukin-2, the contribution of beta-interferon is
proposed to explain the low response rate observed in uncertain.
this trial; dose and schedule were comparable to studies Early experience with gamma-interferon suggested
reporting much higher responses. A similar improve- little if any activity [75, 80]. Gamma-interferon is a
ment in survival for nephrectomy in the setting of meta- product of activated T cells and is unique in its ability to
static disease was also observed in a somewhat smaller stimulate macrophages. Although approved for chronic
study of similar design, and responses to interferon were granulomatous disease, it has demonstrated some activ-
in the usual range (12–19%) [58]. A similar European ity in metastatic renal cancer and in one study using a
trial demonstrated an improvement in the time to dis- low-dose weekly subcutaneous injection of 100 mg, a
ease progression as well as the median survival for 30% response rate was observed in patients with meta-
patients who had a nephrectomy compared to those who static renal cancer [5]. A second study reported a 15%
did not prior to initiating interferon-alpha therapy. In response rate [19]. Combinations of alpha and gamma-
addition, there are some studies that indicate resection interferon have shown additive toxicities without added
of solitary or a small number of metastasis may lead to therapeutic benefit [74]. An effort to improve response
long-term survival in approximately 30% of patients by combining alpha-interferon and gamma-interferon
with metastatic renal cancer. Thus, surgery plays an failed to achieve this intent, perhaps attributable to addi-
important role in MRCC, especially in conjunction with tive toxicities and inability to suitably escalate dosages
effective systemic therapy [22]. [28]. Some of the same principles regarding dose and
schedule of alpha-interferon, discussed above, pertain
to gamma-interferon. A dose–response relationship for
PEG-interferon gamma-interferon may not be straightforward [6]. Foon
The treatment of kidney cancer with inferon alpha ther- et al. conducted a phase I trial of gamma-interferon in
apy requires daily or thrice weekly subcutaneous admin- which anti-tumor responses were observed. However,
istration. The preparation of the interferon with the responses occurred at the lowest doses tested [28].
polyethylene glycol (PEG) sustains the absorption of Follow-up trials with both beta and gamma interferon
interferon from a subcutaneous injection, slows its clear- have yielded equivocal results and only interferon-alpha
ance, and improves its pharmacokinetic parameters. This is approved for use in MRCC.
improvement in pharmacokinetics allows for less fre-
quent administration, for example once weekly, and may Interleukin-2
also improve the effectiveness and side effect profile by In the United States, high-dose interleukin-2 was
avoiding the frequent peaks and troughs associated with approved by the Food and Drug Administration for the
the shorter half-life of conventional interferon. PEG- treatment of advanced kidney cancer, whereas in Europe,
interferon alpha has become commercially available for lower doses and infusional IL-2 were approved and are
the treatment of hepatitis C, and studies were done to more commonly used in MRCC. Kidney cancer cells in
evaluate the effectiveness and tolerability of this agent in culture can grow in spite of the presence of high concen-
renal cell carcinoma. Preliminary studies suggest that trations of interleukin-2, demonstrating that interleukin-2
PEG-interferon retains anti-tumor activity in renal cell has no directly cytotoxic activity. The action of interleu-
carcinoma and has a spectrum of side effects similar to kin-2 appears to be the activation of lymphocytes and
conventional interferon alpha, but improves the conve- natural killer cells that mediate the antitumor activity of
nience of administration to a once weekly dose [62]. this agent. About 10–20% of patients treated with inter-
leukin-2 will achieve an objective response, and a small,
but important fraction of these patients will achieve a
Other Interferons complete remission, which may be quite durable. A
The role of beta-interferon in the treatment of renal-cell review of over 1,900 patients treated with single agent
carcinoma remains to be established. A trial of combina- interleukin-2 demonstrated an overall response rate of
tion interleukin-2 and beta-interferon evoked objective 15% that included complete remissions in 4% of the total
responses in 6 of 22 evaluable patients, with one com- patient population (Table 2) [7, 8]. This review included
plete remission and five partial remissions. Two of these patients treated on phase I and II studies using various
responses lasted for almost 2 years [32]. Although this dose levels, schedules, and routes of administration.
650 Biological therapy of genitourinary cancer

Table 2. Results of therapy with interleukin-2 The administration of interleukin-2 at high doses
Route of Administration N % % complete requires hospital admission to an intensive care unit or a
responding responders dedicated, specialized nursing unit capable of close
Intravenous bolus 733 16 5 monitoring and the administration of vasopressors to
Continuous intravenous 922 13.3 2.5 approximately half of patients, a skilled treatment team,
infusion and in spite of the careful attention of this skilled team
Subcutaneous 290 15.0 3 is nonetheless associated with significant toxicity and
Total 1945 15.0 3.6
occasional mortality. A 4% treatment related death rate
was noted in the original 255 patient dataset, although
Table 2 adapted from(7,8).
more recent series have found the treatment related
death rate to be 1% or less [34, 44, 107]. Because of the
It is likely that the dose intensity of interleukin-2 admin- toxicity associated with the capillary leak syndrome and
istration influences the proportion of patients that the potential cardiac toxicity associated with high dose
respond to treatment as well as the quality and durability interleukin-2, patient selection is very important for its
of the responses, but a linear dose-response relationship safe administration [44]. The toxicity of high dose inter-
has not been demonstrated for interleukin-2, and ques- leukin-2 has limited its application to patients that are
tions of dose and schedule have been the subject of relatively young, have limited co-morbidities, an excel-
several important clinical studies in the late 1990s. lent performance status, and access to one of the rela-
Interleukin-2 was approved by the United States Food tively few treatment centers that offer this intensive
and Drug Administration in 1992 based on a data set of therapy. Because lower intensity and outpatient regi-
over 450 patients treated on a series of phase II studies mens of interleukin-2 lead to responses in a fairly simi-
with high dose, intravenous bolus interleukin-2 with lar proportion of patients in phase II studies, it has been
and without lymphokine-activated killer (LAK) cells, tempting to replace high dose interleukin-2 with low
demonstrating an overall response rate of 21% with 6% dose, intravenous or subcutaneous, outpatient regime ns
complete and 15% partial responses [18, 25, 53]). of interleukin-2. While considerably easier to adminis-
Subsequently, studies have demonstrated that the bene- ter and applicable to a much broader patient population,
fits of high dose interleukin-2 plus LAK cells are derived it is not clear that the response rate and quality of
from the interleukin-2 (as discussed below and reviewed responses are preserved with this approach.
in [48]), and of the over 450 patients analyzed, 255 were In order to directly compare the results of therapy with
treated with high dose interleukin-2 without LAK cells. high dose interleukin-2 compared to lower intensity and
The results of treatment in these 255 patients have been less toxic regimens, two large, prospective, randomized
rigorously scrutinized and followed long-term, updated trials have completed patient accrual at the National
reports have been published periodically [24, 25, 30, Cancer Institute (NCI) and at multiple institutions through
31]. These reports are instructive in an analysis of the the Cytokine Working Group (CWG) comparing high
outcome of treatment of metastatic kidney cancer with dose interleukin-2 with lower dose regimens [55, 107].
interleukin-2. Objective responses following high dose In the former study from the NCI, patients were random-
interleukin-2 have been confirmed in 15% (7% com- ized to high dose intravenous bolus interleukin-2, low
plete and 8% partial), with a median response duration dose intravenous bolus interleukin-2, or subcutaneous
for the entire group of 15 months. Considering this interleukin-2. In the latter study through the CWG,
group as a whole, the results of treatment with high dose patients were randomized to receive high dose intrave-
interleukin-2 appear to be fairly similar to the results nous bolus interleukin-2 or subcutaneous interleukin-2
presented above for treatment with interferon alpha in plus interferon alpha. The response rates with high-dose
which the overall response rate of 16% and median sur- interleukin-2 are approximately twice that observed with
vivals of 7–16 months have been observed in rigorously lower doses of interleukin-2 with or without interferon
conducted trials. However, the importance of aggressive alpha in groups of patients that have been comparably
immunotherapy with high dose interleukin-2 treatment screened and selected on the basis of their ability to
may be the duration and quality of the responses undergo the rigors of treatment with high dose interleu-
observed. In this 255 patient data set treated with high kin-2 [55, 107]. The survival and the response duration
dose interleukin-2, the median survival for responding from these trials await further maturation of the data;
patients is over 84 months, and the median response however, the trend from both studies appears to indicate
duration for completely responding patients has not that the durability of the responses in patients treated
been reached, which is tantamount to cure [24]. with high dose interleukin-2 is superior to that achieved
Robert K. Oldham 651

with low dose interleukin-2 [55, 107]. Overall survival was patients irrespective of the route of interleukin-2 adminis-
not significantly different in the two groups. [55, 107]. tration [8]. Because of this apparent slight but potentially
In summary, the consistent results of the numerous significant improvement in the response rate, several ran-
studies of the treatment of metastatic kidney cancer with domized trials have been performed comparing combina-
cytokine therapy are that a relatively small and consis- tion versus single agent cytokine therapy. The largest of
tent fraction of patients, 10–20%, will achieve evidence these, reported by Negrier et al., comprised over 400
of tumor regression with occasional complete remissions patients and demonstrated an improved response rate for
and long survival noted for a few patients. For the subset interleukin-2 plus interferon alpha (18.6%) compared to
of patients who are able to receive aggressive immuno- interleukin-2 (6.5%) or interferon alpha (7.5%) alone
therapy with high dose interleukin-2, this intensive therapy [65]. Unfortunately, the improvement in the response rate
may result in a higher percentage of complete and partial did not lead to a significant improvement in median sur-
remissions and more durable responses(Table 3). Even vival, although the crossover design for patients failing to
enthusiasts of high dose interleukin-2 hoped to replace respond to interleukin-2 or interferon alpha monotherapy
this labor intensive and toxic treatment with a more makes comparison of the survival times difficult. It is
widely applicable but therapeutically equivalent regimen important to note that the overall response rate to cytokine
(in designing the randomized trial described above, the monotherapy in this study was lower than what has gen-
Cytokine Working Group had expected equivalent thera- erally been observed, again underscoring the importance
peutic results from the low dose regimen and performed of patient selection and the importance of randomized trials
this trial “before accepting the low dose regimens as the for comparing the outcomes of different treatments. An
standard”). However, at the time of this writing, high important observation resulted from the crossover design
dose interleukin-2 cannot be dismissed and remains one of this study: responses to interleukin-2 after interferon
of the standard treatment approaches for appropriately failure or interferon alpha after interleukin-2 failure are
selected patients treated at centers with expertise in the rare [20]. Thus, at present, monotherapy with either inter-
administration of this therapy. leukin-2 or interferon alpha at modest doses remains the
standard of cytokine care for most patients with kidney
cancer and can be expected to result in tumor regression in
Cytokine Combinations 10–20% and overall median survivals of slightly greater
Pre-clinical studies of interleukin-2 and interferon alpha than a year [32].
suggested additive or synergistic activity and led to a A cytokine therapy must be reinterpreted in light of the
number of phase I and II studies to investigate the clinical newer molecularly targeted agents described in a subse-
activity of this combination. A review of over 1,400 quent section. Perspective randomized trials of cytokine
patients has been published demonstrating response rates therapy versus targeted agents, and/or combinations
of approximately 20% and complete remissions in 5% of thereof, are currently underway [32].

Table 3. Randomized trials comparing high dose versus low dose interleukin-2
Study/treatment N Complete responses (%) Partial responses (%) Objective response rate (%)
a
NCI Study(107)
HD-IL-2 115 9 (8) 13 (11) 22 (19)
LD-IL-2 (IV) 112 5 (4) 6 (5) 11 (10)
LD-IL-2 (SC) 53 3 (6) 3 (6) 6 (11)
CWG Study(55)
HD-IL-2 99 8 (8) 17 (17) 25 (25)
LD-IL-2 (SC) + IFN (SC) 94 2 (2) 10 (11) 12 (13)
Totals
HD-IL-2 214 17 (8) 30 (14) 47 (22)
LD-IL-2 (IV or SC) +/− IFN 259 10 (4) 19 (7) 29 (11)

HD-IL-2 high dose interleukin-2 by intravenous bolus injection; LD-IL-2 low dose interleukin-2 by intravenous bolus injection (IV) or
subcutaneous injection (SC); IFN interferon alpha by subcutaneous injection
a
The NCI study was initially designed to evaluate high dose versus low dose intravenous bolus injections of interleukin-2. After the first
117 patients were randomized (56–57 per arm), the third arm of the study was added to compare subcutaneous interleukin-2 admin-
istration. For a statistically valid comparison, these data have been reported comparing only concurrently randomized patients [104].
For informational comparison in this table, the data have been presented as if all patients had been randomized concurrently.
652 Biological therapy of genitourinary cancer

Adoptive Cellular Therapy been done. Perhaps the most extensive dose-related studies
have been accomplished by Oldham and co-workers
Lymphocyte Activated Killer Cells and Tumor [51] in utilizing as many as ten doses of activated T cells
Infiltrating Lymphocytes in treatment of patients with advanced malignancy. With
Historically and for the purposes of review, cancer immu- regard to patients with renal cancer, only four doses have
notherapy has been classified as “active,” including agents been used, giving total cell doses up to 2 × 1011 cells.
such as cytokines that activate the host anti-tumor immune A series of phase I–II trials were performed, including
response, and “passive,” including agents such as mono- 114 patients with renal cell carcinoma, treated with TIL
clonal antibodies or immune effector cells that directly plus IL-2, and demonstrating an objective response rate
(or sometimes indirectly) mediate the anti-tumor response of 23% (range 0–35%) [40]. Again, in a randomized trial
themselves. The transfer of immune effector cells with comparing interleukin-2 therapy alone to the combina-
direct anti-tumor reactivity has also been termed “adop- tion of interleukin-2 plus TIL in 178 patients with renal
tive cellular therapy.” In kidney cancer, the use of lym- cell carcinoma, there was no advantage in terms of
phokine-activated killer (LAK) cells is of great historical response rate, duration of response, or survival to the
significance, since the development of modern cytokine addition of the TIL compared to interleukin-2 alone [23].
therapy using interleukin-2 for renal cell carcinoma was The addition of an expanded population of autologous
originally based on the observation, in the early 1980s, tumor killing cells in the form of TIL or LAK cells to
that LAK cells are lymphoid cells that circulate in the interleukin-2 has been carefully studied and in its present
peripheral blood, are activated in vitro by exposure to form offers no advantage to the treatment of kidney can-
high concentrations of interleukin-2, and have the ability cer over cytokine therapy alone. Nonetheless, early
to selectively lyse neoplastic cells [44]. Early clinical trials observations demonstrated that melanoma patients
of interleukin-2 were therefore done in combination with treated with TIL plus interleukin-2 were capable of
LAK cells, or more accurately stated, early trials of LAK responding even when a prior response to interleukin-2
cells required the systemic administration of interleukin-2 alone was not observed [88]. The administration of an
to demonstrate efficacy of the LAK cells [36, 85, 98, expanded population of highly selected T cells derived
104]. An initial response rate in excess of 75% was from autologous TILs in combination with high dose
reported [85], but a series of phase II studies including IL-2 and following a non-myeloablative chemotherapy
over 500 patients with renal cell carcinoma demonstrated conditioning regimen mediated the regression of meta-
that this treatment approach lead to anti-tumor responses static melanoma and MRC patients refractory to stan-
in 22% of patients (range 9–35%) [40]. Moreover, most dard therapy, including high dose IL-2. This study
of these responses have been partial and transient, typi- demonstrated that prior lymphodepletion improves the
cally lasting no more than several months. Subsequently, persistence and function of adoptively transferred cells
randomized trials at the National Cancer Institute [86] [17]. Ongoing studies at the National Cancer Institute
and elsewhere [40]) comparing interleukin-2 plus or and in Europe indicate that TIL cells can be genetically
minus LAK cells failed to demonstrate a contribution to engineered to enhance their activity against both mela-
response rate or survival by the LAK cells. noma and kidney cancer. Such genetically retargeted
In the hope of reducing the number of cells needed for cells sometimes, accompanied by lympho-depletion
passive transfer and decreasing the requirement for the strategies, have shown high response rates in a small
co-administration of high dose interleukin-2, tumor infil- series of patients over the last 5 years [17, 50].
trating lymphocytes (TIL) were discovered in the search
for a more potent lymphoid effector cell [108]. Whereas
LAK cells are nonspecific natural killer cells, TIL are Allogeneic Stem Cell Transplantion
activated cytotoxic T cells, which show a greater propen- A novel approach to the biological treatment of meta-
sity for target-specific killing. TIL are isolated from a static renal cell carcinoma has employed allogeneic
primary or metastatic tumor, and, similar to LAK cells, hematopoietic stem cell transplantation in an attempt to
are expanded ex vivo in the presence of interleukin-2, mediate anti-tumor activity from donor T-cells from an
then reinfused in combination with systemic cytokine HLA matched sibling donor. This treatment strategy rep-
therapy. In murine models, TIL were found to be 50–100 resents a form of adoptive cellular immunotherapy.
times more potent on a per cell basis in the treatment of Based on data that the transfer of allogeneic stem cells in
established lung micrometastases than LAK cells [87]. the treatment of hematalogic malignancies can induce a
Sufficient studies utilizing acceptable doses of interleukin-2 graft-versus-leukemia reaction that may improve the
along with escalating doses of activated cells have not cure of these malignancies, a similar graft-versus-tumor
Robert K. Oldham 653

effect has been sought in the treatment of metastatic kid- tumor antigen presentation and patient T-lymphocyte
ney cancer. Childs et al. established proof-of-principle activation by enhancing tumor antigenic peptide presen-
for the existence of the GVT effect in metastatic kidney tation in the context of major histocompatibility complex
cancer [10, 11] and have recently reviewed their experi- (MHC) molecules, by restoring co-stimulatory signals
ence at the National Institutes of Health using this treat- that are frequently deficient in tumor cells, and by
ment modality [12]. Over 50 patients have been treated, amplifying recruitment of the patient’s immune effector
and 47% have achieved evidence of tumor regression, cells. Gene therapy strategies for kidney cancer are based
including 18 (38%) partial responses and 4 (9%) com- on the premise that genetic modification of immune
plete responses [12]. Three of the four complete respond- cells can enhance function or that the introduction of a
ers remain disease free, including the first patient treated particular gene into tumor cells can increase their immu-
whose remission exceeds 5 years. Interestingly, the nogenicity and induce an effective immune response not
majority of patients that eventually respond to this treat- only against the gene-modified tumor, but also against
ment experience initial tumor progression in the first few the unmodified tumor cells that are present elsewhere in
months following the procedure, and tumor regression the individual [39, 67]. Tumor cells may be modified in
occurs late in the course of the transplant at a median of vitro or in vivo to express cytokines, co-stimulatory
5 months. Disease regression typically occurred after the molecules, or foreign MHC molecules.
withdrawal of immunosuppression and after the conver-
sion of the host to full donor T-cell chimerism, implying Vaccine therapy
that these responses resulted from the GVT activity of Dendritic cells (DC) are the most potent, professional
donor T-cells. These results demonstrate the proof-of- antigen presenting cells (APC) of the immune system and
principle that a clinically meaningful graft-versus-tumor may be uniquely able to stimulate T-cells because they
response can be achieved in the setting of metastatic possess all of the required co-stimulatory molecules.
renal cell carcinoma. The wide applicability of this Dendritic cells can be isolated from peripheral blood
approach may be limited by the long interval of time mononuclear cells and expanded and modified ex vivo,
required from the initiation of the evaluation of the making vaccination with DC feasible. Strategies to induce
patient for treatment with stem cell therapy until the the expression of tumor antigens on dendritic cells include
withdrawal of immunosuppression when a graft-versus- gene transfer into DC using viral or non-viral vectors,
tumor effect may be expected. This time interval has so tumor cell lysates, apoptotic bodies, tumor-derived RNA,
far required many months, during which time patients and DC-tumor cell hybrids. Alternately, dendritic cells
with even a moderate rate of tumor progression may may be expanded in vivo by the systemic administration
worsen clinically and become ineligible for aggressive of combinations of cytokines such as GM-CSF and IL-4
intervention or succumb to their disease, particularly in [90]. These approaches indicate there is a need to reach a
light of the fact that most patients will have been treated balance between immunity and tolerance [16].
with a traditional treatment approach before consider- A study in patients with metastatic renal cell carci-
ation of this novel form of immunotherapy. Indeed, as noma demonstrated the potential effectiveness of DC
expected, of the first 19 patients reported by Childs et al., vaccines using electrofusion to generate autologous
eight patients died of disease progression within approx- tumor cell-allogeneic DC hybrids [49]. Seventeen
imately 1 year while awaiting the GVT effect during patients were vaccinated subcutaneously, and after vac-
follow-up after transplant [10]. As a result, treatment of cination, four patients achieved complete responses and
patients with stem cell transplantation requires careful two patients had partial responses. Fever and injection
selection of a subset of kidney cancer patients who are fit site pain were the only important toxicities reported.
enough to undergo an aggressive treatment and who Controversy exists regarding the effectiveness and repro-
have a slowly progressive form of the disease. The role ducibility of the electrofusion technique used in this
of stem cell therapy for the treatment of metastatic renal study, but other human clinical trials employing dendritic
cell carcinoma will require further study with large num- cell vaccines in patients with metastatic renal cell cancer
bers of patients and confirmation of these intriguing have also shown anti-tumor responses [33, 41].
results by other investigators at other sites [84]. The field of cancer vaccination has been advanced by
evidence that both cytotoxic and helper T cells recog-
nize intracellularly degraded peptides that are processed
Vaccines and Gene Therapy by specialized antigen presenting cells via the protea-
Vaccines and gene therapy approaches to the treatment some apparatus, inserted into the endoplasmic reticu-
of kidney cancer have generally attempted to improve lum, and transported to the cell surface for association
654 Biological therapy of genitourinary cancer

with major histocompatibility (MHC) molecules [103]. used natural killer cells transfected with the interleu-
Taking advantage of this finding, a specific approach to kin-2 gene for the adoptive immunotherapy of renal cell
improving effective antigen presentation for the treat- carcinoma and other malignancies [91]. This trial dem-
ment of renal cell carcinoma involves the use of peptide onstrated the feasibility of this approach, the relative
vaccines derived from heat shock protein–peptide com- lack of toxicity, a significant increases in other immuno-
plexes from kidney tumors. Heat shock proteins (HSP) stimulatory cytokines (interferon gamma, transforming
are molecular chaperones implicated in “loading” growth factor beta, and granulocyte-macrophage colony
immunogenic peptides onto MHC molecules for pre- stimulating factor), and clinical response in one patient
sentation by antigen presenting cells to T-cells. In this with lymphoma. There are several examples of the latter
vaccination strategy, autologous tumor derived peptides gene therapy approach of genetically modifying tumor
associated with HSP administered to patients are taken cells to increase their immunogenicity in the treatment
up by antigen presenting cells such as DC and processed of kidney cancer. Autologous renal cell carcinoma cells
by the APC so that tumor antigen is “re-presented” to have been retrovirally transduced with the gene for
naïve T-cells, leading to an antigen specific T-cell granulocyte-macrophage colony stimulating factor and
response [94]. Amato et al. have treated patients with re-administered as a vaccine to 16 patients with meta-
metastatic renal cell carcinoma with autologous gp96 static renal cell carcinoma [93]. No significant toxicities
heat shock protein–peptide complex vaccination in a were observed, and one patient experienced a partial
phase I–II trial [3]. The vaccine, at one of three doses, remission. Another strategy to make tumor cells more
was administered once with a week for 4 weeks with immunogenic has involved the transfer of the gene encod-
follow-up doses at weeks 12 and 20 depending on ing human leukocyte antigen (HLA) molecule B7 into
response. Responses were observed in 4 of 16 patients autologous renal cell carcinoma cells. In one study, lipid-
(25%) at the 25 ug dose level, including one patient with a DNA complexes (commercially called “Allovectin-7”)
complete response; three additional patients experienced were transferred into accessible metastatic lesions in 15
prolonged stabilization of disease in excess of 1 year. HLA B7 negative renal cell carcinoma patients [83].
Follow on phase III randomized studies have not con- Conceptually, the intent of these intratumoral injection
firmed these earlier trials, and there has not been a sig- studies is to develop an in vivo autologous tumor vac-
nificant difference seen between the HSP arm and the cine that will lead to a systemic disease response. In this
placebo arm in these adjuvant studies [95]. example, neither toxicity nor responses were observed.
A similar example of this approach includes the direct
Gene Therapy transfer of the lipid complexed interleukin-2 gene into
Human gene therapy generally involves the insertion of accessible metastatic lesions in patients with renal cell
functional DNA into a human cell either in vivo or ex carcinoma. Further details of gene therapy are presented
vivo to replace a defective gene or to provide a new in Chapter 19.
function to the cell. Traditionally, the therapeutic DNA
has been packaged in a viral or lipid vector for delivery
of a functional gene into human cells. Alternately, naked
Targeted therapy
DNA, generally in the form of a plasmid, may be directly It has been known for many years that renal cancer over-
injected into skin or muscle or adsorbed onto micropar- expresses vascular endothelial growth factor (VEGF).
ticles and forcibly bombarded into the skin in vivo or Because of this finding, recent efforts have targeted
other cells ex vivo. Naked DNA injections have been VEGF in an attempt to block vascular supply and blood
alternately termed “polynucleotide vaccines.” Human supply to MRCC.
gene therapy approaches to the treatment of kidney can- Bevacizumab (Avastin) is a recombinant human
cer that have advanced into clinical trials have included monoclonal antibody against VEGF [32, 71]. In a phase
altering immune effector cells with a gene designed to II trial or more than 100 patients with MRCC, random-
improve their function or altering tumor cells with a ized placebo versus a low-dose (3 mg/kg) or high-dose
gene designed to improve their immunogenicity [33]. (10 mg/kg) of bevacizumab given intravenously in treat-
The “polynucleotide vaccine” strategy of transferring a ment-resistant patients every 2 weeks (over 90% had
gene encoding a tumor antigen into endogenous antigen received prior IL-2) demonstrated an improved time to
presenting cells has yet to be exploited in the clinical progression for the bevacizumab group [106].
model of renal cell carcinoma, although this approach As a result of this interesting phase II trial, there are
has been attempted clinically in other solid tumors [89]. ongoing studies with bevacizumab plus other agents
In an example of the former approach, investigators active in MRCC [81]. Bevacizumab plus interferon is
Robert K. Oldham 655

more active than interferon alone [106], and bevaci- MRCC. The daunting task of working through this will
zumab combined with erlotinib (Tarceva), an EGRF require many randomized trials for patients with MRCC
inhibitor, has shown objective responses in patients with [14]. While the best combination is not known, the
resistant, advanced renal cancer [37]. future looks much brighter for these patients than it did
Many clinical trials are underway using various com- a mere 5 years ago.
binations of cytokines and targeted therapies, but no
specific combination has proven superior to an alternative
combination to-date.
In addition to a large molecule inhibitor of VEGF,
References
such as the monoclonal antibody bevacizumab, there 1. Abrams JS, Rayner AA, Wiernik PH. High-dose recombinant
interleukin-2 alone: a regimen with limited activity in the treat-
are small molecule inhibitors, which work through the
ment of advanced renal cell carcinoma. J Natl Cancer Inst 1990;
super family of tyrosine kinases. Three of the agents are 82: 1202.
currently approved for the treatment of MRCC and are 2. Amato RJ. Chemotherapy for renal cell carcinoma. Semin Oncol
being used alone and in various combinations with the 2000; 27:177–186.
other active agents. 3. Amato RJ, Murray L, and Wood L. Active specific immunotherapy
in patients with renal cell carcinoma (RCC) using autologous
Sorafenib (Nexavar) is an oral agent that inhibits Raf
tumor derived heat shock protein-peptide complex-96 (HSPP-96)
kinase. Sorafenib also is a direct inhibitor of the VEGF vaccine. Proc Am Soc Clin Oncol 1999; Abstract 19.
receptor family. In a phase II randomized study with over 4. Angevin E, Kremer F, Gaudin C. Analysis of T-cell immune
200 patients with MRCC, patients received 400 mg twice response in renal cell carcinoma: polarization to type I-like differ-
a day of sorafenib versus placebo. The median progres- entiation pattern, clonal T-cell expansion, and tumor-specific cyto-
toxicity. Int J Cancer 1997; 72:431–440.
sion free for survival was longer in the sorafenib group.. 5. Aulitzky W, Gastl G, Aulitzky WE et al. Successful treatment of
This study was followed by a phase III randomized trial metastatic renal cell carcinoma with a biologically active dose of
or sorafenib versus placebo in cytokine-refractory MRCC recombinant interferon-gamma. J Clin Oncol 1989; 7:1875–1884.
[21]. Although only a small percentage of patients had a 6. Brown TD, Koeller J, Beougher K et al. A phase I clinical trial of
complete or partial response, and many of the patients recombinant DNA gamma interferon. J Clin Oncol 1987;
5:790–798.
exhibit stable disease while receiving the drug. The 7. Bukowski RM. Cytokine therapy for metastatic renal cell carci-
median progression free survival for the sorafenib group noma. Semin Urol Oncol 2001; 19:148–154.
was 24 weeks, double that of placebo. Sorafenib was well 8. Bukowski RM. Natural history and therapy of metastatic renal cell
tolerated, but does have significant side effects which are carcinoma: the role of interleukin-2. Cancer 1997; 80:1198–1220.
9. Cameron DA, Cornbleet MC, Mackie RM et al. Adjuvant inter-
generally manageable, including dermatological, gastro-
feron alpha 2b in high risk melanoma – the Scottish study. Br J
intestinal and neurologic symptoms, along with fatigue Cancer 2001; 84:1146–1149.
and elevates blood pressures [76]. 10. Childs R, Chernoff A, Contentin N et al. Regression of metastatic
Another oral tyrosine kinase inhibitor of VEGF recep- renal-cell carcinoma after nonmyeloablative allogeneic peripheral-
tor is sunitinib (Sutent). This drug was effective in phase blood stem-cell transplantation. N Engl J Med 2000; 343:750–758.
11. Childs RW, Clave E, Tisdale J, Plante M, Hensel N, Barrett J.
II trials and was subsequently approved based on random-
Successful treatment of metastatic renal cell carcinoma with a non-
ized phase III trial in patients with MRCC [59]. Again, the myeloablative allogeneic peripheral-blood progenitor-cell trans-
median progression free for survival was approximately plant: evidence for a graft-versus-tumor effect. J Clin Oncol 1999;
double in the treated group versus the placebo group. 17:2044–2049.
A third small molecule, temsirolimus (Torisel), has 12. Childs R, Srinivasan R. Advances in allogeneic stem cell trans-
plantation: directing graft- versus-leukemia at solid tumors. Cancer
recently been approved for poor risk MRCC patients. This J 2002; 8:2–11.
drug is given intravenously and is an inhibitor or rapamy- 13. Choueiri TK, Dreicer R, Rini BI, et al. Phase II study of lenalido-
cin (mTOR), a molecule important in tumor-promoting mide in patients with metastatic renal cell carcinoma (MRCC)
intracellular signaling pathways. Studies comparing this [abstract]. J Clin Oncol 2006; 24(Suppl):4539. Abstract 4539.
agent to interferon-alpha demonstrated an improvement in 14. Colcher D, Esteban J, Carrasquillo JA et al. Complementation of
intracavitary and intravenous administration of a monoclonal anti-
survival amounting to 3–6 months [42] body (B72.3) in patients with carcinoma. Cancer Res 1987;
Other agents, such as lenalidomide [13], tykerb lapa- 47:4218–4224.
tinib [77] and axitinib [82], are underway. 15. Colcher D, Zalutsky M, Kaplan W, Kufe D, Austin F, Schlom J.
While it is clear that these small molecule and large Radiolocalization of human mammary tumors in athymic mice by
a monoclonal antibody. Cancer Res 1983; 43:736–742.
molecule agents targeting various receptors and mole-
16. Cools N, Ponsaerts P, Van Tendeloo VFI, et al. Balancing between
cules that support neovascularization and/or promote immunity and tolerance: an interplay between dedritic cells, regu-
tumor growth are important individually, it is unknown latory T cells, and effector T cells. J Leuko Biol 2007;
how best to combine them with other agents active in 82:1365–1374.
656 Biological therapy of genitourinary cancer

17. Dudley ME, Wunderlich JR, Yang, JC et al. Adoptive cell transfer 36. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-
therapy following non-ablative but lymphodepleting chemother- activated killer cell phenomenon. Lysis of natural killer- resistant
apy for the treatment of patients with refractor metastatic mela- fresh solid tumor cells by interleukin 2-activated autologous
noma. J Clin Oncol 2005; 23:2346–2357. human peripheral blood lymphocytes. J Exp Med 1982;
18. Dutcher JP, Atkins M, Fisher R et al. Interleukin-2-based therapy 155:1823–1841.
for metastatic renal cell cancer: the Cytokine Working Group expe- 37. Hainsworth JD, Sosman DR, Spigel RC, et al. Phase II trial of
rience, 1989–1997. Cancer J Sci Am 1997; 3(Suppl 1):S73–S78. bevacizumab and erlotinib in patients with metastatic renal carci-
19. Ellerhorst J., Jones E., and Kilbourn R. Fixed low dose gamma noma. J Clin Oncol 2005; 31:7889–7896.
interferon is active against metastatic renal cell carcinoma. Proc 38. Hock LM, Lynch J, Balaji KC. Increasing incidence of all stages of
Am Soc Clin Oncol 1992; 11:220 (abstract). kidney cancer in the last 2 decades in the United States: an analysis
20. Escudier B, Chevreau C, Lasset C et al. Cytokines in metastatic of surveillance, epidemiology, and end results program data. J
renal cell carcinoma: is it useful to switch to interleukin-2 or inter- Urol 2002; 167:57–60.
feron after failure of a first treatment? Groupe Francais 39. Hoffman DM, Figlin RA. Intratumoral interleukin 2 for renal-cell
d’Immunotherape. J Clin Oncol 1999; 17:2039–2043. carcinoma by direct gene transfer of a plasmid DNA/DMRIE/
21. Escudier B, Szczylik C, Eisen T, et al. Randomized phase III trial DOPE lipid complex. World J Urol 2000; 18:152–156.
of sorafenib (BAY 43–9006) – an oral multi-kinase inhibitor – in 40. Hoffman DM, Gitlitz BJ, Belldegrun A, Figlin RA. Adoptive cel-
patients with advanced RCC [abstract]. Proc Am Soc Oncol 2005; lular therapy. Semin Oncol 2000; 27:221–233.
23. Abstract 4510. 41. Holtl L, Rieser C, Papesh C et al. Cellular and humoral immune
22. Figlin RA. Renal cell carcinoma: management of advanced dis- responses in patients with metastatic renal cell carcinoma after
ease. J Urol 1999; 161:381–387. vaccination with antigen pulsed dendritic cells. J Urol 1999;
23. Figlin RA, Thompson JA, Bukowski RM et al. Multicenter, ran- 161:777–782.
domized, phase III trial of CD8(+) tumor-infiltrating lymphocytes 42. Hudes G, Carducci M, Tomczak P, et al. A phase 3, randomized,
in combination with recombinant interleukin-2 in metastatic renal 3-arm study of temsirolimus (TEMSR) or interferon-alpha (IFN)
cell carcinoma. J Clin Oncol 1999; 17:2521–2529. or the combination of TEMSR + IFN in the treatment of first-line,
24. Fisher RI, Rosenberg SA, Fyfe G. Long-term survival update for poor-risk patients with advanced renal cell carcinoma (adv RCC)
high-dose recombinant interleukin-2 in patients with renal cell car- [abstract]. J Clin Oncol 2006; 24(Suppl):LBA4. Abstract LBA4.
cinoma. Cancer J Sci Am 2000; 6(Suppl 1):S55–S57. 43. Jemal A, Siegel R, Ward E, et al. Cancer statistics 2007. CA Canc
25. Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Fyfe G. High- J Clin 2007; 57:43–66.
dose aldesleukin in renal cell carcinoma: long-term survival 44. Kammula US, White DE, Rosenberg SA. Trends in the safety of
update. Cancer J Sci Am 1997; 3(Suppl 1):S70–S72. high dose bolus interleukin-2 administration in patients with meta-
26. Flanigan RC, Mickisch G, Sylvester R, et al. Cytoreductive neph- static cancer. Cancer 1998; 83:797–805.
rectomy in patients with metastatic renal cancer: a combined anal- 45. Kavolius JP, Mastorakos DP, Pavlovich C, et al. Resection of met-
ysis. J Urol 2004; 171:1071–1076. astatic renal cell carcinoma. J Clin Oncol 1998; 16:2261–2266.
27. Flanigan RC, Salmon SE, Blumenstein BA et al. Nephrectomy fol- 46. Kliem V, Kolditz M, Behrend M. Risk of renal cell carcinoma after
lowed by interferon alfa-2b compared with interferon alfa-2b kidney transplantation. Clin Transplant 1997; 11:255–258.
alone for metastatic renal-cell cancer. N Engl J Med 2001; 47. Kowalczyk D, Skorupski W, Kwias Z. Flow cytometric analysis of
345:1655–1659. tumor-infiltrating lymphocytes in patients with renal cell carci-
28. Foon K, Doroshow J, Bonnem E et al. A prospective randomized noma. Br J Urol 1997; 80:543–547.
trial of alpha 2B-interferon/gamma-interferon or the combination 48. Kriegmair M, Oberneder R, Hofstetter A. Interferon alfa and vin-
in advanced metastatic renal cell carcinoma. J Biol Response Mod blastine versus medroxyprogesterone acetate in the treatment of
1988; 7:540–545. metastatic renal cell carcinoma. Urology 1995; 45:758–762.
29. Fossa SD. Interferon in metastatic renal cell carcinoma. Semin 49. Kugler A, Stuhler G, Walden P et al. Regression of human meta-
Oncol 2000; 27:187–193. static renal cell carcinoma after vaccination with tumor cell-den-
30. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie dritic cell hybrids. Nat Med 2000; 6:332–336.
AC. Results of treatment of 255 patients with metastatic renal cell 50. Lamers CJH, Sleijfer S, Vulto AG, et al. Treatment of metastatic
carcinoma who received high-dose recombinant interleukin-2 renal cell carcinoma with autologous T-lymphocytesgenetically
therapy. J Clin Oncol 1995; 13:688–696. retargeted against carbonic anhydrase IX: first clinical experience.
31. Fyfe GA, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie J Clin Oncol May 1, 2006; 24(13):20e-22.
AC. Long-term response data for 255 patients with metastatic 51. Lewko WM, Good RW, Bowman D, Smith TL, Oldham RK.
renal cell carcinoma treated with high-dose recombinant interleu- Growth of tumor derived activated T-cells for the treatment of can-
kin-2 therapy. J Clin Oncol 1996; 14:2410–2411. cer. Cancer Biother 1994; 9:211–224.
32. Garcia JA, Rini BI. Recent progress in the management of 52. Lindner DJ. Curr Oncol Rep 2002 Nov; 4(6):510–4).
advanced renal call carcinoma. CA Cancer J Clin 2007; 53. Margolin KA. Interleukin-2 in the treatment of renal cancer. Semin
57:112–125. Oncol 2000; 27:194–203.
33. Gitlitz BJ, Belldegrun AS, Figlin RA. Vaccine and gene therapy of 54. McCune CS, Schapira DV, Henshaw EC. Specific immunotherapy
renal cell carcinoma. Semin Urol Oncol 2001; 19:141–147. of advanced renal carcinoma: evidence for the polyclonality of
34. Gitlitz BJ, Hoffman DM, Moldawer N, Belldegrun A, Figlin RA. metastases. Cancer 1981; 47:1984–1987.
Treatment of metastatic renal cell carcinoma with high-dose bolus 55. McDermott DF, Regan M, Clark JI, et al. Randomized phase III
interleukin-2 in a non-intensive care unit: an analysis of 124 con- trial of high-dose interleukin-2 versus subcutaneous interleukin-2
secutively treated patients. Cancer J 2001; 7:112–120. and interferon in patients with metastatic renal cell carcinoma. J
35. Gleave ME, Elhilali M, Fradet Y. Inteferon gamma-1b compared Clin Oncol 2005; 23:133–141.***
to placebo in metastatic renal cell carcinoma. N Engl J Med 1998; 56. McLaughlin JK, Lipworth L. Epidemiologic aspects of renal cell
338:1265. cancer. Semin Oncol 2000; 27:115–123.
Robert K. Oldham 657

57. Medical Research Council Renal Cancer Collaborators. Interferon- 75. Quesada JR, Kurzrock R, Sherwin SA, Gutterman JU. Phase II
alpha and survival in metastatic renal cell carcinoma: early results studies of recombinant human interferon gamma in metastatic
of a randomised controlled trial. Lancet 1999; 353:14–17. renal cell carcinoma. J Biol Response Mod 1987; 6:20–27.
58. Mickisch GHJ, Garin A, van Poppel H, de Prijck L, Sylvester R. 76. Ratain MG, Eisen T, Stadler WM, et al. Phase II placebo-controlled
Radical nephrectomy plus interferon-alfa-based immunotherapy randomized discontinuation of trial of sorafenib in patients with
compared with interferon alone in metastatic renal-cell carcinoma: metastatic renal cell carcinoma. J Clin Oncol 2006; 24:2505–2512.
a randomised trial. Lancet 2001; 358:966–970. 77. Ravaud A, Gardner J, Hawkins R, et al. Efficacy of lapatinib in
59. Motzer RJ, Hutson TE, Tomczak P, et al. Phase III randomized trial patients with high tumor EGFR expression: results of a phase III
of sunitinib malate (SU11248) versus interferon-alpha (IFN-α) or trial in advanced renal cell carcinoma (RCC) [abstract]. J Clin
first-line systemic therapy for patients with metastatic renal cell ONcol 2006; 24(Suppl):4502. Abstract 4502.
carcinoma (mRCC) [abstract]. J Clin Oncol 2006; 24(Suppl):LBA3. 78. Reuter VE, Presti JC. Contemporary approach to the classification
Abstract LBA3. of renal epithelial tumors. Semin Oncol 2000; 27:124–137.
60. Motzer RJ, Mazumdar M, Bacik J. Survival and prognostic strati- 79. Ries LAG, Harkins D, Krapcho M, et al. eds. SEER Cancer
fication of 670 patients with advanced renal cell carcinoma. J Clin Statistics Review, 1975–2003, Bethesda, MD: National Cancer
Oncol 1999; 17:2530–2540. Institute; 2006.
61. Motzer RJ, Mazumdar M, Bacik J, Russo P, Berg WJ, Metz EM. 80. Rinehart JJ, Malspeis L, Young D, Neidhart JA. Phase I/II trial of
Effect of cytokine therapy on survival for patients with advanced human recombinant interferon gamma and renal cell carcinoma. J
renal cell carcinoma. J Clin Oncol 2000; 18:1928–1935. Biol Resp Modif 1986; 5:300–308.
62. Motzer RJ, Rakhit A, Ginsberg M et al. Phase I trial of 40-kd 81. Rini BI, Halabi S, Taylor J, et al. Cancer and Leukemia Group B
branched pegylated interferon alfa-2a for patients with advanced 90206: a randomized phase III trial of interferon-alpha or inter-
renal cell carcinoma. J Clin Oncol 2001; 19:1312–1319. feron-alpha plus anti-vascular endothelial growth factor antibody
63. Motzer RJ, Russo P. Systemic therapy for renal cell carcinoma. (bevacizumab) in metastatic renal cell carcinoma. Clin Cancer Res
J Urol 2000; 163:408–417. 2004; 10:2584–2586.
64. Muss HB. Interferon therapy for renal cell carcinoma. Semin 82. Rini BI, Rixe O, Bukowski RM, et al. AG-013736, a multi-target
Oncol 1987; 14:36–42. tyrosine kinase receptor inhibitor, demonstrates anti-tumor activ-
65. Negrier S, Escudier B, Lasset C et al. Recombinant human inter- ity in a phase 2 study of cytokine-refractory metastatic renal cell
leukin-2, recombinant human interferon alfa-2a, or both in meta- cancer (RCC) [abstract]. J Clin Oncol 2005; 23(Suppl):4509.
static renal-cell carcinoma. Groupe Francais d’Immunotherapie. N Abstract 4509.
Engl J Med 1998; 338:1272–1278. 83. Rini BI, Selk LM, Vogelzang NJ. Phase I study of direct intrale-
66. Neidhart JA, Murphy SG, Hennick LA, Wise HA. Active specific sional gene transfer of HLA-B7 into metastatic renal carcinoma
immunotherapy of stage IV renal carcinoma with aggregated lesions. Clin Cancer Res 1999; 5:2766–2772.
tumor antigen adjuvant. Cancer 1980; 46:1128–1134. 84. Rini BI, Zimmerman TM, Gajewski TF, Stadler WM, Vogelzang
67. Neumann E, Engelsberg A, Decker J. Heterogenous expression of NJ. Allogeneic peripheral blood stem cell transplantation for meta-
the tumor-associated antigens RAGE-1, PRAME, and glycoprotein static renal cell carcinoma. J Urol 2001; 165:1208–1209.
75 in human renal cell carcinoma: candidates for T-cell based 85. Rosenberg SA, Lotze MT, Muul LM et al. Observations on the
immunotherapies? Cancer Res 1998; 58:4090–4095. systemic administration of autologous lymphokine- activated
68. Oliver RT, Nethersell AB, Bottomley JM. Unexplained spontaneous killer cells and recombinant interleukin-2 to patients with meta-
regression and alpha-interferon as treatment for metastatic renal static cancer. N Engl J Med 1985; 313:1485–1492.
carcinoma. Br J Urol 1989; 63:128. 86. Rosenberg SA, Lotze MT, Yang JC. Prospective randomized trial
69. Phillips JL, Pavlovich CP, Walther M, Ried T, Linehan WM. The of high-dose interleukin-2 alone or in conjunction with lym-
genetic basis of renal epithelial tumors: advances in research and phokine-activated killer cells for the treatment of patients with
its impact on prognosis and therapy. Curr Opinion Urol 2001; advanced cancer. J Natl Cancer Inst 1993; 85:622.
11:463–469. 87. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the
70. Prager MD, Baechtel FS, and Peters PC. Specific immunotherapy adoptive immunotherapy of cancer with tumor- infiltrating lym-
of human metastatic renal cell carcinoma. Proc Am Assoc Cancer phocytes. Science 1986; 233:1318–1321.
Res 1981; 22:163 (abstract). 88. Rosenberg SA, Yannelli JR, Yang JC et al. Treatment of patients with
71. Presta LG, Chen H, O’Connor SJ, et al. Humanization of an anti- metastatic melanoma with autologous tumor- infiltrating lympho-
vascular endothelial growth factor monoclonal antibody for the cytes and interleukin 2. J Natl Cancer Inst 1994; 86:1159–1166.
therapy of solid tumors and other disorders. Cancer Res 1997; 89. Rousseau RF, Hirschmann-Jax C, Takahashi S, Brenner MK. Cancer
57:4593–4599. vaccines. Hematol Oncol Clin North Am 2001; 15:741–773.
72. Puisieux I, Bain C, Merrouche Y. Restriction of the T-cell reper- 90. Roth MD, Gitlitz BJ, Kiertscher SM et al. Granulocyte macrophage
toire in tumor-infiltrating lymphocytes from nine patients with colony-stimulating factor and interleukin 4 enhance the number
renal-cell carcinoma; relevance of the CDR3 length analysis for and antigen-presenting activity of circulating CD14+ and CD83+
the identification of in situ clonal T-cell expansions. Int J Cancer cells in cancer patients. Cancer Res 2000; 60:1934–1941.
1996; 66:201–208. 91. Schmidt-Wolf IG, Finke S, Trojaneck B et al. Phase I clinical study
73. Pyrhonen S, Salminen E, Ruutu M et al. Prospective randomized applying autologous immunological effector cells transfected with
trial of interferon alfa-2a plus vinblastine versus vinblastine alone the interleukin-2 gene in patients with metastatic renal cancer, col-
in patients with advanced renal cell cancer. J Clin Oncol 1999; orectal cancer and lymphoma. Br J Cancer 1999; 81:1009–1016.
17:2859–2867. 92. Schrodter S, Hakenberg OW, Manseck A, et al. Outcome of surgical
74. Quesada JR, Evans L, Saks SR, Gutterman J. Recombinant treatment of isolated local recurrence after radical nephrectomy
interferon alpha and gamma in combination as treatment for for renal cell carcinoma. J Urol 2002; 167:1630–1633.
metastatic renal cell carcinoma. J Biol Resp Modif 1988; 93. Simons JW, Jaffee EM, Weber CE et al. Bioactivity of autologous
7:234–239. irradiated renal cell carcinoma vaccines generated by ex vivo
658 Biological therapy of genitourinary cancer

granulocyte-macrophage colony-stimulating factor gene transfer. 101. Tykka H, Hjelt L, Oravisto KJ, Turunen M, Tallberg T.
Cancer Res 1997; 57:1537–1546. Disappearance of lung metastases during immunotherapy in five
94. Srivastava P. Interaction of Heat Shock Proteins with Peptides patients suffering from renal carcinoma. Scand J Respir Dis
and Antigen Presenting Cells: chaperoning of the Innate and Suppl 1974; 89:123–134.
Adaptive Immune Responses. Annu Rev Immunol 2002; 102. Vogelzang NJ, Priest ER, Borden L. Spontaneous regression of
20:395–425. histologically proved pulmonary metastases from renal cell car-
95. Srivastava P, Kumar S, Mendonca C. Principles and practice of cinoma: a case with 5-year follow-up. J Urol 1992; 148:1247.
the use of heat-shock protein-peptide complexes for immuno- 103. Weber J. Peptide vaccines for cancer. Cancer Invest 2002;
therapy of human cancer. Principles and Practice of Biologic 20:208–221.
Therapy of Cancer Updates 2001; 2:1–13. 104. West WH, Tauer KW, Yannelli JR et al. Constant-infusion recom-
96. Steele G, Jr., Wang BS, Richie JP, Ervin T, Yankee R, Mannick binant interleukin-2 in adoptive immunotherapy of advanced
JA. Results of xenogeneic I-RNA therapy in patients with meta- cancer. N Engl J Med 1987; 316:898–905.
static renal cell carcinoma. Cancer 1981; 47:1286–1288. 105. Wirth MP. Immunotherapy for metastatic renal cell carcinoma.
97. Steineck G, Strander H, Carbin BE et al. Recombinant leukocyte Urol Clin North Am 1993; 20:283–295.
interferon alpha-2a and medroxyprogesterone in advanced renal 106. Yang JC, Haworth L, Sherry RM, et al. A randomized trial of
cell carcinoma. A randomized trial. Acta Oncol 1990; bevacizumab, an anti-vascular endothelial growth factor anti-
29:155–162. body, for metastatic renal cancer. N Engl J Med 2003:
98. Sznol M, Clark JW, Smith JW et al. Pilot study of interleukin-2 349:427–434.
and lymphokine-activated killer cells combined with immuno- 107. Yang JC, Sherry RM, Steinberg SM, et al. Randomized study of
modulatory doses of chemotherapy and sequenced with inter- high-dose and low-dose interleukin-2 in patients with metastatic
feron alfa-2a in patients with metastatic melanoma and renal cell renal cell cancer. J Clin Oncol 2003; 16:3127–3132.
carcinoma. J Natl Cancer Inst 1992; 84:929–937. 108. Yron I, Wood TA, Jr., Spiess PJ, Rosenberg SA. In vitro growth
99. Tapper H, Klein H, Rubenstein W, Intriere L, Choi Y, Kazam E. of murine T cells. V. The isolation and growth of lymphoid cells
Recurrent renal cell carcinoma after 45 years. Clin Imaging 1997; infiltrating syngeneic solid tumors. J Immunol 1980;
21:273–275. 125:238–245.
100. Tsui K, Shvarts O, Smith RB, Figlin R, deKernion JB, Belldegrun 109. Zisman A, Pantuck AJ, Dorey F et al. Improved prognostication
A. Renal cell carcinoma: prognostic significance of incidentally of renal cell carcinoma using an integrated staging system. J Clin
detected tumors. J Urol 2000; 163:426–430. Oncol 2001; 19:1649–1657.
21.4 Biological therapy of colon cancer
ROBERT O. DILLMAN

Colon Cancer demonstrated in the smaller subset of patients with stage


II disease [60]. On the basis of this trial, 5FU plus
Non-specific Immune Stimulants levamisole became standard therapy for the adjuvant
treatment of stage III colon cancer during 1990–1997.
Levamisole A major limitation of the early levamisole trial was
Levamisole is a synthetic phenylimidazothiazole oral that it did not have a control arm of 5FU plus folinic
antihelminth that exhibited non-specific immunostimu- acid (leucovorin), which was shown to be more active
latory properties in animal models [82]. Interest in this than 5FU alone while the trial was being conducted.
agent as a cancer therapy dates back to 1971. Small Two large U.S. trials subsequently showed that levami-
exploratory clinical studies were inconclusive, but con- sole did not add benefit to such a regimen. During
sidered encouraging enough to lead to randomized 1989–1990 2,151 patients who had stage II or III colon
phase III trials in colon cancer. Two randomized trials cancer were randomized to receive weekly 5FU plus
failed to demonstrate a benefit for levamisole alone leucovorin, 5FU plus levamisole, or 5FU plus leuco-
compared to placebo in the adjuvant setting. In one vorin plus levamisole [98]. After more than 7 years of
small double-blind U.S. trial 78 patients were random- follow up, the 5-year survival rates were 74% for 5FU
ized to receive 18 months of levamisole 2.5 mg/kg/day plus leucovorin, 73% for all three drugs, and 70% for
given on days 1 and 2 of each week, or placebo follow- 5FU plus levamisole. During 1988–1992 3,794 patients
ing resection of Dukes B or C colorectal cancer [8]. No were randomized in a four-arm trial to receive 5FU plus
benefit for levamisole was observed. In a double-blind levamisole for 1 year or 6 months of low-dose leuco-
randomized European trial comparing levamisole at 100 vorin plus 5FU (“Mayo Clinic regimen”), or high-dose
to 250 mg twice a week to placebo as adjuvant therapy leucovorin plus 5FU (“Roswell Park regimen”) or the
for 297 patients with node-positive colon cancer, there Mayo clinic regimen plus levamisole, plus FU (LDLV
again was no demonstrable benefit for levamisole [1]. plus LEV) regimen [33]. After 10 years of follow up
Other trials combined levamisole with 5-fluorouracil there were no differences in survival among the arms,
(5FU). In a large adjuvant trial 1,296 patients, who had and the 5FU plus leucovorin regimens replaced the
resected colon cancer that was either locally invasive levamisole regimens as standard therapy when the
(n = 318) or metastatic to regional lymph nodes (n = 929), results were first reported in 1997.
were randomized to observation, or to 5FU plus levami- Other randomized trials of adjuvant therapy in colon
sole for 1 year, or to levamisole alone [58]. Patients cancer confirmed beyond a doubt that the addition of
were assigned to observation, or to levamisole alone levamisole provided no benefit in the adjuvant therapy
(50 mg p.o. three times a day for 3 days, repeated every of colon cancer. In a trial in which 379 patients received
2 weeks for 1 year), or to this regimen of levamisole 5FU plus folinic acid, and 374 received 5FU plus
plus 5-FU (450 mg/m2 i.v. daily for 5 days and then, levamisole, after a median follow up of 6.8 years, there
beginning at 28 days, weekly for 48 weeks). Once again was a 72% survival rate in both arms [63]. In a four-arm
levamisole alone provided no benefit compared to trial in which 598 patients were randomized to receive 1
observation, but after a median follow up of 3 years, the year of weekly 5FU, or 5-FU plus levamisole, or 5-FU
combination of 5-FU plus levamisole was associated plus IFN-α or all three agents, recurrence rates and sur-
with a significantly reduced risk of recurrence and death vival were worse in the combined arms that contained
for patients who had lymph node metastases (p = 0.006). levamisole (p = 0.003) [77]. In another randomized
After a median follow up of 6.5 years the advantages of trial there was no difference in survival for 92 patients
levamisole + 5FU persisted in stage III disease, with a who received 5FU plus levamisole compared to 92 who
40% reduced recurrence rate (p < 0.0001) and 33% received 5FU alone, but leucopenia and hepatic toxicity
reduced death rate (p = 0.0007) [59]. No benefit was were more frequent in the levamisole arm [5]. In a two-arm

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 659
© Springer Science + Business Media B.V. 2009
660 Biological therapy of colon cancer

trial in which 1,703 colon cancer patients were random- but no responses in previously treated patients [89–91].
ized to 5FU plus levamisole or 5FU plus leucovorin Unfortunately, other investigators typically reported much
plus levamisole, there was no difference in survival, but lower response rates for this combination in colorectal
the three drug regimen was more toxic [13]. cancer, with a range of 20% to 50% [12, 49, 68, 92, 93].
Randomized trials were unable to confirm response
rates any higher than 20% to 25%, which is no different
Thymosin Fraction 5 that 5FU alone. In a randomized trial for patients with
The importance of the thymus gland and thymic proteins previously untreated advanced colon cancer, the response
for normal T-lymphoycte cellular immunity has been rate was 21% for 104 patients who received 5FU alone
recognized for many decades. One of the preparations of (750 mg/m2 as a 4-h i.v. infusion on days 1–5 and then
interest was a crude lysate called thymosin fraction 5. As i.v. bolus weekly) compared to 25% for 101 patients who
summarized in Table 1, subsequent dose-exploring and received 5FU plus IFN (3 to 9 MIU thrice weekly s.c.)
phase II trials failed to confirm significant activity in [67]. In a similar trial that utilized the same dose and
lung cancer or immune enhancing activity for thymosin schedule of 5FU, but IFN at a dose of 10 MIU s.c. thrice
fraction 5 [14, 15]. weekly, the response rate was 30% for 54 patients who
received 5FU alone and only 19% for 52 patients
who received 5FU plus IFN [39]. The lower response rate
Interferons was probably due to patients withdrawing from the trial
As shown in Table 1, single agent alpha-interferon (IFN- because of toxicity. Patients who received IFN experienced
α) rarely produces objective tumor responses in patients more toxicity, including four toxic deaths, leucopenia,
with metastatic colorectal cancer [4, 62]. In three phase II lymphopenia, depression, and alopecia.
trials there were no objective responses observed with either Subsequent trials combined 5FU, folinic acid, and
lymphoblastoid interferon or alpha interferon [6, 9, 54]. IFN-α, but the response rates were no higher than the
However, there is laboratory evidence that IFN-α can doublets [32, 44, 69]. Randomized trials confirmed that
enhance the antitumor activity of 5-FU by a mechanism IFN-α adds toxicity, but not benefit, to 5FU and folinic
which is different from that of leucovorin [61, 89]. acid. In a trial that included 204 untreated patients, there
An early clinical trial which combined 5-FU with IFN-α was no difference in response rate was (23% to 24%) or
for the treatment of advanced colorectal carcinoma survival (medians 11 to 12 months) [10]. The addition
claimed a 75% objective response rate among patients of IFN was associated with more toxicity, especially
who had not been treated previously with chemotherapy, fever, diarrhea, and mucositis. In another trial 102

Table 1. Single-agent activity of various biologicals in patients with metastatic colon cancer
Modality class Biological agent Reference Patients Response rate (%)
Non specific Thymosin fraction 5 Dillman 12 0
Cytokine Lymphoblastoid Interferon [6] 19 0
Cytokine Interferon-α [54] 18 0
Cytokine Interferon-α [9] 36 0
Cytokine Interleukin-2 (IL-2) [73, 74] 22 0
Cell therapy IL-2 + LAK [73, 74] 38 18
Cell therapy IL-2 + LAK [17] 35 0
Cell therapy IL-2 + TIL [64] 8 0
Cell therapy IL-2 + TIL [20] 8 0
Cell therapy CD4+ T cells [19] 15 7
Mab Anti-CEA [16] 30 0
Mab Edrocolomab 40 0
Mab Edrocolomab [24] 20 5
Mab Edrocolomab [52] 25 4
Mab Edrocolomab [57] 52 2
Mab Bevacizumab [28] 234 3

Mab = monoclonal antibody


LAK = lymphokine activated killer cells
TIL = tumor infiltrating lymphocytes
CEA = carcinoembryonic antigen
Robert O. Dillman 661

patients were randomized to receive 5FU and leuco- has been in combination with adoptive cell therapy or in
vorin with or without IFN; there was no difference in combination with other biologicals. High-dose bolus IL-2
response rates (8% and 10%) but time to progression yielded no objective responses in 22 patients [73, 74].
and overall survival both favored the group that did not The response rate for high-dose continuous infusion
received IFN (p = 0.002) [50]. Thus, clinical trials IL-2 and tumor necrosis factor (TNF-α) was 0/16 [18].
showed that 5FU plus IFN-α was no better than 5FU The response rate for high-dose continuous infusion
plus leucovorin, but more toxic, and that there was no IL-2 and IFN-α was 1/10 [65].
advantage for the addition of IFN-α to 5FU plus leuco-
vorin in the treatment of metastatic colorectal cancer.
Several randomized trials have established that there
Adoptive Cell Therapy
is no role for IFN-α in the adjuvant treatment of high- There has been only limited exploration of cell-based
risk colon cancer. In a Greek trial that included stage II therapies for colon cancer. As described in detail else-
and III colon cancer 139 patients received 5FU plus foli- where in this book, lymphokine activated killer [LAK]
nic acid and 141 received the same therapy plus IFN (3 cells are derived by taking peripheral blood lymphocytes
MU s.c. thrice weekly) [23]. After a median follow up of (PBL) and stimulating them in vitro with IL-2 while
4 years, there was no difference in disease free survival tumor infiltrating lymphocytes (TIL) are produced by
or overall survival. In an Italian trial in which 322 incubating tumor samples with IL-2. For patients with
patients with stage II or III colon cancer were random- metstatic colon cancer, LAK administered with high dose
ized to infusional 5FU plus leucovorin with or without bolus IL-2 produced responses in 7/38 (18%) patients
IFN (5 MU s.c. thrice weekly), there was no difference in [73, 74] AK cells infused with high-dose continuous
either disease-free survival or overall survival but toxic- infusion IL-2 (civ IL-2) produced no responses in 35
ity was greater in the IFN-arm because of flu-like syn- patients [18]. There were no responses among 16 patients
drome [26]. In a three-arm German trial, 813 patients treated with TIL, eight of whom received high doses of
with stage II and 750 with stage III colon cancer were TIL plus civ IL-2, and eight who received variable doses
randomized to 5FU plus levamisole alone or with leuco- of TIL with other schedules of IL-2 [20, 64].
vorin, or with IFN [81]. The 4-year survival rate was Autolymphocyte therapy and autologous activated lym-
highest in the arm that included leucovorin (78%), but phocytes were used to describe autologous lymphocyte
was no better in the IFN-arm than 5FU plus levamisole preparations that were derived from peripheral blood and
alone (both 66%). In a four-arm trial in which 598 stimulated with anti-CD3 monoclonal antibodies in vitro to
patients were randomized to receive 1 year of weekly produce an auto-cytokine preparation that was then used
5FU, or 5-FU plus levamisole, or 5-FU plus IFN-α or all for incubation with additional peripheral blood lympho-
three agents, there was increased toxicity in the IFN- cytes [19, 66]. The autolymphokine preparation contained
arms but no effect on survival [77]. The National Surgical significant amounts of TNF-α, IL-1β, interferon-γ, and
Adjuvant Breast and Bowel Project (NSABP) group ran- IL-6, but no IL-2. Culturing peripheral blood lymphocytes
domized 2,176 patients with stage II or III colon cancer in this media resulted in enrichment of CD3+ cells, but
to receive 5FU plus leucovorin with or without IFN [97]. decreased CD3+/CD56+ cells (NK), decreased CD4+/
After 4.5 years of follow up there was no difference CD45RA(2H4) suppressor T cells, and increased CD8+/
in disease free survival (69–70%) or overall survival HLA-Dr+, and CD4+/CD29(4B4)/HLA-DR+ helper T cells.
(80–81%), but there was more toxicity associated with IFN. There was a slight decrease in NK cytolytic activity in Cr51
In one small randomized trial 99 patients with stage assays compared to the mononuclear cells obtained prior to
II, III, or IV colon cancer were randomized to receive incubation in ALK. These cells, which lacked cytolytic
IFN-gamma 0.2 mg s.c. daily for 6 months or observa- activity, were infused in doses of about 109 cells in patients
tion after surgical resection of their disease [96]. Despite who were receiving high doses of cimetidine to inhibit
evidence that IFN-γ induced immune response, after a suppressor or negative regulatory T cells. A response rate
median follow-up of almost 5 years, disease free sur- of 1/15 was observed with such a product in the setting of
vival was worse for patients who received IFN-γ (p = metastatic colon cancer [19].
0.03) and they had a trend to worse survival (p = 0.12).
Monoclonal Antibodies
Interleukin-2 Early trials with murine anti-CEA monoclonal antibodies
There is only limited clinical trial data with interleukin-2 (Mabs) were not associated with clinical responses [16].
(IL-2) in colorectal cancer, and most of the experience However as described in detail elsewhere in this book,
662 Biological therapy of colon cancer

there are now three monoclonal antibody products that trials compared 5-FU + levamisole, or 5-FU + leuco-
are commercially available with marketing indications for vorin to the same agents plus edrecolomab in Dukes C
colon cancer. These include the anti-VEGF Mab bevaci- colon cancer using the same dose and schedule used
zumab, the chimeric anti-EGFR Mab cetuximab, and in the German trial. European trials compared 17-1A
the human anti-EGFR Mab panitumumab. The murine antibody alone, to 5-FU + eucovorin, and to 5-FU +
anti-Epcam Mab edrocolomab was approved in Germany leucovorin + 17-1A. Edrecolomab was also studied in
for the treatment of colon cancer several years earlier. combination with chemotherapy and radiotherapy
in Dukes B and C rectal cancer. Results of the large ran-
domized European adjuvant trial did not demonstrate a
Edrocolomab (17-1A, PANOREX®) benefit for the use of edrecolomab as adjuvant therapy
Edrecolomab is a murine IgG2a now known to react with [70]. There were 2,761 patients randomized in the three-
the 37–40 kd glycoprotein epithelial cell adhesion mole- arm trial. The major toxicities observed in the edreco-
cule (epcam) that is expressed on various adenocarcino- lomab-alone arm were diarrhea in 32%, although severe
mas and on normal epithelial tissues [31, 38]. Single or life-threatening diarrhea was noted in only 2%.
injections of edrecolomab at doses of 15–1,000 mg were Results in the edrecolomab-alone arm were inferior to
well tolerated, but 50% of patients developed HAMA after the 5FU-containing arms (p < 0.05), and there was no
a single injection [22, 78, 79] summarized in Table 1, as a evidence of better results when edrocolomab was com-
single agent, edrocolomab exhibited minimal anti-tumor bined with 5FU (p = 0.53). Three-year survival rates
effects against colon cancer. There were no objective were 76% for 5FU plus leuovorin, 75% for the antibody
responses among 40 patients who received single infu- plus chemotherapy combination, and 70% for the anti-
sions of 15–1,000 mg of 17-1A during phase I trials in body alone. The differences in 3-year event free survival
patients with various gastrointestinal adenocarcinomas also revealed inferior results for antibody alone, and no
[78, 79]. In a phase II trial of 20 patients who received advantage for the chemotherapy plus antibody combi-
200–850 mg of 17-1A, there was one response in a patient nation. In another trial 377 patients with stage 2 colon
with recurrent rectal cancer [24]. In a trial of 25 patients cancer, stratified by whether the primary tumor was T3
who received one to four doses of 400 mg of 17-1A, one or T4, were randomized post-operatively to treatment
patient had a complete response [52]. Subsequently, this with 900 mg edrecolomab or observation [35]. The
group treated eight patients with a 17-1A chimeric anti- study was stopped because of discontinuation of drug
body with a human IgG4 subclass heavy chain but there supply in Germany after negative results from other tri-
were no responders [53]. Mellstedt and colleagues reported als became public. After a median follow-up of 3.5 years
one objective response among 52 patients who received there was no difference in overall survival or disease-
17-1A by a variety of doses and schedules [24, 57]. free survival.
An apparent benefit for adjuvant edrocolomab was Several trials have explored edrecolomab in combina-
demonstrated in a multicenter randomized German trial in tion with other biologicals with or without chemother-
patients with resected stage III colorectal cancer [71]. apy. Weiner et al. gave 150 mg of 17-1A on days 2 to 4 in
There were 90 patients randomized to observation, and 99 combination with 1.0 MIU/m2 of IFN-γ on days 1 to 4, to
to a post-operative regimen of edrecolomab 500 mg i.v., 19 colorectal cancer patients, but no antitumor responses
then 100 mg i.v. once a month for 4 months. After a median were noted [95]. In a second trial 27 patients with colon
follow up of 5 years, there was superior disease-free and or pancreatic cancer were given IFN-γ at doses up to 40
overall survival for the Mab-treated group. This included MIU/day for 4 days followed by 400 mg 17-1A on day 5
a 30% reduction in death rate and 27% reduction in tumor [94]. No objective tumor responses were seen and the
recurrence, which was similar to results reported in U.S. authors concluded that the low dose of IFN-γ was as
trials of 5-fluououracil and levamisole that had led to effective as higher doses. Saleh et al. treated 15 colorec-
acceptance of 5-FU based chemotherapy in the adjuvant tal cancer patients with 0.1 mg/m2 IFN-γ on days 1 to 15
treatment of colon cancer. After a median follow up of 7 and 400 mg of 17-1A at a dose of 400 mg on days 5, 7, 9
years, the apparent benefits of edrecolomab persisted with and 22 [75]. No significant objective tumor responses
a 23% reduction in recurrence and 32% reduction of death were described.
in the antibody group [72]. On the basis of this trial,
edrecolomab was approved for the treatment of Dukes C
colorectal cancer in Germany in 1995.
Bevacizumab (Avastin®)
Unfortunately these promising results were not con- Bevacizumab is a humanized Mab that binds to the
firmed by four additional phase III trials. North American VEGF ligand. As a single agent bevacizumab rarely
Robert O. Dillman 663

produced objective responses in a three-arm random- leucovorin and bevacizumab (FL-BV) as initial treat-
ized trial in patients with metastatic colorectal cancer ment for patients with metastatic colorectal cancer [47].
[28]. However, a three-arm randomized phase II trial in For the combined data, FL-BV (n = 249) was superior to
patients who had untreated metastatic colorectal cancer FL (n = 241) in terms of response rate (34% versus 24%,
suggested that bevacizumab enhanced 5-FU based che- p = 0.019), PFS (9 versus 6 months, HR = 0.63, p < 0.001),
motherapy [46]. The 144 patients were randomized to and OS (18 versus 15 months, HR 0.74, p = 0.008). In
fluorouracil and leucovorin (both at 500 mg/m2) as the contrast to these excellent results in previously untreated
control arm, or to the same therapy plus bevacizumab q patients, in an expanded access trial 350 patients with
2 weeks at either 5 mg/kg or 10 mg/kg. 5FU and leuco- metastatic colorectal cancer, who had relapsed after or
vorin were given weekly for the first 6 weeks of each progressed during both irinotecan and oxaloplatin based
8-week cycle. Better results were seen with the addition therapy, the response rate was only 4% in the first 100
of bevacizumab, and there appeared to be an advantage patients enrolled by investigator analysis, and only 1%
for the lower dose. based on blinded central review [7].
In the pivotal trial for regulatory approval, 923 patients A response rate of 49% was observed among 81 eval-
with metastatic colorectal cancer were randomized to uable patients with untreated advanced colorectal cancer
receive irinotecan, 5-FU and leucovorin (IFL) and beva- who were treated with IFL and bevacizumab [27]. The
cizumab (IFL-BV), IFL and placebo (control), or FL doses of irinotecan and 5FU both had to be reduced by
with bevacizumab (FL-BV) at a dose of 5 mg/kg every 2 20% to 25% because of vomiting, diarrhea and neutro-
weeks [42, 43] the first phase of the trial, 313 patients penia. Bleeding occurred in 37 patients (46%) and nine
were randomly assigned to these three arms, then; after a patients (11%) had grade 3 or 4 thromboembolic events.
planned initial analysis, the FL-BV arm was discontin- In another trial FOLFOX4 and bevacizumab was used
ued after enrollment of 110 patients, not because of infe- to treat 53 patients with previously untreated metastatic
rior results, but because IFL had become the standard colorectal cancer [21]. The response rate was 68%.
control arm based on other trials in metastatic colorectal Hemorrhage was not a problem in this trial, and only
cancer. IFL-BV was superior to IFL-placebo for the 813 one patient had a severe thromboembolic event.
patients randomized to treatment with one of these arms,
in terms of response rate (p = 0.004), PFS (HR 0.54, p <
0.001), and OS (HR 0.66, p < 0.001) [42]. The IFL-BV
Cetuximab (Erbitux®)
arm was associated with more grade 3 or 4 hypertension Cetuximab is a chimeric Mab that binds to EGFR.
(11% versus 2%). The FL-BV arm also was superior to Clinical trials with cetuximab used an initial loading
the IFL-placebo arm [43]. dose of 400 mg/m2 i.v. over 2 h, then 250 mg/m2 i.v. over
In a trial of initial therapy for patients with metastatic 1 h weekly. Saltz et al. treated 57 patients whose met-
colorectal cancer who were not considered optimal can- static cancer had not responded to prior irinotecan alone
didates for first-line irinotecan treatment, 209 patients or as part of combination chemotherapy [76]. Patients
were randomized to receive FL + becacizumab at 5 mg/kg were required to have EGFR demonstrated by immuno-
or FL + placebo [48]. The addition of bevacizumab histochemistry on formalin-fixed paraffin-embedded
resulted in a better PFS (HR = 0.50, p < 0.001), higher tumor tissue, but it is now known that quantitative
response rate (p = 0.055), and a trend toward better sur- expression of EGFR is not predictive of response to
vival (HR = 0.79, p = 0.16). Hypertension was more cetuximab treatment. The most common adverse events
frequent with bevacizumab. were an acne-like skin rash, predominantly on the face
In a three-arm trial 829 patients whose cancers had and upper chest (86%) and the constellation of asthenia,
recurred after IFL therapy were randomized to receive fatigue, malaise, or lethargy (56%). Three patients were
oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) reported to have had a grade 3 allergic reaction; two
with bevacizumab or to FOLFOX4 alone, or to bevaci- were withdrawn.
zumab alone [28]. In this trial bevacizumab was given at Lenz et al. treated reported a response rate of 12% for
10 mg/kg every 2 weeks. As noted earlier, bevacizumab 346 patients whose metastatic cancer was considered
showed little activity as a single agent, but FOLFOX4 refractory to fluoropyrimidines, irinotecan and oxalipla-
plus bevacizumab was superior to FOLFOX alone for all tin [51]. EGFR positivity by IHC was an eligibility
key endpoints, including OS (HR 0.75, p = 0.001), PFS requirement, but degree of positivity did not correlate
(HR 0.61, p < 0.0001) and response rate (p < 0.0001). with clinical benefit. The most prevalent toxicity was
A metanalysis was performed using the raw data the acneiform rash which was observed in 83% of
from three randomized trials that included 5-FU plus patients, and which was predictive of clinical benefit.
664 Biological therapy of colon cancer

In a randomized trial, 572 patients who had pro- Panitumumab (Vectibix®)


gressed despite 5FU, irinotecan, and oxaloplatin, were
either observed or received standard dose cetuximab Panitumumab is a human Mab that reacts with EGFR.
[45]. The objective response rate was only 8%, but PFS A pivotal trial for regulatory approval compared panitu-
and OS were better in the cetuximab arm. Skin rash, mumab at 6 mg/kg i.v. every 2 weeks to best supportive
infection and hypomagnesemia were more common in care in patients with metastatic colorectal cancer whose
the cetuximab arm. Eleven patients discontinued cetux- disease had progressed during or after standard therapy
imab because of infusion reactions. with fluoropyrimidine-, oxaliplatin-, and irinotecan-
In the pivotal trial Cunningham et al. randomized 329 containing chemotherapy regimens [29, 30, 84, 86]. All
patients with colorectal cancer that had progressed patients had epidermal growth factor receptor (EGFR)-
during or within 3 months after discontinuation of expressing tumors. Panitumumab produced a 10%
irinotecan, to receive either, irinotecan plus cetuximab, objective response rate and a longer progression free
or cetuximab alone, using a 2:1 randomization [11]. survival compared to supportive care alone. There was
Protocol prescribed irinotecan was to be the same as no difference in survival, but approximately 75% of
used prestudy. The response rate was higher for the patients in the supportive care alone arm crossed over to
combination therapy (23% versus 11%, p = 0.007), as receive panitumumab after disease progression. Quality
was PFS (4.0 months versus 1.5 months, p < 0.001). of life was also better in the patients who received pani-
Gebbia et al. reported a response rate of 20% for tumumab [80]. For 176 patients who progressed on the
cetuximab plus irinotecan in 60 patients who had supportive care arm, and then subsequently did receive
received at least two prior therapies, and whose meta- panitumumab, 12% had an objective response and two
static cancer was considered refractory to oxaliplatin patients had complete responses [86].
and irinotecan [25]. Standard cetuximab dosing was Hecht et al. treated 148 patients whose metastatic col-
combined with irinotecan 120 mg/m2 weekly for 4 out orectal cancer had progressed on chemotherapy that
of 6 weeks. Responses were not predicted by EGFR included a fluoropyrimidine and irinotecan or oxaliplatin,
expression. Other than the acneiform skin rash that was or both [37]. Panitumumab was given i.v. at a dose of
severe in 13% of patients, the main grade 3 or 4 toxici- 2.5 mg/kg weekly for 8 of each 9 weeks until disease pro-
ties were attributable to the chemotherapy. gression or excessive toxicity. Skin toxicity occurred in
Vincenzi et al. reported a 25% response rate for cetux- 95% and 5% were grade 3 or 4. Four patients discontinued
imab and irinotecan in 55 patients whose metastatic can- therapy because of toxicity and one patient had an infu-
cer was considered refractory to oxaliplatin and irinotecan sion reaction but was able to resume treatment. EGFR of
[88]. Standard cetuximab dosing was combined with iri- at least 1+ by IHC was an eligibility requirement. There
notecan 90 mg/m2 weekly. Skin toxicity was observed in was no difference in response for 105 patients who were
89% of patients. The most common grade 3 to 4 adverse judged as having high EGFR by IHC compared to 43
events were dermatologic (33%), diarrhea (16%), fatigue patients who were characterized as having a low EGFR.
(13%) and stomatitis (7%). Fever was noted in 25%, Berlin et al. tested the combination of irinotecan, 5-FU
typically in association with the first infusion of cetux- and leucovorin, and panitumumab as initial therapy in
imab, but no allergic reactions were recorded. patients with metastatic colorectal cancer [2]. This was a
More recently cetuximab has been combined with two-part, multicenter, phase II study of panitumumab
modern combination therapies such as FOLFOX and 2.5 mg/kg weekly with bolus 5-FU (IFL) in the first part
FOLFIRI as the initial treatment of patients with metastatic of the trial, and infusional 5-FU (FOLFIRI) in the second
colorectal cancer. Tabernero et al. used FOLFOX and part. Grade 3 to 4 diarrhea occurred in 11 patients (58%)
cetuximab as the initial treatment for 43 patients with in part 1 and six patients (25%) in part 2. All patients had
metastatic colorectal cancer [83]. Cetuximab was given dermatologic toxicity, but none was grade 4. The authors
day 1 at a dose of 400 mg/m2 during week 1, and then concluded that panitumumab + FOLFIRI was better tol-
250 mg/m2 weekly thereafter. Treatment was well-tolerated erated than panitumumab + IFL.
and the response rate was 72%. In a randomized trial of
337 patients, a preliminary report showed that cetuximab
plus FOLFOX was superior to FOLFOX alone in terms of
Vaccines
response rate (46% versus 36% p = 0.064) [3]. In a ran- A number of trials were carried out with a vaccine consist-
domized trial of 1,198 patients, an initial report showed ing of about 10 million irradiated autologous cells obtained
that cetuximab plus FOLFIRI was superior to FOLFIRI directly from fresh tumor, admixed with about 10 million
alone in terms of response rate (47% versus 39%, p = 0.004) bacillus Calmette-Guérin (BCG) organisms and injected
and PFS (8.9 versus 8.0 months p = 0.048) [85]. i.d. weekly for 2 weeks, and then an additional injection
Robert O. Dillman 665

of irradiated tumor cells during the third week. Initial edrocolomab. All three of these products had no or little
trials had shown that injections of such a product were activity in the advanced disease setting, and all eventu-
well-tolerated, associated with local delayed type hyper- ally fell out of favor in the adjuvant setting because of
sensitivity reactions to autologous tumor, and isolation of results from randomized trials. Combinations of inter-
human monoclonal antibodies from circulating lympho- feron and 5FU appeared encouraging in patients with
cytes [36, 40]. In a European phase III trial 254 patients advanced disease, but eventually were found to be want-
with stage II or stage III colon cancer were randomized to ing in both the metastatic and adjuvant settings based on
observation or active specific immunotherapy with this results of large randomized trials. Recently, despite lim-
preparation beginning 4 weeks after surgery [87]. At a ited activity as single agents in the advanced setting,
median follow-up of more than 5 years, there was a 44% two monoclonal antibodies have gained approval based
reduction in the risk of recurrence in the treatment arm on their impressive activity in combination with chemo-
(p = 0.023). In the subset analysis there was no benefit for therapy in patients with metastatic disease, including
patients with stage III disease, but there was a great benefit enhanced survival. Bevacizumab and cetuximab are
in terms of decrease risk of recurrence and death in the now standard components of systemic treatment regi-
vaccine arm. In a U.S. phase III trial 297 patients with mens, but only in combination with chemotherapy.
stage II colon cancer, and 115 with stage III colon cancer,
were randomized to observation or to the autologous
active specific immunotherapy starting 1 month after
surgery [34]. After a median follow up of more than 7.5
References
years, there was no difference in outcomes between the 1. Arnaud JP, Buyse M, Nordlinger B, et al. Adjuvant therapy of poor
prognosis colon cancer with levamisole: results of an EORTC
two treatments based on an intent-to-treat analysis,
double-blind randomized clinical trial. Br J Surg 1989;76:284–9.
although there were substantial problems in compliance 2. Berlin J, Posey J, Tchekmedyian S, et al. Panitumumab with irino-
with the injection schedule. This product was not granted tecan/leucovorin/5-fluorouracil for first-line treatment of meta-
regulatory approval. In addition to the inconclusive trials, static colorectal cancer. Clin Colorectal Cancer 2007;6:427–32.
there were issues regarding sterility of such a product 3. Bokemeyer C, Bondarenko I, Makhson A, et al. Cetuximab plus
5-FU/FA/Oxaliplatin (FOLFOX-4) versus FOLFOX-4 in the
derived from fresh colon tumor, and the variability of the first-line treatment of metastatic colorectal cancer: OPUS, a ran-
product in terms of tumor cells versus stromal and immune domized phase II study. Proc Am Soc Clin Oncol 2007;25:172s
cells in fresh tumor. [abstract 4035].
Another vaccine that has been explored in colon 4. Bonnem EM, Spiegel RJ. Interferon-alpha: current status and
cancer consists of a non-replicating canarypoxvirus future promise. J Biol Response Mod 1984; 3:580–8.
5. Cascinu S, Catalano V, Piga A, et al. The role of levamisole in the
(ALVAC) engineered to express carcinoembryonic anti- adjuvant treatment of stage III colon cancer patients: a randomized
gen (CEA) and the B7.1 costimulatory molecule. In a trial of 5-fluorouracil and levamisole versus 5-fluorouracil alone.
phase I trial eight patients were treated with the vaccine Cancer Invest 2003;21:701–7.
[55]. The investigators were able to isolate cytotoxic T 6. Chaplinski T, Laszlo J, Moore J, Silverman P. Phase II trial of lym-
phoblastoid interferon in metastatic colon carcinoma. Cancer Treat
cells that could lyse allogeneic and autologous tumor
Rep. 1983;67:1009–12.
cells in a MHC-restricted manner [99]. In a second phase 7. Chen HX, Mooney M, Boron M, et al. Phase II multicenter trial of
I trial involving patients with CEA-expressing adenocar- bevacizumab plus fluorouracil and leucovorin in patients with
cinomas, three cohorts of six patients were treated with advanced refractory colorectal cancer: an NCI Treatment Referral
increasing doses of an ALVAC-CEA-B7.1 vaccine at Center Trial TRC-0301. J Clin Oncol 2006;24:3354–60.
8. Chlebowski RT, Nystrom S, Reynolds R, et al. Long-term survival
doses of 45 million, 450 million, and 0.45 billion plaque-
following levamisole or placebo adjuvant treatment of colorectal can-
forming units delivered i.m. monthly for 3 months [41, cer: a Western Cancer Study Group Trial. Oncology 1988;45:141–3.
56]. In this trial there again was evidence of induction of 9. Clark PI, Slevin ML, Reznek RH, et al. Two randomised phase II
specific T cell immune reactivity to CEA. trials of intermittent intravenous versus subcutaneous alpha-2
interferon alone (trial 1) and in combination with 5-fluorouracil
(trial 2) in advanced colorectal cancer. Int J Colorectal Dis
1987;2:26–9.
Summary 10. Colucci G, Maiello E, Gebbia V, et al. 5-fluorouracil and levofolinic
acid with or without recombinant interferon-2b in patients with
The history of immunotherapy in colon cancer is note- advanced colorectal carcinoma: a randomized multicenter study
worthy because of the brief periods of success enjoyed with stratification for tumor burden and liver involvement by the
Southern Italy Oncology Group. Cancer 1999;85:535–45.
in the adjuvant setting by the BCG-tumor cell vaccine, 11. Cunningham D, Humblet Y, Siena S, et al. Cetuximab monotherapy
the non-specific immune stimulator levamisole in com- and cetuximab plus irinotecan in irinotecan-refractory metastatic
bination with 5FU, and the anti-epcam murine Mab colorectal cancer. N Engl J Med 2004;351:337–45.
666 Biological therapy of colon cancer

12. Diaz RE, Jimeno J, Camps C, et al. Treatment of advanced col- results from the Eastern Cooperative Oncology Group Study
orectal cancer with recombinant interferon alpha and fluorouracil: E3200. J Clin Oncol 2007;25:1539–44.
activity in liver metastasis. Cancer Invest 1992; 10:259–64. 29. Gibson TB, Ranganathan A, Grothey A. Randomized phase III
13. Di Costanzo F, Sobrero A, Gasperoni S, et al. Adjuvant chemo- trial results of panitumumab, a fully human anti-epidermal growth
therapy in the treatment of colon cancer: randomized multicenter factor receptor monoclonal antibody, in metastatic colorectal cancer.
trial of the Italian National Intergroup of Adjuvant Chemotherapy Clin Colorectal Cancer 2006;6:29–31.
in Colon Cancer (INTACC). Ann Oncol 2003;14:1365–72. 30. Giusti RM, Shastri KA, Cohen MH, et al. FDA drug approval
14. Dillman RO, Beauregard JC, Zavanelli MI, et al. In vivo immune summary: panitumumab (Vectibix). Oncologist 2007;12:577–83.
restoration in advanced cancer patients after administration of 31. Gottlinger HG, Funke I, Hohnson JP, et al. The epithelial cell sur-
thymosin fraction 5 or thymosin alpha 1. J Biol Response Mod face antigen PANOREX, a target for antibody-mediated tumor
1983;2:139–49. therapy: its biochemical nature, tissue distribution and recognition
15. Dillman RO, Beauregard J, Royston I, Zavanelli MI. Phase II trial by different monoclonal antibodies. Int J Cancer 1986;38:47–53.
of thymosin fraction 5 and thymosin alpha 1. J Biol Response Mod 32. Grem JL, Jordan E, Robson ME, et al. Phase II study of fluorouracil,
1987a;6:263–7. leucovorin, and interferon alfa-2a in metastatic colorectal carci-
16. Dillman RO, Beauregard J, Ryan KP, et al. Radioimmunodetection noma. J Clin Oncol 1993; 11:1737–45.
of cancer using indium-labeled monoclonal antibodies. International 33. Haller DG, Catalano PJ, Macdonald JS, et al. Phase III study of
symposium on labeled and unlabeled antibodies in cancer diagnosis fluorouracil, leucovorin, and levamisole in high-risk stage II and
and therapy. NCI Monogr 1987b; 3:33–6. III colon cancer: final report of Intergroup 0089. J Clin Oncol
17. Dillman RO, Oldham RK, Tauer KW, et al. Continuous interleukin-2 2005;23:8671–8.
and lymphokine-activated killer cells for advanced cancer: a National 34. Harris JE, Ryan L, Hoover HC Jr, et al. Adjuvant active specific
Biotherapy Study Group trial. J Clin Oncol 1991; 9:1233–40. immunotherapy for stage II and III colon cancer with an autolo-
18. Dillman RO, Church C, Oldham RK, et al. Inpatient continuous- gous tumor cell vaccine: Eastern Cooperative Oncology Group
infusion interleukin-2 in 788 patients with cancer. The National Study E5283. J Clin Oncol 2000;18:148–57.
Biotherapy Study Group experience. Cancer 1993; 71:2358–70. 35. Hartung G, Hofheinz RD, Dencausse Y, et al. Adjuvant therapy with
19. Dillman RO, Soori G, DePriest C, et al. Treatment of human solid edrecolomab versus observation in stage II colon cancer: a multi-
malignancies with autologous activated lymphocytes and cimeti- center randomized phase III study. Onkologie 2005;28:347–50.
dine: a phase II trial of the Cancer Biotherapy Research Group. 36. Haspel MV, McCabe RP, Pomato N, et al. Generation of tumor
Cancer Biother Radiopharm 2003;18:727–33. cell-reactive human monoclonal antibodies using peripheral
20. Dillman RO, Schiltz PM, DePriest C, et al. Tumor- infiltrating lym- blood lymphocytes from actively immunized colorectal carcinoma
phocytes and interleukin-2: dose and schedules of administration in patients. Cancer Res 1985;45:3951–61.
the treatment of metastatic cancer. Cancer Biother Radiopharm 37. Hecht JR, Patnaik A, Berlin J, et al. Panitumumab monotherapy in
2004;19:730–7. patients with previously treated metastatic colorectal cancer.
21. Emmanouilides C, Sfakiotaki G, Androulakis N, et al. Front-line Cancer 2007;110:980–8.
bevacizumab in combination with oxaliplatin, leucovorin and 38. Herlyn M, Steplewski Z, Herlyn D, et al. CO17–1A and related
5-fluorouracil (FOLFOX) in patients with metastatic colorectal monoclonal antibodies: their production and characterization.
cancer: a multicenter phase II study. BMC Cancer 2007;7:91. Hybridoma 1986;5:S3–S10.
22. Fagerberg J, Ragnhammar P, Liljefors M, et al. Humoral anti-idio- 39. Hill M, Norman A, Cunningham D, et al. Royal Marsden phase III
typic and anti-antiidiotypic immune response in cancer patients trial of fluorouracil with or without interferon alfa-2b in advanced
treated with monoclonal antibody 17–1A. Cancer Immunol colorectal cancer. J Clin Oncol 1995;13:1297–302.
Immunother 1996;42:81–7. 40. Hoover JC Jr, Brandhorst JS, Peters LC, et al. Adjuvant active spe-
23. Fountzilas G, Zisiadis A, Davni U, et al. Fluorouracil and leuco- cific immunotherapy for human colorectal cancer: 6.5-year median
vorin with or without interferon alfa-2a as adjuvant treatment, in follow-up of a phase III prospectively randomized trial. J Clin
patients with high-risk colon cancer: a randomized phase III study Oncol 1993;11:390–9.
conducted by the Hellenic Cooperative Oncology Group. Oncology 41. Horig H, Lee DS, Conkright W, et al. Phase I clinical trial of a
2000;58:227–36. recombinant canarypoxvirus (ALVAC) vaccine expressing human
24. Frodin JE, Harmenberg U, Biberfeld P, et al. Clinical effects of carcinoembryonic antigen and the B7.1 co-stimulatory molecule.
monoclonal antibodies [MAb 17–1A] in patients with metastatic Cancer Immunol Immunother 2000;49:504–14.
colorectal carcinomas. Hybridoma 1988;7:309–321. 42. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus
25. Gebbia V, Del Prete S, Borsellino N, et al. Efficacy and safety of irinotecan, fluorouracil, and leucovorin for metastatic colorectal
cetuximab/irinotecan in chemotherapy-refractory metastatic col- cancer. N Engl J Med 2004;350:2335–42.
orectal adenocarcinomas: a clinical practice setting, multicenter 43. Hurwitz HI, Fehrenbacher L, Hainsworth JD, et al. Bevacizumab
experience. Clin Colorectal Cancer 2006;5:422–8. in combination with fluorouracil and leucovorin: an active regimen
26. Gennatas C, Mouratidou D, Androulakis G, et al. Adjuvant sys- for first-line metastatic colorectal cancer. J Clin Oncol 2005;23:
temic therapy protocol for Dukes’ B2 and C resectable colon car- 3502–8.
cinoma. Tumori 2002;88:32–6. 44. John WJ, Neefe JR, Macdonald JS, et al. 5-fluorouracil and inter-
27. Giantonio BJ, Levy DE, O’dwyer PJ, et al. A phase II study of feron-alpha-2a in advanced colorectal cancer. Results of two treat-
high-dose bevacizumab in combination with irinotecan, 5-fluorou- ment schedules. Cancer 1993; 72:3191–5.
racil, leucovorin, as initial therapy for advanced colorectal cancer: 45. Jonker DJ, O’Callaghan CJ, Karapetis C, et al. Cetuximab for the
results from the Eastern Cooperative Oncology Group study treatment of colorectal cancer. N Engl J Med 2007;357:2040–8.
E2200. Ann Oncol 2006;17:1399–403. 46. Kabbinavar F, Hurwitz HI, Fehrenbacher L, et al. Phase II, ran-
28. Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in domized trial comparing bevacizumab plus fluorouracil (FU)/
combination with oxaliplatin, fluorouracil, and leucovorin leucovorin (LV) with FU/LV alone in patients with metastatic
(FOLFOX4) for previously treated metastatic colorectal cancer: colorectal cancer. J Clin Oncol 2003;21:60–5.
Robert O. Dillman 667

47. Kabbinavar FF, Hambleton J, Mass RD, et al. Combined analysis advanced solid tumors: a National Biotherapy Study Group Trial.
of efficacy: the addition of bevacizumab to fluorouracil/leucovorin Mol Biother 1991; 3:68–73.
improves survival for patients with metastatic colorectal cancer. J 65. Oldham RK, Blumenschein G, Schwartzberg L, et al. Combination
Clin Oncol 2005a;23:3706–12. biotherapy utilizing interleukin-2 and alpha interferon in patients
48. Kabbinavar FF, Schulz J, McCleod M, et al. Addition of bevaci- with advanced cancer: a National Biotherapy Study Group Trial.
zumab to bolus fluorouracil and leucovorin in first-line metastatic Mol Biother 1992;4:4–9.
colorectal cancer: results of a randomized phase II trial. J Clin 66. Osband ME, Lavin PT, Babayan RK, et al. Effect of autolympho-
Oncol. 2005b;23:3697–705. cyte therapy on survival and quality of life in patients with meta-
49. Kemeny N, Younces A. Alfa-2a interferon and 5-fluorouracil for static renal cell carcinoma. Lancet 1990;335:994–8.
advanced colorectal carcinoma: the Memorial Sloan-Kettering 67. Palmeri S, Meli M, Danova M, et al. 5-Fluorouracil plus interferon
experience. Semin Oncol 1992; 19(Suppl 3):171–5. alpha-2a compared to 5-fluorouracil alone in the treatment of
50. Kosmidis PA, Tsavaris N, Skarlos D, et al. Fluorouracil and leuco- advanced colon carcinoma: a multicentric randomized study. J
vorin with or without interferon alfa-2b in advanced colorectal cancer: Cancer Res Clin Oncol 1998;124:191–8.
analysis of a prospective randomized phase III trial. Hellenic 68. Pazdur R, Moore DR Jr, Bready B. Modulation of fluorouracil
Cooperative Oncology Group. J Clin Oncol 1996;14:2682–7. with recombinant alfa interferon: M.D. Anderson Clinical Trial.
51. Lenz HJ, Van Cutsem E, Khambata-Ford S, et al. Multicenter Semin Oncol 1992; 19 (Suppl 3):176–9.
phase II and translational study of cetuximab in metastatic col- 69. Punt CJ, Burghouts JT, Croles JJ, et al. Continuous infusion of
orectal carcinoma refractory to irinotecan, oxaliplatin, and fluoro- high-dose 5-fluorouracil in combination with leucovorin and
pyrimidines. J Clin Oncol 2006;24:4914–21. recombinant interferon-alpha-2b in patients with advanced colorec-
52. LoBuglio AF, Saleh MN, Lee J, et al. Phase I trial of multiple large tal cancer. A Multicenter Phase II study. Cancer 1993;72:2107–11.
doses of murine monoclonal antibody CO17–1A. Clinic aspects. J 70. Punt CJ, Nagy A, Douillard JY, et al. Edrecolomab alone or in
Natl Cancer Inst 1988; 17:932–6. combination with fluorouracil and folinic acid in the adjuvant
53. LoBuglio AF, Wheeler RH, Trang J, et al. Mouse/human chimeric treatment of stage III colon cancer: a randomised study. Lancet
monoclonal antibody in man: kinetics and immune response. Proc 2002;360:671–7.
Natl Acad Sci USA 1989; 86:4220–4. 71. Riethmuller G, Schneider-Gadicke E, Schlimok G, et al. Randomized
54. Lundell G, Blomgren H, Cedermark B et al. High dose rDNA trial of monoclonal antibody for adjuvant therapy of resected Dukes
human alpha 2 interferon therapy in patients with advanced col- C colorectal carcinoma. Lancet 1994; 343:1177–83.
orectal adenocarcinoma: a phase II study. Radiother Oncol 72. Riethmuller G, Holz E, Schlimok G, et al. Monoclonal antibody
1984;1:325. therapy for resected Dukes’ C colorectal cancer: seven-year
55. Marshall JL, Hawkins MJ, Tsang KY, et al. Phase I study in cancer outcome of a multicenter randomized trial J Clin Oncol 1998;
patients of a replication-defective avipox recombinant vaccine that 16:1788–94.
expresses human carcinoembryonic antigen. J Clin Oncol 73. Rosenberg SA, Lotze MT, Yang JC, et al. Experience with the use
1999;17:332–7. of high-dose interluekin-2 in the treatment of 652 patients. Ann
56. Marshall JL, Hoyer RJ, Toomey MA, et al. Phase I study in Surg 1989;210:474–85.
advanced cancer patients of a diversified prime-and-boost vaccina- 74. Rosenberg SA, Lotze MT, Yang JC, et al. Prospective randomized
tion protocol using recombinant vaccinia virus and recombinant trial of high-dose interluekin-2 alone or in conjunction with
nonreplicating avipox virus to elicit anti-carcinoembryonic anti- lymphokine-activated killer cells for the treatment of patients with
gen immune responses. J Clin Oncol 2000;18:3964–73. advanced cancer. J Natl Cancer Inst 1993;85:622–32.
57. Mellstedt H, Frodin J-E, Masucci G, et al. The therapeutic use of 75. Saleh MN, LoBuglio AF, Wheeler RH, et al. A phase II trial of
monoclonal antibodies in colorectal carcinoma. Semin Oncol murine monoclonal antibody 17–1A and interferon-gamma: Clinical
1991; 18:462–77. and immunological data. Cancer Immunol 1990; 32:185–90.
58. Moertel CG, Fleming TR, Macdonald JS, et al. Levamisole and 76. Saltz LB, Meropol NJ, Loehrer PJ Sr, et al. Phase II trial of
fluorouracil for adjuvant therapy of resected colon carcinoma. N cetuximab in patients with refractory colorectal cancer that
Engl J Med 1990;322:352–8. expresses the epidermal growth factor receptor. J Clin Oncol
59. Moertel CG, Fleming TR, Macdonald JS, et al. Fluorouracil plus 2004;22:1201–8.
levamisole as effective adjuvant therapy after resection of stage III 77. Schippinger W, Jagoditsch M, Sorre C, et al. A prospective ran-
colon carcinoma: a final report. Ann Intern Med 1995a;122:321–6. domised trial to study the role of levamisole and interferon alfa in
60. Moertel CG, Fleming TR, Macdonald JS, et al. Intergroup study of an adjuvant therapy with 5-FU for stage III colon cancer. Br J
fluorouracil plus levamisole as adjuvant therapy for stage II/ Cancer 2005;92:1655–62.
Dukes’ B2 colon cancer. J Clin Oncol 1995b;13:2936–43. 78. Sears HF, Atkinson B, Mattis J, et al. Phase I clinical trial of mono-
61. Namba M, Miyoshi T, Kanamori T. Combined effects of 5-fluro- clonal antibody in treatment of gastrointestinal tumors. Lancet
rouracil and interferon on proliferation of human neoplastic cells 1982; 1:762–5.
in culture. Jpn J Cancer Res 1982;73:819–24. 79. Sears HF, Herlyn D, Steplewski Z, et al. Phase II clinical trial of a
62. Neefe JR, Silgals R, Ayoob M, Schein PS. Minimal activity of murine monoclonal antibody cytotoxic for gastrointestinal adeno-
recombinant clone A interferon in metastatic colon cancer. J Biol carcinoma. Cancer Res 1985; 45:5910–13.
Resp Modif 1984; 3:366–70. 80. Siena S, Peeters M, Van Cutsem E, et al. Association of progression-
63. Nordlinger B, Rougier P, Arnaud JP, et al. Adjuvant regional che- free survival with patient-reported outcomes and survival: results
motherapy and systemic chemotherapy versus systemic chemo- from a randomised phase 3 trial of panitumumab. Br J Cancer
therapy alone in patients with stage II-III colorectal cancer: a 2007;97:1469–74.
multicentre randomised controlled phase III trial. Lancet Oncol 81. Staib L, Link KH, Beger HG, et al. Toxicity and effects of adjuvant
2005;6:459–68. therapy in colon cancer: results of the German prospective, con-
64. Oldham RK, Dillman RO, Yannelli JR, et al. Continuous infusion trolled randomized multicenter trial FOGT-1. J Gastroint Surg
interleukin-2 and tumor-derived activated cells as treatment of 2001;5:275–81.
668 Biological therapy of colon cancer

82. Stevenson HC, Green I, Hamilton JM, et al. Levamisole: known 92. Wadler S, Lembersky B, Atkins M, et al. Phase II trial of fluorou-
effects on the immune system, clinical results, and future applica- racil and recombinant interferon alfa-2a in patients with advanced
tions to the treatment of cancer. J Clin Oncol 1991;9:2052–66. colorectal carcinoma: an Eastern Cooperative Oncology Group
83. Tabernero J, Van Cutsem E, Diaz-Rubio E, et al. Phase II trial of study. J Clin Oncol 1991;9:1806–10.
cetuximab in combination with fluorouracil, leucovorin, and oxali- 93. Weh HJ, Platz D, Braumann D. Treatment of metastatic colorectal
platin in the first-line treatment of metastatic colorectal cancer. J carcinoma with a combination of fluorouracil and recombinant
Clin Oncol 2007;25:5225–32. interferon alfa-2b: preliminary data of a phase II study. Semin
84. Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial Oncol 2003; 19(Suppl. 3):180–4.
of panitumumab plus best supportive care compared with best sup- 94. Weiner LM, Steplewski Z, Koprowski H, et al. Biologic effects of
portive care alone in patients with chemotherapy-refractory meta- gamma interferon pre-treatment followed by monoclonal antibody
static colorectal cancer. J Clin Oncol 2007a;25:1658–64. 17–1A administration in patients with gastrointestinal carcinoma.
85. Van Cutsem E, Nowacki M, Lang I, et al. Randomized phase III study Hybridoma 1986;5 Suppl 1:S65–77.
of irinotecan and 5-FU-FA with or without cetuximab in the first-line 95. Weiner LM, Moldofsky PJ, Gatenby RA, et al. Antibody delivery
treatment of patients with metastatic colorectal cancer: the CRYSTAL and effector cell activation in a phase II trial of recombinant
trial. Proc Am Soc Clin Oncol 2007b;25:164s [abstract 4000]. gamma-interferon and the murine monoclonal antibody CO17–
86. Van Cutsem E, Siena S, Humblet Y, et al. An open-label, single- 1A in advanced colorectal carcinoma. Cancer Res 1988;48:
arm study assessing safety and efficacy of panitumumab in patients 2568–73.
with metastatic colorectal cancer refractory to standard chemo- 96. Wiesenfeld M, O’Connell MJ, Wieand HS, et al. Controlled
therapy. Ann Oncol 2008;19:92–8. clinical trial of interferon-gamma as postoperative surgical
87. Vermorken JB, Claessen AM, van Tinteren H, et al. Active specific adjuvant therapy for colon cancer. J Clin Oncol 1995;13:
immunotherapy for stage II and stage III human colon cancer: a 2324–9.
randomised trial. Lancet 1999;353:345–50. 97. Wolmark N, Bryant J, Smith R, et al. Adjuvant 5-fluorouracil and
88. Vincenzi B, Santini D, Rabitti C, et al. Cetuximab and irinotecan leucovorin with or without interferon alfa-2a in colon carcinoma:
as third-line therapy in advanced colorectal cancer patients: a single National Surgical Adjuvant Breast and Bowel Project protocol
centre phase II trial. Br J Cancer 2006;94:792–7. C-05. J Natl Cancer Inst 1998;90:1810–6.
89. Wadler S, Wiernik PH. Clinical update on the role of fluorouracil 98. Wolmark N, Rockette H, Mamounas E, et al. Clinical trial to assess
and recombinant interferon alfa-2a in the treatment of colorectal the relative efficacy of fluorouracil and leucovorin, fluorouracil
carcinoma. Semin Oncol 1990; 17:16–21 and levamisole, and fluorouracil, leucovorin, and levamisole in
90. Wadler S, Schwartz EL, Goldman M, et al. Fluorouracil and recom- patients with Dukes’ B and C carcinoma of the colon: results from
binant alfa-2a-interferon: an active regimen against advanced col- National Surgical Adjuvant Breast and Bowel Project C-04. J Clin
orectal carcinoma. J Clin Oncol 1989; 7:1769–75. Oncol 1999;17:3553–9.
91. Wadler S, Wersto R, Weinberg V, et al. Interaction of fluorouracil 99. Zhu MZ, Marshall J, Cole D, et al. Specific cytolytic T-cell
and interferon in human colon cancer cell lines: cytotoxic and responses to human CEA from patients immunized with recombi-
cytokinetic effects. Cancer Res 1990; 50: 5735–9. nant avipox-CEA vaccine. Clin Cancer Res 2000;6:24–33.
21.5 Biological therapy of breast cancer
ROBERT O. DILLMAN

Breast Cancer survival. However, because of its design, this trial did
not address whether BCG made any contribution to
Several studies indicate that patients with carcinoma these results. U.S. trials of adjuvant cyclophosphamide,
of the breast remain reasonably immune competent doxorubicin, and 5-fluorouracil, with or without BCG,
throughout the natural history of their disease [45, 91]. in patients with stage II or III breast cancer [12], or stage
Anti-tumor immunity is demonstrable in patients by a IV breast cancer [52]. A randomized trial of BCG after
number of techniques. An in vitro immune reaction to radiation therapy for locally advanced breast cancer also
tumor-associated antigens was demonstrated using the found no advantage for BCG [79].
leukocyte migration inhibition (LMI) and the leukocyte A methanol-extracted residue (MER) of BCG was
adherence inhibition (LAI) assays, using as stimulants tested in a trial in which 395 evaluable patients with
tumor cell extracts, soluble membrane extracts of MCF-7 metastatic breast cancer were randomized to one of
cells (a breast cancer cell line), and soluble membrane three different chemotherapy combinations with or
extracts of biopsy-derived tumor tissue [15, 32, 75]. without MER [1]. MER was injected at five sites at a
Several of these studies demonstrated blocking factors, dose of 100 μg or at the lowest tenfold dilution that pro-
presumably immune complexes, in the serum [83]. The duced a 1-cm indurated lesion. After adjustments for
T-antigen, a precursor of the M and N blood group anti- chemotherapy regimen, collectively response rates were
gens whose expression is masked on normal tissue, is inferior in the combined chemoimmunotherapy arms
expressed on nearly all breast carcinoma tissue. Patients (38% vs. 52%, p = 0.02). MER was associated with sig-
also mount a strong cellular immune reaction to this nificant toxicity including painful ulcers and fevers, but
antigen as demonstrated by both delayed hypersensitivity without response or survival benefit.
and in vitro LMI. The demonstration of delayed hyper-
sensitivity to tumor-associated antigens has not been
particularly useful clinically. However 85% of breast
Corynbacterium Parvum
cancer patients had such a reaction to a crude membrane NSABP conducted a trial in which breast cancer
extract prepared from cultured MCF-7 breast tumor cells patients were randomized to receive l-phenylalanine
previously infected with vesicular stomatitis virus [3]. mustard (L-PAM) and 5FU (PF) with or without C par-
vum [30]. After 8 years of follow-up there was no
improvement in disease free survival (DFS) or overall
Non-specific Immune Stimulation survival (OS), and the trend was actually for inferiority
Because of the prevalence of breast cancer, in the 1970s in the immunomodulating arm. There was also more
many studies were carried out with various nonspecific toxicity, especially fever and chills associated with
immune stimulating agents including BCG, levamisole, C parvum administration.
Corynbacterium parvum, poly A:U, and cis-retinoic
acid. As a generalization, these agents failed to demon-
strate significant benefit.
Pseudomonas Antigens
In a trial for metastatic breast cancer patients, 133 previ-
ously untreated women were treated with combination
Bacillus Calmette Guerre (BCG) chemotherapy and then randomized to no further treat-
In a European trial 254 patients with surgically resected ment or nonspecific immunotherapy with a heptavalent
breast cancer (stages T1–3a/N0–1/M0) were random- pseudomonas vaccine [38]. Women with estrogen receptor
ized to either surgery alone or surgery plus adjuvant positive tumors also received tamoxifen after completion
chemotherapy with chlorambucil, methotrexate and 5FU of the chemotherapy. The addition of the pseudomonas
with BCG [78]. After 10 years of follow up, the treat- vaccine failed to increase the response rate, duration of
ment arm was associated with better relapse and overall response, or survival.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 669
© Springer Science + Business Media B.V. 2009
670 Biological therapy of breast cancer

Levamisole Immunoactivation/Absorption/
The antihelminth levamisole exhibited limited benefit Ultrafiltration
as a single-agent in patients with metastatic breast cancer. Staphylococcal protein A is a polypeptide with a mole-
The agent was generally well-tolerated although granu- cular weight of 42,000 that has a high affinity for the Fc
locytopenia and agranulocytosis were observed in some portion of mammalian immunoglobulin (IgG). Preclincal
patients. Most trials focused on utilizing the agent as an studies demonstrated therapeutic efficacy in a spontane-
adjunctive therapy. In an early small randomized trial in ous canine mammary carcinoma 2and a chemically
patients with inoperable stage III breast cancer who had induced mammary carcinoma of rats) following the
been treated with radiation therapy, there was a better exposure of plasma to staphylococcal protein A [37, 70, 86].
median DFS (25 vs. 9 months) and OS at 30 months It was postulated that the removal of immunosuppres-
(90% vs. 35%) for 20 patients who received adjuvant sive immune complexes was enabling an endogenous
levamisole compared to 23 patient who did not [72]. anti-tumor response. In a highly publicized pilot study,
In an adjuvant study conducted before adjuvant che- four of five patients with advanced breast cancer exhib-
motherapy and/or hormonal therapy had been estab- ited significant anti-tumor effects following repeated
lished as standard therapy, 72 post-surgery stage II exposure of plasma to protein A imbedded in a colloidal
breast cancer patients were randomized to receive charcoal mixture [87]. Unfortunately several attempts to
levamisole or placebo [43]. After 5 years of follow up, confirm the anti-tumor effects results were unsuccessful
there was a trend for survival advantage in the levami- [29, 60]. It was eventually determined that the anti-
sole arm, primarily because of an apparent benefit in tumor effects were the result of elution of the Staph A
post-menopausal patients. protein and possibly other enterotoxins off the column
During 1976 to 1978 101 patients with advanced breast [59, 85]. In the blood stream this protein acted as a
cancer were randomized to receive doxorubicin, vincristine “super antigen”, and induced a cytokine cascade associ-
and cyclophosphamide with either placebo or levamisole ated with fever, chills, hypotension, and other systemic
2.5 mg/kg given on 2 consecutive days every week except symptoms in patients.
on the days chemotherapy was given [44]. There was a The Prosorba® immunoadsorbent column consisting of
higher response rate in patients treated with levamisole Staphylococcal aureus protein A bound covalently to an
(63% vs. 47%) and after 10 years of follow up, they had inert silica matrix to remove immune complexes, received
experienced longer survival. regulatory approval for the treatment of immune-mediated
In another trial conducted in metastatic breast cancer, 97 thrombocytopenia (IMRE Corporation, Seattle, WA). In
women were randomized to receive cyclophosphamide, one trial that enrolled 114 patients with various tumor
doxorubicin, and 5-fluorouracil chemotherapy with pla- types, objective partial responses were reported for 5/22
cebo or levamisole, 2.5 mg/kg, 2 days of each week [16]. patients with measurable metastatic breast cancer [51].
The patient population was limited to those with only bone The final commercial product appeared to overcome the
metastases or local chest wall recurrence. There was no problems with elution of the protein A off the column, but
significant difference in response rate or survival. this also appeared to remove clinical activity since there
was no evidence of anti-tumor effects in another clinical
trial conducted in 16 patients with metastatic breast
Poly A:U cancer [27].
Poly A:U, an interferon inducer and stimulator of natural A related approach, utilized dialysis filters to ultrafil-
killer cytotoxicity, has been evaluated in a randomized trate plasma to remove suppressive factors or induce
adjuvant study (not incorporating chemotherapy) in immune activation, which appeared to induce tumor
patients with stage II carcinoma of the breast, all of whom regression [47]. As was the case with the Staph A col-
had previously undergone mastectomy [42]. Patients umns, the patients who had tumor responses were the
receiving poly A:U had a better overall survival than same ones who developed symptoms consistent with a
those not receiving this polynucleotide. At the time this cytokine-release syndrome. An effort to confirm the ben-
benefit was comparable to other studies using adjuvant efits of this approach by other investigators was aborted
chemotherapy in breast cancer. Benefit was essentially by the proponents of the procedure after no responses
limited to patients with one to three involved lymph were seen in seven patients (R.O. Dillman, personal
nodes and could be correlated with natural killer cyto- observation 1990). The clinical reactions experienced by
toxicity augmentation and 2′,5′-A synthetase production these patients suggested that impurities eluted off the
(an indicator of interferon induction). dialysis filters were responsible for the severe immune
Robert O. Dillman 671

reactions that were producing anti-tumor effects in some lowest response rate and lowest median survival, perhaps
patients. because more patients discontinued treatment because
of the various toxicities that accompanied administration
of interferon.
Cytokines Since there were over 12 IFN-α molecules in the
human-derived leukocyte interferon preparation, and at
Interferon least eight in the lymphoblastoid product, but only one in
When clinical trials with interferon began, there was each recombinant IFN-α product, it is possible that other
great optimism that these natural proteins would be clin- molecules present in the natural products are important
ically useful in most malignancies [7]. As summarized for anti-tumor effect. However, an analysis of many
in Table 1, several early trials suggested interferon- reported studies suggests that that IFN-α has negligible
alpha (IFN-α) had significant anti-tumor activity against activity in breast cancer [61].
breast cancer. Early studies investigating leukocyte-
derived IFN-α reported response rates of 20% to 40%
[8, 35]. However, a subsequent study using human lym-
Interleukin-2
phoblastoid interferon documented responses in only Because of the recognized efficacy of chemotherapy and
1/29 evaluable patients [46]. A trial involving 32 women hormonal therapy, and the substantial toxicity of IL-2, few
with recurrent loco-regional breast cancer randomized patients with breast cancer were included in early clinical
patients to observation or recombinant human IFN-α at trials of IL-2 [24, 74]. As shown in Table 1, no anti-tumor
a dose of 3 MU s.c. daily for 1 year after local treatment responses were seen with these high dose regimens. Lower
[28]. There were no differences in the rates of new or doses of IL-2 have been explored in combination with
recurrent breast cancer, but there was a high incidence other therapy. Following high-dose chemotherapy with
of side effects. Homogenates of breast cancer cells autologous peripheral blood stem cell rescue, 72 women
exposed to interferon express increased levels of estro- with high-risk stage II or III breast cancer were random-
gen receptor protein. In a trial for postmenopausal ized to receive either a low dose of IL-2 (1 MU/m2) s.c.
patients with advanced breast cancer that was not known daily for 28 days or combined cyclosporine A at 1.25 mg/
to be hormone receptor negative, 99 women were ran- kg i.v. daily from day 0 to +28 and interferon gamma
domized to receive tamoxifen alone, or with IFN-α2a 3 0.025 mg/m2 s.c. every 2 days from day +7 to +28 [88].
MU i.m. thrice weekly, or daily oral 13-cis-retinoic acid Both treatments were well tolerated. At a median follow-
[19]. Response rates did not differ significantly, and up of 67 months, there were no significant differences in
ranged from 38% to 44%. After 8 years median follow- progression free or overall survival. The combination of
up, there was no significant difference in overall sur- high dose continuous infusion IL-2+ IFN-α yielded an
vival. In terms of trends, the interferon arm had the objective response in 4/23 patients [62].

Table 1. Single-agent activity of various biologicals in patients with metastatic breast cancer
Modality class Biological agent Reference Patients Response rate
Non specific Staph A protein column Terman 1981 5 80%
Non-specific Staph A protein column Messerschmidt 1988 22 23%
Non specific Staph A protein column Fennelly 1995 16 0%
Cytokine Interferon-α Gutterman 1980 16 44%
Cytokine Interferon-α Borden 1982 23 22%
Cytokine Lymphoblastoid Interferon Laszlo 1986 29 3%
Cytokine Interleukin-2 ± LAK Dillman 1993 7 0%
Cytokine Interleukin-2 ± LAK Rosenberg 1989 6 0%
Mab Trastuzumab Burris 2004 52 33%
Mab Trastuzumab Vogel 2002 114 26%
Mab Trastuzumab Baselga 2005 105 19%
Mab Trastuzumab Cobleigh 1999 222 15%
Mab Trastuzumab Baselga 1996 43 12%
Mab Bevacizumab Cobleigh 75 9%
Retinoid Bexarotene Esteva-2003 95 6%

Mab = monoclonal antibody.


LAK = lymphokine activated killer cells.
672 Biological therapy of breast cancer

Retinoids 250 mg i.v. followed by weekly doses of 100 mg for 9


additional weeks yielded five responses among 43 eval-
Cis-retinoic Acid uable patients [5]. The largest trial of trastuzumab as a
single-agent was conducted in 222 patients with Her2-
In a trial for postmenopausal women with advanced
positive metastatic breast cancer that had recurred after
breast cancer that was not known to be hormone receptor
chemotherapy [20]. Trastuzumab alone was given at an
negative, 99 patients were randomized to receive tamox-
initial dose of 4mg/kg followed by weekly doses of
ifen alone, or with s.c. IFN-α with 13-cis-retinoic acid
2 mg/kg. There were eight complete and 26 partial
1 mg/kg p.o. daily [19]. After 8 years median follow-up,
responses for a response rate of 15% with a median
response rates and overall survival were similar for the
duration of response of 13 months. Burris et al. gave
tamoxifen control arm and the retinoid containing arm.
trastuzumab at twice the standard doses by the weekly
schedule as initial treatment for 8 weeks for patients
Bexarotene with Her2-positive metastatic breast cancer who had
Bexarotene is a retinoid X receptor-selective retinoid never received chemotherapy [9]. They reported a
that exhibited anti-tumor activity against breast cancer in response rate of 33% among 52 evaluable patients in a
preclinical testing. A multicenter phase II trial random- trial that enrolled 61 patients. This response rate is the
ized 145 women with metastatic breast cancer to oral highest reported for single-agent trastuzumab. However,
bexarotene at a doses of 200 or 500 mg/m2 per day or in a randomized phase II trial conducted by Vogel et al.,
placebo [26]. A response rate of 6% was observed in 48 there was no suggestion that this higher dose produced
tamoxifen-refractory patients and in 47 chemotherapy- a higher response rate [90]. In this trial there was no
refractory patients, and 3% in 51 patients with tamox- significant difference in results for an 8 mg/kg loading
ifen-resistant disease. Hypertriglyceridemia is noted in dose and 4 mg/kg per week maintenance versus the stan-
84% of patients. Other common drug-related adverse dard 4 mg/kg loading dose and 2 mg/kg per week main-
events were dry skin (34%), asthenia (30%), and head- tenance. The failure to demonstrate any difference
ache (27%). between these doses is not surprising in view of the sus-
tained serum levels of trastuzumab that were achieved
at the lower dose. In this trial trastuzumab alone as ini-
Monoclonal Antibodies tial treatment for metastatic breast cancer resulted in
seven complete and 23 partial responses among 114
Trastuzumab
patients (26%), but the response rates were 35% for
As summarized in Table 1, an extensive clinical experi- patients whose tumors were IHC3+ compared to 0% for
ence has confirmed the anti-tumor activity of the anti- tumors that were IHC2+, and 34% among patients
Her2 humanized Mab trastuzumab in patients whose whose tumors were FISH+ compared to 7% for tumors
breast cancers over express Her2. Trastuzumab is now that were FISH-. Baselga et al. tested an every 3-week
standard therapy for the treatment of patients with high- schedule of trastuzumab delivery as initial therapy for
risk or advanced breast cancer. Various clinical trials 105 patients with Her2-positive metastatic breast cancer
have confirmed the efficacy of trastuzumab, especially [6]. They observed a response rate was 19%, but it was
in combination with chemotherapy, for the treatment of 34% for patients whose tumors were IHC3+ and/or
Her-2 positive breast cancer in patients with recurrent FISH+ for Her2.
metastatic breast cancer, as the initial treatment for met- Numerous phase II trials have been reported in which
astatic breast cancer, and as adjuvant therapy. At this trastuzumab was combined with single agent chemo-
time most physicians continue trastuzumab maintenance therapy for treatment of patients with Her2-positive
indefinitely or until disease progression because the breast cancer. These doublet combinations were associ-
Her2 receptor is continually being produced by some ated with response rates between 25% and 75% with
malignant cells. median PFS of 6 to 12 months. Response rates of 50%
As shown in Table 1, as a single agent, trastuzumab or greater were reported in 17 of the 20 trials, and
alone produced objective response rates of 12–33% in appeared to be much higher than observed with chemo-
patients with Her2-positive metastatic breast cancer that therapy alone. Many of these reports found higher
had relapsed after chemotherapy eHer2 [5, 6, 9, 20, 90]. response rates in the subsets of patients who had 3+ IHC
In a phase II trial in 46 patients with metastatic breast Her2 expression by IHC and/or overexpression by
cancer in whom at least 25% of tumor cells expressed FISH, as opposed to 2+ IHC Her2 or negative FISH.
Her2 by immunohistochemistry (IHC), a schedule of The addition of trastuzumab was not associated with
Robert O. Dillman 673

any additive toxicities, other than cardiac disease in hazard ratio 0.66, p = 0.03). In consecutive phase II trials,
association with anthracyclines. weekly paclitaxel and carboplatin plus trastuzumab
Most of the single agents used in these trials were the appeared superior to an every 3-week schedule of the
taxanes paclitaxel or docetaxel, or the vinca vinorelbine. same agents with response rates of 81% versus 65%, median
In phase II trials with 23 to 40 patients, various sched- PFS 14 versus 10 months, and median OS 3.2 versus 2.3
ules of docetaxel were associated with response rates years, and was associated with less hematologic toxicity
ranging from 50% to 70% depending on prior treatment [68]. In 40 patients who had received no prior chemo-
[2, 26, 57, 76, 84]. In phase II trials with 25 to 88 therapy the combination of paclitaxel, gemcitabine and
patients, various schedules of paclitaxel and trastu- trastuzumab produced a response rate of 52% [31]. The
zumab produced response rates ranging from 36% to same combination had a response rate of 92% in 13 previ-
62% depending on prior treatment [31, 34, 48, 71, 77]. ously untreated patients [54]. Gemcitabine, cisplatin and
In phase II trials with 30 to 62 patients, various sched- trasutuzumab produced a response rate of 40% in 20
ules of vinorelbine and trastuzumab produced response patients, but most had previously received both a taxane
rates ranging from 50% to 78% depending on prior and anthracycline [82]. Gemcitabine, vinorelbine and
treatment [4, 10, 11, 17, 24, 40, 64]. There was a 38% trastuzumab as second-line therapy produced a 50%
response rate for gemcitabine and trastuzumab among response rate in 30 patients [58].
61 patients who had previously been treated with a tax- Randomized trials have established the superiority of
ane and anthracycline [63]. Liposomal doxubicin and combining trastuzumab with chemotherapy compared
trastuzumab produced a 52% rate in 29 patients who to chemotherapy alone. One of the pivotal trials that led
had received little or no prior chemotherapy [18]. A to regulatory approval of trastuzumab compared trastu-
response rate of 24% was observed for cisplatin and zumab plus chemotherapy to chemotherapy alone in
trastuzumab in 37 patients who had received multiple 469 patients with metastatic disease who had not
prior chemotherapy regimens [66]. received prior chemotherapy for metastatic disease [80].
Many phase II clinical trials have used combination All patients had tumors that overexpressed Her2, which
chemotherapy plus trastuzumab for treatment of patients was defined as 2+ or 3+ by IHC on a scale of 0–3. An
with Her2-positive breast cancer. Most of these trials uti- initial 4 mg/kg trastuzumab dose was followed by 2 mg/kg
lized chemotherapy combinations of taxanes and plati- weekly. The first dose of Mab was infused i.v. over
num agents. Combination chemotherapy has consistently 90 min, but in the absence of significant infusion related
produced higher response rates and longer progression toxicity, subsequent doses were infused i.v. over 30 min.
free survival in clinical trials in patients with metastatic Patients who had not received an anthracycline previ-
breast cancer, although survival advantages have been ously were randomized to receive trastuzumab alone or
harder to establish. Phase II trials of combination chemo- with cyclophosphamide 600 mg/m2 i.v. and doxorubicin
therapy plus trastuzumab appear to produce slightly 60 mg/m2 or epirubicin (AC) i.v. every 3 weeks for six
higher response rates (40% to 92%) than were seen with cycles. Patients who had received adjuvant chemother-
single agents plus trastuzumab, and the PFS was some- apy that included an anthracycline were randomized to
what longer (range 6–16 months). receive paclitaxel 175 mg/m2 i.v. over 3 h alone every 3
Docetaxel, carboplatin and trastuzumab yielded a weeks, or with trastuzumab. The combination of che-
response rate of 58% in 59 patients with metastatic motherapy plus trastuzumab was superior for key end
breast cancer, 15% of whom had received prior taxane points including: response rate (50% vs. 32%, p <
therapy in the adjuvant setting [67]. Docetaxel, cisplatin 0.0001), duration of response (9.1 vs. 6.1 months), PFS
and trastuzumab produced response rate of 79% in 62 (median 7.4 vs. 4.6 months, p < 0.001), and OS (death at
previously untreated patients [67]. Docetaxel, epirubicin 1 year 22% vs. 33%, p = 0.008; median survival 25 vs.
and trastuzumab produced a response rate of 67% as ini- 20 months, p = 0.046) with a 20% risk reduction of
tial therapy for 45 patients, but the use of an anthracy- death. The differences were most striking for paclitaxel
cline was associated with an unacceptable rate of cardiac plus trastuzumab versus paclitaxel alone. The median
toxicity [89]. OS was 22 months for paclitaxel plus trastuzumab ver-
In a randomized trial, trastuzumab and paclitaxel with sus 18 months for paclitaxel alone (p = 0.17) and 27
or without carboplatin were compared as first-line therapy months for AC plus trastuzumab versus 21 months for
for 196 women with HER-2-overexpressing (2+ or 3+ by AC alone (p = 0.16).
IHC) metastatic breast cancer [71]. The addition of In another randomized trial, docetaxel plus trastu-
carboplatin produced a higher response rate 52% versus zumab was superior to docetaxel alone in patients with
36% (p = 0.04), and longer median PFS (11 vs. 7 months, metastatic breast cancer who had received no prior
674 Biological therapy of breast cancer

chemotherapy [50]. Docetaxel was given every 3 weeks of fluorouracil, epirubicin, and cyclophosphamide in both
and trastuzumab was given weekly. The addition of tras- groups, with a secondary post-chemotherapy random-
tuzumab roughly doubled the response rate and PFS, and ization for the 232 patients whose tumors were Her2-
yielded a better OS 31 versus 23 months (p = 0.032). positive to either observation or trastuzumab for 9 weeks
Three large randomized trials have confirmed the [41]. The 3-year DFS was better for patients who received
expected advantage of combining trastuzumab with che- trastuzumab (91% vs. 86%, p = 0.005) with a 42%
motherapy in the adjuvant treatment of patients with reduction in the risk of recurrence.
Her2 positive breast cancer [41, 69, 73, 81]. Two U.S. Based on Her2 biology, it is probable that there is
cooperative group trials were combined for one prelimi- more of an advantage to give trastuzumab during, rather
nary analysis [73]. The National Surgical Adjuvant than only after chemotherapy, but this has not yet been
Breast and Bowel Project (NSABP) trial B-31 compared established in trials. It is also probable that there is an
doxorubicin and cyclophosphamide followed by pacli- advantage for giving trastuzumab indefinitely, even if it
taxel every 3 weeks with the same regimen plus 52 weeks has already been given with chemotherapy, but this also
of trastuzumab beginning with the first dose of pacli- has not been proven. Finally, completed trials have
taxel. The North Central Cancer Treatment Group attempted to address whether it is better to give adjuvant
(NCCTG) trial N9831 compared three regimens: doxo- trastuzumab for 2 years or 1 year, but it is probable that
rubicin and cyclophosphamide followed by weekly it is better to give trastuzumab indefinitely if there is
paclitaxel, the same regimen plus 52 weeks of trastu- reason to believe that the breast cancer has not been
zumab initiated concomitantly with paclitaxel, and the completely eliminated.
same chemotherapy regimen followed by 52 weeks of Trastuzumab is also being combined with chemother-
trastuzumab after paclitaxel. For an early analysis at a apy as part of neo-adjuvant therapies with pathologic
median follow up of 2 years, the two similar arms from complete response rates ranging from 13% to 65% [11,
each trial were combined and compared: doxorubicin 13, 14, 22, 39, 56]. After 3 weeks of neoadjuvant treat-
plus cyclophosphamide followed by paclitaxel with or ment with trastuzumab alone, 23% of patients had a par-
without trastuzumab concurrent with paclitaxel [73]. At tial response before going on to receive 12 more weeks
a median follow up of 3 years, DFS was superior in the of trastuzumab combined with paclitaxel before surgery
there trastuzumab arm (87% vs. 75%, p < 0.0001) with a [56]. One randomized trial, which was designed to
52% reduction in the risk of disease recurrence. The accrue 164 patients, was closed early because of the
3-year OS also favored the use of trastuzumab (94% vs. marked superiority of the trastuzumab-containing arm
92%, p = 0.015). Because of the use of anthracyclines in [13]. Chemotherapy consisted of four cycles of pacli-
this trial, patients have been closely monitored for car- taxel followed by four cycles of Z5FU, epirubicin, and
diac toxicity, which has been higher in the trastuzumab cyclophosphamide, with weekly trastuzumab for all 24
arms. In the NSABP B-31 trial 16% of women discontin- weeks. After 3 years of follow up, there had been no
ued trastuzumab therapy due to clinical evidence of recurrences in the patients who received neoadjuvant
myocardial dysfunction or significant decline in left ven- therapy, and they have a better PFS (p = 0.041) [14].
tricular function. About half of Her2-positive patients are hormone
An international, multicenter trial (HERA) random- receptor positive which has led to interest in the interac-
ized more than 5,000 women with HER2-positive node tion “cross-talk” between Her2 and hormone receptors
positive or high-risk node negative breast cancer, com- which appears to increase resistance to hormonal thera-
pared 1 or 2 years of trastuzumab given every 3 weeks pies. There is a recent report of a 26% response rate for 31
with observation after completion of locoregional ther- evaluable patients who were treated with the aromatase
apy and at least four cycles of neoadjuvant or adjuvant inhibitor letrozole in combination with weekly trastu-
chemotherapy (Piccart-Gebhart et al. 2007) [81]. Initial zumab [49]. Her2 positivity was confirmed for 82%
reports after 2 years of follow up indicated that 1-year of (IHC3+ and/or FISH+) and 82% had previously been
trastuzumab conveys an advantage over observation treated with tamoxifen. The median PFS was 6 months.
with a 46% reduction in disease recurrence (p < 0.0001) The anti-vascular endothelial growth factor Mab bev-
(Piccart-Gebhart et al. 2007), and a 34% reduction in acizumab has been approved for the treatment of breast
death (p < 0.0001) [81]. cancer in combination with chemotherapy. As shown in
In a European trial 1,010 women with node-positive Table 1, bevacizumab had demonstrable, but limited
or high risk node-negative breast cancer were random- activity as a single agent in breast cancer [21]. In that
ized to adjuvant therapy that consisted of three cycles of phase I/II trial, cohorts of patients received 3, 10 or
either docetaxel or vinorelbine, followed by three cycles 20 mg/kg of bevacizumab i.v. every other week. However,
Robert O. Dillman 675

as a third-line treatment for patients with metastatic sTn(c)-KLH conjugate containing 30, 10, 3, or 1 μg of
breast cancer, the addition of bevacizumab to capecit- Tn(c) plus 100 micrograms of QS-21 [33]. All patients
abine produced a higher response rate compared to developed significant IgM and IgG antibody titers
capecitabine alone (19% vs. 9%, p = 0.001), although it against sTn(c).
did not increase DFS or OS [55]. In subsequent trial the
addition of bevacizumab to paclitaxel as initial therapy
for metastatic disease, produced higher response rates
(37% vs. 21%) and doubled the PFS from 6 to 12 months References
compared to paclitaxel alone [53]. 1. Aisner J, Weinberg V, Perloff M, et al. Chemotherapy versus
Many other antibody products are in development, chemoimmunotherapy (CAF v CAFVP v CMF each +/− MER) for
metastatic carcinoma of the breast: a CALGB study. Cancer and
but none have been approved by regulatory authorities Leukemia Group B. J Clin Oncol 1987; 5:1523–1533.
for standard treatment of breast cancer. These are 2. Arnould L, Gelly M, Penault-Llorca F, et al. Trastuzumab-based
described elsewhere in this text in other chapters that treatment of HER2-positive breast cancer: an antibody-dependent
cover monoclonal antibodies, radioimmunotherapy and cellular cytotoxicity mechanism? Br J Cancer 2006; 94:259–267.
3. Austin FC, Boone CW, Levin DL, et al. Breast cancer skin test
immunotoxins.
antigens of increased sensitivity prepared from vesicular stomatitis
virus-infected tumor cells. Cancer 1982; 49:2034–2042.
4. Bartsch R, Wenzel C, Altorjai G, et al. Results from an observa-
Vaccines tional trial with oral vinorelbine and trastuzumab in advanced
Because of the importance of HER-2 as a target, a vaccine breast cancer. Breast Cancer Res Treat 2007; 102:375–381.
5. Baselga J, Tripathy D, Mendelsohn J, et al. Phase II study of
was formulated with peptides 15–18 amino acids in
weekly intravenous recombinant humanized anti-p185HER2 mono-
length derived from the HER-2/neu domains and clonal antibody in patients with HER2/neu-overexpressing meta-
admixed with GM-CSF as an adjuvant [25]. Patients static breast cancer. J Clin Oncol 1996; 14:737–744.
with breast or ovarian cancer underwent i.d. immunization 6. Baselga J, Carbonell X, Castaneda-Soto NJ, et al. Phase II study of
once a month for a total of two to six immunizations. All efficacy, safety, and pharmacokinetics of trastuzumab monotherapy
administered on a 3-weekly schedule. J Clin Oncol 2005; 23:
eight patients immunized with HER-2/neu peptides 2162–2171.
developed HER-2/neu peptide-specific T-cell responses 7. Bonnem EM, Spiegel RJ. Interferon-alpha: current status and
and six also developed HER-2/neu protein-specific future promise. J Biol Response Mod 1984; 3:580–598.
responses. 8. Borden EC, Holland JF, Dao TL, et al. Leukocyte-derived inter-
BA7072 is a recombinant fusion protein consisting of feron (alpha) in human breast carcinoma. The American Cancer
Society phase II trial. Ann Intern Med 1982; 97:1–6.
sequences from intracellular and extracellular domains 9. Burris H 3rd, Yardle D, Jones S, et al. Phase II trial of trastuzumab
of HER-2 linked to GM-CSF. Lapuleucel-T (APC8024) followed by weekly paclitaxel/carboplatin as first-line treatment
is product produced by Dendreon Inc. that consists of for patients with metastatic breast cancer. J Clin Oncol 2004;
patient-specific peripheral-blood mononuclear cells 22:1621–1629.
10. Burstein HJ, Harris LN, Marcom PK, et al. Trastuzumab and
obtained by leukapheresis and then activated in vitro
vinorelbine as first-line therapy for HER2-overexpressing meta-
with BA7072. In an exploratory trial, 18 patients with static breast cancer: multicenter phase II trial with clinical out-
HER-2 positive breast cancer underwent leukapheresis comes, analysis of serum tumor markers as predictive factors,
and subsequent lapuleucel-T infusion 2 days later every and cardiac surveillance algorithm. J Clin Oncol 2003a; 21:
2 weeks for at least three doses [65]. Laboratory assays 2889–2895.
11. Burstein HJ, Harris LN, Gelman R, et al. Preoperative therapy
demonstrated cellular immune responses specific for
with trastuzumab and paclitaxel followed by sequential adjuvant
BA7072 and HER-2 sequences. One patient had a par- doxorubicin/cyclophosphamide for HER2 overexpressing stage
tial response that persisted for 6 months. II or III breast cancer: a pilot study. J Clin Oncol 2003b;
MUC1 is a tumor-associated antigen that is expressed 21:46–53.
on many adenocarcinomas including breast cancer. 12. Buzaid AC, Blumenschein GR, Hortovagyi GN. Adjuvant chemo-
therapy with 5-fluorouracil doxorubicin (Adriamycin) and cyclo-
Sialyl-Tn is a carbohydrate associated with MUC1. phosphamide, with or without BCG immunotherapy in stage II or
Theratope® is a synthetic Sialyl-Tn antigen produced by stage III breast cancer. In: Terry WD, Rosenberg SA, eds.
Biomira, Inc. Most of the clinical trials with this product Immunotherapy of human cancer. New York: Elsevier North
have been conducted in breast cancer patients [36]. A Holland, 1982:175–181.
related formulation combined clustered sTn conjugated 13. Buzdar AU, Ibrahim NK, Francis D, et al. Significantly higher
pathologic complete remission rate after neoadjuvant therapy
to keyhole limpit hemocyanin (KLH) (sTn(c)-KLH) with trastuzumab, paclitaxel, and epirubicin chemotherapy:
combined with the adjuvant QS-21. sTn(c)-KLH was results of a randomized trial in human epidermal growth factor
tested in 27 patients with high risk breast cancer who receptor 2-positive operable breast cancer. J Clin Oncol 2005;
received 1, 3, 10, or 30 μg. Immunizations consisted of 23:3676–3685.
676 Biological therapy of breast cancer

14. Buzdar AU, Valero V, Ibrahim NK, et al. Neoadjuvant therapy with 29. Fer MF, Oldham RK. An immunoadsorption/immunoactivation: a
paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophos- critical review. Immune complexes and human cancer. New York:
phamide chemotherapy and concurrent trastuzumab in human epi- Plenum, 1985:257–276.
dermal growth factor receptor 2-positive operable breast cancer: 30. Fisher B, Brown A, Wolmark N, et al. Evaluation of the worth of
an update of the initial randomized study population and data of corynebacterium parvum in conjunction with chemotherapy as
additional patients treated with the same regimen. Clin Cancer Res adjuvant treatment for primary breast cancer. Eight-year results
2007; 13:228–233. from the National Surgical Adjuvant Breast and Bowel Project
15. Cannon B, McCoy JL, Connor RJ, et al. Use of the leukocyte B-10. Cancer 1990; 66:220–227.
migration inhibition assay to evaluate antigenic differences in 31. Fountzilas G, Tsavdaridis D, Kalogera-Fountzila A, et al. Weekly
human breast cancers and melanomas. J Natl Cancer Inst 1978; paclitaxel as first-line chemotherapy and trastuzumab in patients
60:969–978. with advanced breast cancer. A Hellenic Cooperative Oncology
16. Carpenter JT Jr, Smalley RV, Raney M, et al. Ineffectiveness of Group phase II study. Ann Oncol 2001; 12:1545–1551.
levamisole in prolonging remission or survival of women treated 32. Fujisawa T, Waldman SR, Yonemoto RH. Leukocyte adherence
with cyclophosphamide, doxorubicin, and 5-fluorouracil for good- inhibition by soluble tumor antigens in breast cancer patients.
risk metastatic breast carcinoma: a Southeastern Cancer Study Cancer 1977; 39:506–513.
Group Trial. Cancer Treat Rep 1986; 70:1073–1079. 33. Gilewski TA, Ragupathi G, Dickler M, et al. Immunization of
17. Chan A, Martin M, Untch M, et al. Vinorelbine plus trastuzumab high-risk breast cancer patients with clustered sTn-KLH conjugate
combination as first-line therapy for HER 2-positive metastatic plus the immunologic adjuvant QS-21. Clin Cancer Res 2007;
breast cancer patients: an international phase II trial. Br J Cancer 13:2977–2985.
2006; 95:788–793. 34. Gori S, Colozza M, Mosconi AM, et al. Phase II study of weekly
18. Chia S, Clemons M, Martin LA, et al. Pegylated liposomal doxo- paclitaxel and trastuzumab in anthracycline- and taxane-pretreated
rubicin and trastuzumab in HER-2 overexpressing metastatic patients with HER2-overexpressing metastatic breast cancer. Br J
breast cancer: a multicenter phase II trial. J Clin Oncol 2006; Cancer 2004; 90:36–40.
24:2773–2778. 35. Gutterman JU, Blumenschein GR, Alexanian R, et al. Leukocyte
19. Chiesa MD, Passalacqua R, Michiara M, et al. Tamoxifen vs interferon-induced tumor regression in human metastatic breast
tamoxifen plus 13-cis-retinoic acid vs tamoxifen plus interferon cancer, multiple myeloma, and malignant lymphoma. Ann Intern
alpha-2a as first-line endocrine treatments in advanced breast can- Med 1980; 93:399–406.
cer: updated results of a phase II, prospective, randomised multi- 36. Holmberg LA, Sandmaier BM. Vaccination with Theratope (STn-
centre trial. Acta Biomed 2007; 78:204–209. KLH) as treatment for breast cancer. Expert Rev Vaccines 2004;
20. Cobleigh MA, Vogel CL, Tripathy D, et al. Multinational study of the 3:655–663.
efficacy and safety of humanized anti-HER2 monoclonal antibody in 37. Holohan TV, Phillips TM, Bowles C, Deisseroth A. Regression of
women who have HER2-overexperessing metastatic breast cancer canine mammary carcinoma after immunoadsorption therapy.
that has progressed after chemotherapy for metastatic disease. J Clin Cancer Res 1982; 42:3663–3668.
Oncol 1999; 17:2639–2648. 38. Hortobagyi GN, Buzdar AU, Frye D, et al. Combined antiestrogen
21. Cobleigh MA, Langmuir VK, Sledge GW, et al. A phase I/II dose- and cytotoxic therapy with pseudomonas vaccine immunotherapy
escalation trial of bevacizumab in previously treated metastatic for metastatic breast cancer. A prospective, randomized trial.
breast cancer. Semin Oncol 2003; 30(5 Suppl 16):117–124. Cancer 1987; 60:2596–2604.
22. Coudert BP, Largillier R, Arnould L, et al. Multicenter phase II 39. Hurley J, Doliny P, Reis I, et al. Docetaxel, cisplatin, and trastu-
trial of neoadjuvant therapy with trastuzumab, docetaxel, and car- zumab as primary systemic therapy for human epidermal growth
boplatin for human epidermal growth factor receptor-2-overex- factor receptor 2-positive locally advanced breast cancer. J Clin
pressing stage II or III breast cancer: results of the GETN(A)-1 Oncol 2006; 24:1831–1838.
trial. J Clin Oncol 2007; 25:2678–2684. 40. Jahanzeb M, Mortimer JE, Yunus F, et al. Phase II trial of weekly
23. De Maio E, Pacilio C, Gravina A, et al. Vinorelbine plus 3-weekly vinorelbine and trastuzumab as first-line therapy in patients with
trastuzumab in metastatic breast cancer: a single-centre phase 2 HER2(+) metastatic breast cancer. Oncologist 2002; 7:410–417.
trial. BMC Cancer 2007; 7:50. 41. Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant doc-
24. Dillman RO, Church C, Oldham RK, et al. Inpatient continuous- etaxel or vinorelbine with or without trastuzumab for breast can-
infusion interleukin-2 in 788 patients with cancer. The National cer. N Engl J Med 2006; 354:809–820.
Biotherapy Study Group experience. Cancer 1993; 71:2358–2370. 42. Lacour J, Lacour F, Spira A, et al. Adjuvant treatment with poly-
25. Disis ML, Grabstein KH, Sleath PR, Cheever MA. Generation of adenylic-polyuridylic acid in operable breast cancer: updated
immunity to the HER-2/neu oncogenic protein in patients with results of a randomised trial. Br Med J (Clin Res Ed) 1984;
breast and ovarian cancer using a peptide-based vaccine. Clin 288:589–592.
Cancer Res 1999; 5:1289–1297. 43. Klefstrom P, Holsti P, Grohn P, et al. Levamisole in the treatment
26. Esteva FJ, Valero V, Booser D, et al. Phase II study of weekly of stage II breast cancer. Five-year follow-up of a randomized
docetaxel and trastuzumab for patients with HER-2-overexpressing double-blind study. Cancer. 1985; 55:2753–2757.
metastatic breast cancer. J Clin Oncol 2002; 20:1800–1808. 44. Klefstrom P, Nuortio L. Levamisole in the treatment of advanced
27. Fennelly DW, Norton L, Sznol M, Hakes TB. A phase II trial of breast cancer. A ten-year follow-up of a randomized study. Acta
extracorporeal plasma immunoadsorption of patient plasma with Oncol 1991; 30:347–352.
PROSORBA columns for treating metastatic breast cancer. Cancer 45. Krown SE, Pinsky CM, Wanebo HJ, et al. Immunologic reactivity
1995; 75:2099–2102. and prognosis in breast cancer. Cancer 1980; 46:1746–1752.
28. Fentiman IS, Balkwill FR, Cuzick J, et al. A trial of human alpha 46. Laszlo J, Hood L, Cox E, Goodwin B. A randomized trial of low
interferon as an adjuvant agent in breast cancer after loco-regional doses of alpha interferon in patients with breast cancer. J Biol
recurrence. Eur J Surg Oncol 1987; 13:425–428. Response Mod 1986; 5:206–210.
Robert O. Dillman 677

47. Lentz MR. Continuous whole blood UltraPheresis procedure in 65. Park JW, Melisko ME, Esserman LJ, et al. Treatment with autolo-
patients with metastatic cancer. J Biol Response Mod 1989; gous antigen-presenting cells activated with the HER-2 based anti-
8:511–527. gen Lapuleucel-T: results of a phase I study in immunologic and
48. Leyland-Jones B, Gelmon K, Ayoub JP, et al. Pharmacokinetics, clinical activity in HER-2 overexpressing breast cancer. J Clin
safety, and efficacy of trastuzumab administered every three weeks Oncol 2007; 25:3680–3687.
in combination with paclitaxel. J Clin Oncol 2003; 21:3965–3971. 66. Pegram MD, Lipton A, Hayes DF, et al. Phase II study of receptor-
49. Marcom PK, Isaacs C, Harris L, et al. The combination of letrozole enhanced chemosensitivity using recombinant humanized anti-
and trastuzumab as first or second-line biological therapy produces p185HER2/neu monoclonal antibody plus cisplatin in patients
durable responses in a subset of HER2 positive and ER positive with HER2/neu-overexpressing metastatic breast cancer refractory
advanced breast cancers. Breast Cancer Res Treat 2007; to chemotherapy treatment. J Clin Oncol 1998; 16:2659–2671.
102:43–49. 67. Pegram MD, Pienkowski T, Northfelt DW, et al. Results of two
50. Marty M, Cognetti F, Maraninchi D, et al. Randomized phase II trial open-label, multicenter phase II studies of docetaxel, platinum
of the efficacy and safety of trastuzumab combined with docetaxel salts, and trastuzumab in HER2-positive advanced breast cancer. J
in patients with human epidermal growth factor receptor 2-positive Natl Cancer Inst 2004; 96:759–769.
metastatic breast cancer administered as first-line treatment: the 68. Perez Ea, Suman VJ, Rowland KM, et al. Two concurrent phase II
M77001 study group. J Clin Oncol 2005; 23:4265–4274. trials of paclitaxel/carboplatin/trastuzumab (weekly or every-3-
51. Messerschmidt GL, Henry DH, Snyder HW Jr, et al. Protein A week schedule) as first-line therapy in women with HER2-
immunoadsorption in the treatment of malignant disease. J Clin overexpressing metastatic breast cancer: NCCTG study 983252.
Oncol 1988; 6:203–212. Clin Breast Cancer 2005; 6:425–432.
52. McCulloch PB, Poon M, Dent PB, Dawson P. A stratified random- 69. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab
ized trial of 5-flurouracil, doxorubicin (Adriamycin) and cyclo- after adjuvant chemotherapy in HER2-positive breast cancer.
phosphamide alone or with BCG in stage IV breast cancer. In: N Engl J Med 2005; 353:1659–1672.
Terry WD, Rosenberg SA, eds. Immunotherapy of human cancer. 70. Ray PK, Raychaudhuri S, Allen P. Mechanism of regression of
New York: Elsevier, 1982:183–186. mammary adenocarcinomas in rats following plasma adsorption
53. Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab over protein A-containing Staphylococcus aureus. Cancer Res
versus paclitaxel alone for metastatic breast cancer. N Engl J Med 1982; 42:4970–4974.
2007; 357:2666–2676. 71. Robert N, Leyland-Jones B, Asmar L, et al. Randomized phase III
54. Miller KD, Sisk J, Ansari R, et al. Gemcitabine, paclitaxel, and study of trastuzumab, paclitaxel, and carboplatin compared with
trastuzumab in metastatic breast cancer. Oncology 2001; 15(2 trastuzumab and paclitaxel in women with HER-2-overexpressing
Suppl 3):38–40. metastatic breast cancer. J Clin Oncol 2006; 24:2786–2792.
55. Miller KD, Chap LI, Holmes FA, et al. Randomized phase III trial 72. Rojas AF, Feierstein JN, Mickiewicz E, et al. Levamisole in
of capecitabine compared with bevacizumab plus capecitabine in advanced human breast cancer. Lancet 1976; 1:211–215.
patients with previously treated metastatic breast cancer. J Clin 73. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant
Oncol 2005; 23:792–799. chemotherapy for operable HER2-positive breast cancer. N Engl J
56. Mohsin Sk, Weiss HL, Gutierrez MC, et al. Neoadjuvant trastu- Med 2005; 353:1673–1684.
zumab induces apoptosis in primary breast cancers. J Clin Oncol 74. Rosenberg SA, Lotze MT, Yang JC, et al. Experience with the use
2005; 23:2460–2468. of high-dose interleukin-2 in the treatment of 652 patients. Ann
57. Montemurro F, Choa G, Faggiuolo R, et al. Safety and activity of Surg 1989; 210:474–485.
docetaxel and trastuzumab in HER2 overexpressing metastatic breast 75. Rudczynski AB, Dyer CA, Mortensen RF. Detection of cell-mediated
cancer: a pilot phase II study. Am J Clin Oncol 2003; 26:95–97. immune reactivity of breast cancer patients by the leukocyte adher-
58. Morabito A, Longo R, Gattuso D, et al. Trastuzumab in combina- ence inhibition response to MCF-7 extracts. Cancer Res 1978;
tion with gemcitabine and vinorelbine as second-line therapy for 38:3590–3594.
HER-2/neu overexpressing metastatic breast cancer. Oncol Rep 76. Sato N, Sano M, Tabei T, et al. Combination docetaxel and trastu-
2006; 16:393–398. zumab treatment for patients with HER-2-overexpressing metastatic
59. Murphy RM, Colton CK, Yarmush ML, et al. Staphylococcal pro- breast cancer: a multicenter, phase-II study. Breast Cancer 2006;
tein A adsorption in neoplastic disease: analysis of physicochemi- 13:166–171.
cal aspects. Mol Biother 1989; 1:186–206. 77. Seidman AD, Fornier MN, Esteva FJ, et al. Weekly trastuzumab
60. Oldham R. Symposium on ex vivo plasma immunoabsorption and and paclitaxel therapy for metastatic breast cancer with analysis of
protein A in cancer therapy. J Biol Resp Modif 1984; 3:229–230. efficacy by HER2 immunophenotype and gene amplification. J
61. Oldham RK. Biologicals for cancer treatment: interferons. Hosp Clin Oncol 2001; 19:2587–2595.
Pract 1985; 20:72–91. 78. Senn HJ, Barett-Mahler AR, Jungi WF, et al. Adjuvant chemoim-
62. Oldham RK, Blumenschein G, Schwartzberg L, et al. Combination munotherapy with Leukeran, Methotrexate and 5FU (LMF) +
chemotherapy utilizing interleukin-2 and alpha interferon in BCG in node-negative and node-positive breast cancer patients: 10
patients with advanced cancer. Mol Biother 1991; 3:68–73. year results. Eur J Cancer Clin Oncol 1989; 25:513–525.
63. O’Shaughnessy JA, Vukelja S, Marsland T, et al. Phase II study of 79. Serrou B, Sancho-Garnier H, Cappelaere P. Inefficacy of post
trastuzumab plus gemcitabine in chemotherapy-pretreated patients radiotherapeutic BCG immunotherapy in T3-T4 breast cancer
with metastatic breast cancer. Clin Breast Cancer 2004; 5:142–147. patients: a randomized trial. In: Terry WD, Rosenberg SA, eds.
64. Papaldo P, Fabi A, Ferretti G, et al. A phase II study on metastatic Immunotherapy of human cancer. New York: Elsevier, 1982:
breast cancer patients treated with weekly vinorelbine with or 195–198.
without trastuzumab according to HER2 expression: changing the 80. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy
natural history of HER2-positive disease. Ann Oncol 2006; plus a monoclonal antibody against HER2 for metastatic breast
17:630–636. cancer that overexpresses HER2. N Engl J Med 2001; 344:783–792.
678 Biological therapy of breast cancer

81. Smith I, Procter M, Gelber RD, et al. 2-year follow-up of trastu- immunohistochemical, immunologic, and serologic findings.
zumab after adjuvant chemotherapy in HER2-positive breast can- J Immunol 1980; 124:795–805.
cer: a randomised controlled trial. Lancet 2007; 369:29–36. 87. Terman DS, Young JB, Shearer WT, et al. Preliminary observations
82. Stemmler HJ, Kahlert S, Brudler O, et al. High efficacy of gemcit- of the effects on breast adenocarcinoma of plasma perfused over
abine and cisplatin plus trastuzumab in patients with HER2- immobilized protein A. N Engl J Med 1981; 305:1195–1200.
overexpressing metastatic breast cancer: a phase II study. Clin 88. Vahdat LT, Cohen DJ, Zipin D, et al. Randomized trial of low-dose
Oncol 2005; 17:630–635. interleukin-2 vs cyclosporine A and interferon-gamma after high-
83. Tanaka F, Yonemoto RH, Waldman SR. Blocking factors in sera of dose chemotherapy with peripheral blood progenitor support in
breast cancer patients. Cancer 1979; 43:838–847. women with high-risk primary breast cancer. Bone Marrow
84. Tedesco KL, Thor AD, Johnson DH, et al. Docetaxel combined Transplant 2007; 40:267–272.
with trastuzumab is an active regimen in HER-2 3 + overexpressing 89. Venturini M, Bighin C, Monfardini S, et al. Multicenter phase II
and fluorescent in situ hybridization-positive metastatic breast study of trastuzumab in combination with epirubicin and docetaxel
cancer: a multi-institutional phase II trial. J Clin Oncol 2004; as first-line treatment for HER2-overexpressing metastatic breast
22:1071–1077. cancer. Breast Cancer Res Treat 2006; 95:45–53.
85. Terman DS. Protein A and staphylococcal products in neoplastic 90. Vogel CL, Cobleigh MA, Tripathy D, et al. Efficacy and safety of
disease. Crit Rev Oncol Hematol 1985; 4(2):103–124. trastuzumab as a single agent in first-line treatment of HER2-
86. Terman DS, Yamamoto T, Mattioli M, et al. Extensive necrosis overexpressing metastatic breast cancer. J Clin Oncol 2002;
of spontaneous canine mammary adenocarcinoma after extra- 20:719–726.
corporeal perfusion over Staphylococcus aureus Cowans I. 91. Weese J, Oldham RK, Tormey DC. Immunologic monitoring in
Description of acute tumoricidal response: morphologic, histologic, carcinoma of the breast. Surg Gynecol Obstet 1997; 145:208–218.
21.6 Biological therapy of lung cancer
ROBERT O. DILLMAN

Non-specific Immunostimulants included two different types of cutaneous BCG therapy


or no immuotherapy [70].
Bacillus Calmette Guerin (BCG) SRL172, a preparation of heat-killed Mycobacterium
vaccae, was combined with chemotherapy to treat
Bacillus Calmette Guerin (BCG) was the first biotherapy patients with inoperable non-small cell lung cancer
tested in lung cancer in the modern era. Early randomized (NSCLC) or mesothelioma [75]. In a small study, 28
trials suggested that various forms or extracts of BCG patients were randomized to receive chemotherapy alone
could induce immune responses in patients with lung or with monthly i.d. injections of SRL172. The response
cancer [88]. However, after the reporting of numerous rate was higher for the SRL172 arm (54% versus 33%),
randomized trials that explored intrapleural, cutaneous, as was median survival (9.7 months versus 7.5 months)
and intralesional BCG, this approach was abandoned. and 1-year survival (42% versus 18%). SRL172 pro-
Intrapleural BCG: Following an initial encouraging duced mild inflammation at the injection site.
report based on 40 patients, after 4 years of follow up In 298 patients with small cell lung cancer (SCLC) a
of 169 patients with stage I lung cancer, there appeared randomized trial of induction chemotherapy and radiation
to be a survival advantage for intrapleural BCG follow- therapy with or without concurrent BCG by scarification
ing surgery [68, 69]. However, other small randomized or found no beneficial effect on response rate, duration of
trials of intrapleural BCG involving 52, 92 and 118 response, or survival [67]. A second study in 102 patients
patients respectively, found no difference in survival SCLC found no advantage for the MER form of BCG
and documented significant toxicity [56, 61, 89]. A [34]. A third randomized trial in limited stage SCLC com-
large U.S. randomized trial of 425 patients also failed pared MER to observation after four cycles of chemother-
to confirm a survival advantage for intrapleural BCG apy and again there was no difference in outcome in terms
in patients with stage I lung cancer [72]. A large of maximum response or survival [66].
European randomized trial of 407 patients also found no
advantage for intrapleural BCG, and noted a shortened
survival in patients who received BCG and underwent
Corynebacterium Parvum
pneumonectomy [58]. Another non-specific immunostimulant form of biother-
Intratumoral BCG: A randomized trial in 88 patients apy that was studied in lung cancer is Corynebacterium
with lung cancer examined the effects of pre-operative parvum (C parvum). There was no difference in response
intratumoral treatment with BCG, and found no survival rate in a small randomized trial of 49 NSCLC patients
advantage [64]. who received C parvum in combination with ifos-
Cutaneous BCG: A report of 48 patients suggested an phomide and doxorubicin chemotherapy compared to
increased progression free survival (PFS) for s.c. BCG those who received chemotherapy alone [33]. A 49
in lung cancer patients treated with radiotherapy [80]. patient randomized trial of i.v. C parvum as an adjuvant
After 2 years of follow up, a randomized trial of 500 to surgery also found no advantage for this approach
patients suggested a slight, but statistically insignificant, [111]. A randomized trial in 79 patients failed to confirm
survival advantage for patients who received i.d. BCG a positive effect on response rate or PFS in patients who
post-operatively [23]. In a randomized trial of 103 received chemotherapy with doxorubicin, cyclophosph-
patients with advanced or metastatic cancer, a methanol amide and vincristine, and methotrexate with or without
extracted residue (MER) of BCG provided no response C-parvum [96]. A large European trial randomized 286
or survival benefit beyond chemotherapy with CCNU, evaluable patients with stage I or II non-small cell lung
methotrexate, and doxorubicin [85]. In a three-arm trial cancer to no further treatment or adjuvant therapy with
in 92 surgically resected patients, investigators observed a combination of intrapleural and intravenous C parvum
the same 37% 5-year survival rate in all three-arms that [57]. There was no difference in outcome.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 679
© Springer Science + Business Media B.V. 2009
680 Biological therapy of lung cancer

Levamisole three treatment groups in terms of response rate or sur-


vival [16]. Another trial randomized 141 patients with
Levamisole alone: The antihelminth levamisole has also resected stage II and III adenocarcinoma and large-cell
been studied in lung cancer. A randomized trial in which undifferentiated carcinoma to receive postoperative
211 post-operative patients were either observed or chemotherapy (cyclophosphamide, cisplatin and doxo-
treated with levamisole found no difference in survival rubicin) or immunotherapy (intrapleural BCG and oral
[1]. In a randomized trial of 318 patients, there was a levamisole) [32]. Survival was better for the patients
15% increase risk of death associated with a regimen of who received chemotherapy.
pre and post operative levamisole compared to a placebo
group, primarily because of post surgical respiratory
failure [2]. OK-432
Levamisole and chemotherapy: A Veterans Adminis- OK-432 (Picibanil) is a lyophilized preparation of peni-
tration trial randomized 381 patients with metastatic cillin-treated and heat killed Streptococcus pyogenes that
lung cancer to cyclophosphamide and CCNU alone or has been studied for many years as an immunotherapy in
with levamisole [20]. After statistical adjustments, Japan. OK-432 has also been explored as treatment for
levamisole had a statistically significant negative effect malignant ascites and malignant pleural effusions, and
on survival. also as inhalational therapy for bronchoalveolar cell car-
Levamisole and radiation therapy: In a four-arm cinoma [74, 113]. A prospective randomized study in 93
trial in 107 patients with squamous cell NSCLC, there patients with primary lung cancer failed to demonstrate
was no advantage for levamisole or levamisole plus an immunological or survival benefit from adjuvant intra-
doxorubicin over radiation therapy alone [110]. The pleural OK-432 immunotherapy after complete resection
Southeastern Cancer Study Group randomized 251 of the primary lung tumor [49]. A recent three-arm ran-
patients with inoperable NSCLC to radiation therapy domized trial compared intrapleural OK-432, intrapleural
alone or with levamisole [46]. The trends for response bleomycin, or systemic cisplatin plus etoposide in 102
rate and local control favored radiation alone; survival patients with NSCLC who had malignant pleural effu-
was the same in both arms. The Radiation Therapy sions [114]. Pleural PFS and OS both favored the OK-432
Oncology Group (RTOG) conducted a similar trial in arm, but the differences were not statistically different.
285 patients with inoperable stage I or II or unresectable Intrapleural OK-432 as also been given in combina-
T3 N0 or N1 NSCLC [79]. Patients received radiation tion with chemotherapy as adjuvant treatment for
therapy and were randomized to either placebo or patients with resected non-small-cell lung cancer. A
twice weekly levamisole for 2 years. Levamisole failed recent meta-analysis was conducted based on data from
improve PFS or OS. RTOG also found no difference in 1,520 such patients enrolled in 11 randomized clinical
a study of 74 patients with resected stage II–III non- trials, all of which were started before 1991 in which
small cell lung cancer with positive nodes randomized standard chemotherapy was compared with the chemo-
to post-operative thoracic irradiation plus either placebo therapy plus OK-432 [92]. The 5-year survival rate for
or levamisole [31]. all eligible patients in the 11 trials was 51% in the immu-
BCG and/or C parvum and/or levamisole: Some ran- nochemotherapy group versus 44% in the chemotherapy
domized trials explored C parvum and/or levamisole group (hazard ratio 0.70, p = 0.001). Because, only four
and/or BCG simultaneously. In one trial, 76 patients of these trials actually utilized a centralized randomiza-
with stage III lung cancer were randomized to chemo- tion, an analysis of that subset of trials was analyzed and
therapy alone (cisplatin, doxorubicin, cyclophosph- also showed longer survival time for the OK-432 plus
amide and vincristine) chemotherapy plus BCG, or chemotherapy group (odds ratio = 0.66, p = 0.049).
chemotherapy plus C-parvum. Survival favored the Confirmatory trials have not been performed in the
group that received C-parvum (p = 0.05) [12]. A ran- United States or Europe.
domized trial in 100 patients with resectable lung cancer
showed no difference in post-operative survival among
intrapleural BCG, BCG plus C-Parvum, or placebo
Thymic Hormones
[112]. A total of 109 patients with advanced lung cancer A number of different thymic hormones, including crude
of various cell types, were randomized to MACC che- preparations such as Thymosin fraction V, and purified
motherapy alone (methotrexate, doxorubicin cyclophos- agents such as thymosin alpha-1, have significant effects
phamide, and CCNU), or with oral levamisole or with on T cells and cell mediated immunity that led to inves-
s.c. C parvum. There were no differences among the tigation of their immunopotientiation in lung cancer [55].
Robert O. Dillman 681

In a small randomized trial in SCCL, thymosin fraction


V plus chemotherapy did not increase the response rate
Interferon alpha
compared to chemotherapy alone, but was associated with Non Small Cell Lung Cancer: In a phase II trial in 38
a significant prolongation of survival [19]. Unfortunately, patients with measurable NSCLC, human leukocyte
a larger study of 91 patients found no advantage for the interferon, which is predominantly interferon-alpha,
addition of thymosin fraction 5 over chemotherapy alone produced no objective responses [95] Preclinical and
in terms of response rate or survival [97]. A randomized preliminary clinical data suggested IFN-α potentiates
trial involving 105 NSCLC patients found a higher the cytotoxic activity of several chemotherapy agents
response rate for patients who received VAP (vindesine, against NSCLC which provided the rationale for several
doxorubicin, cisplatin) alone compared to patients who trials of combination therapy in NSCLC. In one trial
received VAP plus thymosin fraction V, and there was a 182 patients were randomized to receive either cisplatin-
statistically insignificant trend for better survival for epidoxorubicin-cyclophosphamide (CEP) alone or with
chemotherapy alone [8]. i.m. IFN-α [3]. The response rate and toxicity were
As summarized in Table 1, dose-exploring and phase statistically higher in the combination therapy arm,
II trials failed to confirm significant anti-tumor effects but there was no difference in OS.. In a phase II trial
or enhancing activity for thymosin alpha-1 in NSCLC the 3/41 (7%) response rate and median survival of only
[21, 22]n a phase II trial in patients with advanced 6 months for patients treated with carboplatin and IFN-α
NSCLC, the combination of cisplatin and etoposide was considered disappointing [62]. In a randomized
chemotherapy combined with thymosin-alpha 1 and phase II trial a combination of ifosfamide, platinum
low-dose IFN-α2a produced a response rate of 43% and IFN-α produced a response rate that similar to
(24/56) and a median survival of 12.6 months [25]. In a the same chemotherapy alone, but was associated
small randomized trial involving 22 patients with with much greater neutropenia [82]. In a small pilot
advanced NSCLC, following ifosfamide chemotherapy study the combination of 5FU, leucovorin, and IFN-α
half of patients received the combination of thymosin resulted in responses in 7/18 (39%) of patients with
alpha 1 with interferon alpha (IFN-α) [93]. The chemo- metastatic NSCLC, but this same response rate has been
immunotherapy cohort had a higher response rate (33% reported for 5FU chemotherapy alone in patients
versus 10%) and longer PFS as well as less suppression with adenocarcinoma for the lung [82]. A phase II study
of T cell counts and less hematologic toxicity. in 100 previously untreated patients with unresectable,

Table 1. Single-agent activity of various biologicals in patients with metastatic lung cancer
Modality class Biological agent Lead author Patients Response rate (%)
Non-specific Thymosin alpha 1 [22] 10 0
Non-specific All-trans retinoic acid [104b] 28 7
Cytokine Interferon-α (IFN) [95] 38 0
Cytokine Interferon-α + isotretinoin [5] 17 0
Cytokine Interleukin-2 (IL-2) [4] 11 0
Cyokines IL-2 ± IFN-β [103] 76 3
Cytokines IL-2 + IFN-α [76] 7 0
Cytokines IL-2 + IFN-α [35] 11 0
Cytokine IL-4 [109] 55 2
Cell therapy IL-2 + LAK [9] 15 6
Immunotoxin Ricin A-Anti-CD56 [59] 21 4
Mab Anti-Epcam [24] 6 0
Mab Anti-GRP [40] 13 8
Mab Cetuximab [29] 66 5
Mab Trastuzumab [18] 24 1
Mab Trastuzumab [48] 13 0
Vaccine Belagenpumatucel-L [73] 61 15

Mab = monoclonal antibody


LAK = lymphokine activated killer cells
Epcam = epithelial cell adhesion molecule
GRP = gastrin releasing peptide
682 Biological therapy of lung cancer

measurable or evaluable stage III or IV NSCLC examined both NSCLC and SCLC, but this has been explored in
the combination of IFN-α2a and cisplatin in NSCLC only a limited manner clinically. In one study 20 patients
[39]. After an initial planned 6 months of therapy, with inoperable NSCLC, were randomly assigned to
responding patients could receive maintenance IFN-α receive either hyperfractionation radiotherapy alone or
for up to 6 more months. The overall response rate was concurrently with IFN-α 3MU i.m. and 1.5 MU inhaled
33% among 84 evaluable patients with a median survival via a dosimeter-equipped jet nebulizer 30 min before
of 6.4 months. As would be expected, better results each radiotherapy session [60]. Combined treatment
were achieved in stage III than IV patients. In another with radiotherapy was feasible but impractical. There
phase II trial 35 patients received MVP chemotherapy was no difference in response rate and some early deaths
(mitomycin C, vindesine or vinblastine, cisplatin) plus occurred in the combination arm that could have been
IFN-α2a-IFN, 3 MIU during the first week of each 28 treatment related. In a phase I study, escalating doses of
day cycle of therapy [99]. There was a tumor response IFN-α2a were combined with three different schedules
rate of 51% (18/35), median PFS of 6 months, and of cisplatin and twice-daily radiotherapy [108]. IFN-α was
median OS of 9.5 months. injected s.c. 2 h before the first daily fraction of radia-
Small Cell Lung Cancer: Following induction che- tion. The objective response rate was 46% (11/24).
motherapy with cisplatin, doxorubicin, cyclophosph- The Radiation Therapy Oncology Group (RTOG)
amide (CAP) 237 SCLC patients were randomized to conducted a trial in 123 patients with stage III NSCLC
receive no further treatment or IFN-α as maintenance in which patients were randomized to 50 Gy RT over 6
therapy [65]. Although there were more long term survi- weeks alone or with IFN-α 16 MIU by i.v. bolus every
vors among the subset of patients with limited stage dis- other day thrice weekly on weeks 1, 3, and 5 during
ease who received IFN-α, there was no difference in radiation therapy [14]. Because of toxicity, only 76%
OS. In a trial involving 215 patients with limited stage completed IFN-α, and only 82% completed radiotherapy
SCLC, 171 responded to induction chemoradiotherapy compared to 94% in the control arm. Median survival
and 140 were randomized to receive maintenance IFN-α and 1-year survival rates were similar, respectively 9.5
or no further treatment [41]. The IFN-α afforded no to 10.3 months and 42% to 44%.
improvement in duration of response or survival, and
was tolerated poorly. In a trial that included patients
with both limited and extensive stage SCLC, 90 previ-
ously untreated patients were randomly assigned to Retinoids
receive Ifosfamide, Carboplatin, and Etoposide (ICE)
alone or in combination with twice weekly low dose s.c.
Prevention Trials
IFN-α [115]. There was no significant difference in It had been proposed that analogs of vitamin A such as
overall response rates, but toxicity, complete responses arytenoids and retinoids might help prevent lung cancer
and survival were higher in the IFN-α arm (p < 0.05). and heart disease. The effects of a combination of 30 mg
Most of the impact was in patients who had limited of beta carotene per day and 25,000 IU of retinol (vita-
stage disease. In a similar trial with different induction min A) in the form of retinyl palmitate were explored in
chemotherapy 85 patients were randomized to chemo- a multicenter, randomized, double-blind, placebo-con-
therapy alone or concurrent with IFN [81]. The IFN-α trolled primary prevention trial involving 18,314 smok-
arm was associated with higher rates of complete (30% ers, former smokers, and workers exposed to asbestos
versus 15%) and partial remission (42% versus 29%), [77]. There were 388 new cases of lung cancer diag-
and longer survival (p < 0.02). In patients with exten- nosed during a mean follow-up of 4 years. Surprisingly,
sive stage SCLC, a single arm phase II trial that included the vitamin-treated group had a higher relative risk 1.28
IFN-α in both the induction and maintenance therapy (p = 0.02) of developing lung cancer compared with the
did not suggest results that were better than the histori- placebo group. There were no differences in the risks of
cal experience with the etoposide-cisplatin chemother- other types of cancer, but there was also a higher risk of
apy alone [42]. A three-arm trial in 219 patients with cardiac related mortality in the retinoid treatment group.
any stage of SCLC randomized patients to cisplatin plus The investigators concluded that the combination of
etoposide alone, or to low doses of natural or recombi- beta carotene and vitamin A had no benefit and may
nant IFN [90]. Hematologic toxicity was greater in the have had an adverse effect on the incidence of lung
interferon arms and there was no difference in survival. cancer and on the risk of death from lung cancer, cardio-
Radiation Therapy and Interferon Alpha: In vitro vascular disease, and for these reasons the study was
studies suggest that IFN-α may be a radiosensitizer for stopped early.
Robert O. Dillman 683

Beta carotene (50 mg on alternate days) alone was randomized to no treatment or retinol palmitate (300,000
tested in a similar randomized, double-blind, placebo- IU p.o. daily for 12 months) [78]. After 46 months there
controlled trial involving some 22,071 male physicians, were fewer new tumors in the retinol group (p = 0.045)
about half of whom were current or former smokers but few recurrences had taken place.
[30]. After 12 years of follow up 170 cases of lung can-
cer had been diagnosed. There were no differences in
the overall incidence of lung cancer, overall malignancy
Cis-retinoic Acid and Interferon
or cardiovascular disease, or in overall mortality. The combination of IFN-α plus CRA, which had pro-
National Cancer Institute (NCI) Intergroup phase III duced high objective response rates in squamous cell
trial (NCI #I91-0001) tested the hypothesis that retinoid cancers of the skin and cervix, produced objective
chemoprevention would decrease recurrences or second responses in only 1/21 patients with squamous cell cancer
primary tumors in the setting of stage I NSCLC [52]. The of the lung, and that response was in a patient who only
1,166 patients had pathologic stage I NSCLC and were 6 had regionally advanced disease [86]. A three-arm ran-
weeks to 3 years post potentially curative surgery without domized phase II multi-center study of 85 patients with
other adjuvant therapy. They were randomized to received SCLC revealed no differences in survival among the
placebo or 13-cisretinoic acid (isotretinoin, CRA, three arms, one of which included IFN-α during induc-
Accutane®) at a dose of 30 mg/day for 3 years. After a tion, and IFN plus CRA as maintenance therapy [91].
median follow-up of 3.5 years, there were no differences
between the placebo and isotretinoin arms, but there was
more mucocutaneous toxicity and noncompliance in the
Cis-retinoic Acid and Chemotherapy
isotretinoin arm. Secondary multivariate and subset anal- CRA has also been combined with chemotherapy.
yses suggested that isotretinoin was harmful in current Carboplatin, vindesine and 5-fluorouracil/leucovorin were
smokers, but beneficial in those who had never smoked. combined CRA 1 mg/kg orally in 28 patients with
Squamous metaplasia is frequently observed in bron- advanced, measurable NSCLC [84]. Toxicity was man-
chial biopsy samples from chronic smokers. In a small ageable. The response rate was 39% (11/28) with a median
randomized trial the synthetic retinoid etretinate was survival of 9.7 months for the whole group.
evaluated to see if could reverse such squamous meta-
plasia [50]. Following bronchoscopy 86 individuals
with significant dysplasia and/or metaplasia were ran-
Trans-retinoic Acid and Interferon
domized to receive either 1 mg/kg isotretinoin or pla- The combination of all trans-retinoic acid (ATRA) and
cebo daily for 6 months, 69 were reevaluated at the IFN-α was tested in patients with various types of non-
completion of treatment. A reduction in the metaplasia small cell lung cancer that were unresectable, locally
was noted in more than 50% of subjects in each group, advanced, or metastatic [6]. The authors reported an
but no significant change in metaplasia was found objective response rate of 21% (6/29) with several
among those who continued to smoke. responses lasting more than a year in duration. ATRA
In an exploratory trial, the frequency of abnormalities alone produced two objective response in 28 patients
in the expression of retinoic acid receptor-beta (RARbeta) with NSCLC [104b].
in bronchial cells and the ability of CRA to correct such
abnormalities, were examined in 188 smokers who under-
went bronchoscopy [7]. Bronchial brushing samples were
Transretinoic Acid and Chemotherapy
obtained for cytology analysis and for molecular analy- In a phase II study 38 patients with advanced squamous
sis. Forty-four individuals with diminished RARbeta cell NSCL were treated with a combination of low-dose
expression were randomized to receive a placebo or CRA ATRA (40 mg/m2/day), IFN-α (6 MU/day) and monthly
30 mg p.o. daily for 6 months. Only 27 patients completed cisplatin (40 mg/m2) for 12 weeks [27]. There was a
treatment and 18 underwent repeat bronchoscopy. There response rate of 21% but only 16% of patients were able
was no difference between the results of RARbeta expres- to complete the planned treatment.
sion before and after treatment in the placebo group (p = Two large randomized trials were conducted in which
0.43), but there was upregulation of RARbeta expression patients with pleural effusions or distant metastatic NSCLC
in the CRA group (p = 0.001). The authors felt that these were randomized to receive standard combination chemo-
results supported undertaking a phase III chemopreven- therapy with placebo or bexarotene capsules (Targretin®) at
tion trial of CRA treatment for lung cancer. In another a dose of 400 mg/m2 p.o. daily [106]. In both trials the
trial 307 post operative patients with stage I NSCLC were 2-year survival rate was 16% for chemotherapy alone, and
684 Biological therapy of lung cancer

12% to 13% in the chemotherapy plus bexarotene arms; so


the studies were conclusively negative in terms of any ben-
Interleukin-2 Alone,
efit for this retinoid [13, 36]. in Combination, or with
Adoptive Cellular Therapy
Interferon Gamma IL-2 in NSCLC
Randomized trials have failed to support a benefit for Eleven subjects with Stage III–IV non small cell lung
IFN-gamma in lung cancer, even in the setting of mini- cancer were treated with continuous i.v. IL-2 at a dose of
mal residual disease. 18 MIU/m2/day from day 1 to day 13 with a rest on day
7 [4]. There were no tumor responses, but immune
effects were noted. In an Italian study, 20 patients with
Non Small Cell Lung Cancer NSCLC received the pineal hormone melatonin orally at
In a small randomized phase II trial, 37 patients with a dose of 10 mg day-1 each evening starting 1 week
inoperable NSCLC were randomized to receive either before starting s.c. IL-2 at a dose of 3 MIU/m2 every 12 h
two cycles of etoposide and cisplatin chemotherapy or 6 for 5 days/week for 4 weeks [53]. The authors reported
weeks of thrice weekly IFN-β (30 MU) plus IFN-γ partial responses in 4/20 (20%), with little toxicity. In a
(200 μg) followed by two cycles of chemotherapy [98]. subsequent trial, the same group conducted a random-
There was more hematologic toxicity during chemo- ized trial in 60 patients with locally advanced n = 8) or
therapy on the combined modality arm (p = 0.02), but metastatic NSCLC (n = 52) who received either low-
no difference in response rates (11–17%) or survival. dose s.c. IL-2 plus melatonin or cisplatin plus etoposide
chemotherapy [54]. Similar response rates (all partial)
were observed (7/29 patients treated with biotherapy and
Small Cell Lung Cancer 6/31 in patients receiving biotherapy, but the survival
Of 71 patients with extensive stage SCLC, 41 had a com- rate at 1 year actually favored the biotherapy group (45%
plete or partial response after four cycles of PACE (cis- versus 19%, p = 0.02). This study is of interest because it
platin, doxorubicin, cyclophosphamide, and etoposide) suggests that a relative non toxic immunotherapy regimen
and then were treated with INF-γ 0.2 mg s.c. daily until may be as efficacious as standard chemotherapy in the
grade IV toxicities or disease progression occurred [11]. treatment of advanced NSCLC.
There was an increased tumor regression in only 2/30 In a phase II trial, 76 patients with stage IV NSCLC
(7%) patients who had a partial response at the end of were randomized to receive either IL-2 alone 18 MIU/
chemotherapy, and no suggestion of a survival benefit. m2 i.v. 3 days weekly or IL-2 plus i.v. IFN-β (6 MU/m2),
Based on this the authors concluded that INF-γ was not both given thrice weekly [103]. Objective responses
active against SCLC. In a phase III trial, 100 patients in were observed in only 3/76 (4%) patients; grade 4 toxicity
complete remission following treatment with six cycles was about 10% in each arm.
of combination chemotherapy, thoracic radiotherapy, Continuous infusion IL-2 (18 MIU/m2/day for 5 days)
and prophylactic cranial irradiation (PCI), were random- combined with IFN-α (5 MU s.c. every other day during
ized to observation or IFN-γ at a dose of 4 MU s.c. per IL-2) produced no responses in seven patients with
day for 6 months [37]. Significant toxicity including NSCLC [76]. In another trial of continuous i.v. IL-2 (18
chills, myalgia, lethargy, and alteration of mood-person- MIU/m2/day for 3 days) and IFNα (5 MU/m2/day i.m.
ality were observed in the IFN- γ group. There was no for 3 days) produced no responses in 11 patients [35].
difference in progression free survival or overall sur- IL-2 at a dose of 3 MU/m2 s.c. twice daily, and IFN-α
vival, although the trends favored the group who did not at a dose of 3 MU once daily, 5 days a week was given
receive IFN-γ. The study had sufficient power to exclude as a consolidation treatment to patients with NSCLC
a 33% improvement in survival (P = 0.04) for the IFN-γ. cancer who had responded to chemotherapy, but no ben-
In another phase III trial, 127 of 177 patients with SCLC efit was observed in a study of 52 patients in which
who were in complete or nearly-complete response fol- many patients dropped out because of toxicity [105].
lowing chemotherapy with or without thoracic radiother-
apy, were randomized to observation or to receive either
IL-2 in SCLC
IFN-γ 4 MU (0.2 mg) sc every other day for 4 months or
observation [107]. There was no difference in progres- In a phase II trial, 24 of 50 patients with extensive
sion free or overall survival. SCLC who had not achieved complete remission with
Robert O. Dillman 685

PACE (cisplatin, doxorubicin, cyclophosphamide, and and 32 IIIB NSCLC who were randomized to receive
etoposide) chemotherapy, were treated with IL-2 by s.c. IL-2 with TIL with either chemotherapy or radiation
4-day continuous i.v. infusion of 4.5 MIU/m2 per day for stage III A disease, or to observation alone in patients
for up to 8 weeks [17]. Four patients improved to a with stage II disease [83]. The radiation therapy con-
complete response and one improved from stable dis- sisted of 60 Gy and the chemotherapy of cisplatin and
ease to a partial response for an improvement rate of vinblastine followed by radiation therapy. The radia-
21% (5/21), but the treatment was quite toxic, and only tion/chemotherapy was not given until 2–3 months after
five patients completed the planned 8 weeks of treat- the administration of IL-2 and TIL. Better survival was
ment. These radiographic improvements in response seen for the patients who received IL-2 plus TIL in
may have been because of faster resolution of dead stage IIIA (median survival 22 months versus 9 months,
tumor cells by activated macrophages rather than a p = 0.06) and stage III B (median survival 24 months
cytotoxic anti-tumor effect. versus 7 months, p < 0.01). There was no difference in
survival for patients with stage II disease. There has
been no effort to replicate this study in the United States.
IL-2 and LAK One criticism of this study is that the median survival
A phase II study of interleukin-2 (IL-2) at a fixed dose of rates for this chemotherapy followed by radiation therapy
6 MU/m2 per day as a 24 h continuous intravenous infu- has been 13–14 months in U.S. trials, and the analysis
sion (CIV) with lymphokine-activated killer (LAK) cells was based on treatment rather than intent to treat.
yielded one near complete response that lasted 18 months
[9]. Two Japanese trials have suggested that IL-2 plus
LAK may be of value in NSCLC. A randomized trial of
immunotherapy with IL-2 plus LAK cell was conducted
Interleukin-4
in 105 patients after noncurative resection of primary In a randomized phase II trial, 63 patients with advanced
lung cancer [43]. All patients received standard chemo- NSCLC, 44 of whom had received prior combination
therapy and/or radiotherapy with or without the addition chemotherapy were randomized to receive one of two
of the immunotherapy. The 7-year survival rate was different dose of Interleukin-4 (IL-4) (0.25 μg/kg and
greater in the immunotherapy group than in the control 1.0 μg/kg) given s.c. thrice weekly [109]. Common side
group (39% versus 13%, p < 0.01). The same investiga- effects were fatigue and fever. Only one of 55 evaluable
tors conducted a randomized prospective study of post patients had an objective tumor response. A subsequent
surgical adjuvant immunotherapy using IL-2 and LAK randomized three-arm placebo-controlled phase III trial
cells in 82 patients who had undergone a curative resec- of chemotherapy alone or with one of two different dose of
tion of primary lung cancer [44]. The three arms included IL-4 showed no difference in response rate or survival
observation alone, adjuvant chemotherapy (cisplatin, (unpublished results).
vindesine, and mitomycin C) and adjuvant chemotherapy
for two cycles followed by IL-2 plus LAK. Chemotherapy
plus immunotherapy produced a 5-year survival rate
of 50% compared to 33% for observation and 30% for
Amifostine (Ethyol™)
chemotherapy alone (p = 0.03). Amifostine is an analog of cysteamine that selectively
protects normal tissues of various organ systems against
the toxic effects of various cytotoxic drugs and radiation.
IL-2 and TIL Because of this it has been suggested that amifostine be
In a Phase I clinical-trial retroviral-mediated IL-2 genes given concurrently with chemotherapy and radiation
were transferred to tumor infiltrating lymphocytes (TIL) therapy in the treatment of lung cancer in an effort to
that were infused into ten lung cancer patients with decrease toxicity [102, 15]. It was originally named
refractory pleural effusions [101]. Autologous TIL were Walter Reed 2721, a reflection of its origin from a clas-
exposed to the retroviral plasmid pL(IL-2)SN contain- sified military research program that was trying to
ing the human IL-2 gene with about 10 to 16 billion develop agents that would protect personnel from the
IL-2-transfected TIL cells infused into the chest cavity lethal effects of radiation. Amifostine is actually a prod-
of each patient. Pleural effusions did not re-accumulate rug that is converted by tissue alkaline phosphatase to
for at least 4 weeks in six of ten patients. WR-1065, a free thiol with a sulfhydryl group that binds
In a randomized study, TIL cultures were successfully to oxygen-free radicals generated by radiation and chemo-
established for 113 of 131 patients, 13 stage II, 42 IIIA, therapy. It may also upregulate p53 as an additional
686 Biological therapy of lung cancer

mechanism of chemoresistance. It acts a broad-spectrum treated SCLC patients. One patient had complete reso-
cytoprotective agent that seems to protect virtually all lution of radiographically detectable tumor that lasted 4
normal tissues except perhaps the brain. The alkaline months. Four patients (33%) had stable disease. No
phosphatase enzyme that converts WR-2721 to WR-1065 toxic reactions were observed.
is expressed in much greater amounts in normal blood The anti-VEGF Mab bevacizumab (Avastin®) is now
vessels and normal tissues than in neoplastic vasculature part of standard therapy for patients with non-squamous
or membranes of malignant cells. Numerous clinical NSCLC. In a three-arm randomized phase II trial 99
trials have shown that amifostine decreases the toxic previously untreated NSCLC patients were randomized
effects of chemotherapy and radiation on normal tissues to treatment every 3 weeks with paclitaxel 200 mg/m2
without any diminution in clinical efficacy as manifest and carboplatin (AUC = 6) (PC) alone or in combina-
by equivalent response rates and survival. Cytoprotective tion with bevacizumab at 15 or 7.5 mg/kg [38]. The
effects without decreased anti-tumor efficacy were dem- response rate and PFS were higher for the 15 mg/kg
onstrated for patients with NSCLC who were receiving bevacizumab arm compared to PC alone with a trend
carboplatin [10, 51]. A small randomized trial in 62 toward better survival. Bleeding was the most signifi-
patients suggested a better survival for patients with cant complication associated with bevacizumab and
stage II and III NSCLC who received chemoradiotherapy included minor mucocutaneous hemorrhage, and major
with amifostine compared to chemoradiotherapy alone hemoptysis associated with centrally located squamous
[45]. However, a larger national trial in 242 patients cell cancers accompanied by tumor necrosis and cavita-
stage II to IIIA/B non-small-cell lung cancer failed to tion. In a large two-arm randomized trial, 878 patients
confirm any benefit for amifostine to carboplatin/paclitaxel with previously untreated non-squamous NSCLC were
chemotherapy [71]. randomized to PC + bevacizumab (15 mg/kg) or PC
alone on a 3 week schedule [94]. The bevacizumab arm
was associated with a higher response rate (p < 0.001),
better OS (HR = 0.79, p = 0.003), and PFS (HR 0.66,
Monoclonal Antibodies p < 0.001). Even though patients with squamous cell
The murine anti-Epcam monoclonal antibody (Mab) histology were not enrolled in this trial, there were still
KS1/4 and KS1/4-methotrexate immunoconjugate were five hemorrhagic deaths in the bevacizumab arm (1.2%)
administered to patients with Stage IIIB or IV NSCLC. and significant bleeding was more frequent in the beva-
Six patients received KS1/4 alone and five patients cizumab arm (4.4% versus 0.7%, p < 0.001). In a large
received KS1/4-methotrexate conjugate [24]. Mild to European trial 1,043 patients with previously untreated
moderate side effects including fever, chills, anorexia, non-squamous NSCLC were randomized to gemcit-
nausea, vomiting, diarrhea, anemia, and brief transami- abine plus cisplatin with bevacizumab at 10 mg/kg or
nasemia were seen in both groups. There was one possible 5 mg/kg or placebo [63]. Results favored the bevaci-
clinical response in a patient treated with unconjugated zumab arms, although the benefit was not as impressive
antibody. Results were not better with a KS1/4 metho- as in the U.S. trial with PC.
trexate immunoconjugate, but the latter was associated As shown in Table 1, the anti-EGFR Mab cetuximab
with greater GI toxicity because of reactivity with antigen (Erbitux®) also enhances the effects of chemotherapy in
on normal small bowel. NSCLC. In NSCLC Hanna et al. observed a response
An immunotoxin consisting of murine Mab N901, that rate of only 5% for cetuximab alone in 66 patients with
binds to the CD56 (neural cell adhesion molecule metastatic disease who had progressed after previous
[NCAM]) antigen found on cells of neuroendocrine ori- systemic therapy [29]. Other studies have combined
gin, and blocked ricin was tested in a phase I trial in 21 cetuximab with chemotherapy. Thienelt et al. reported a
patients with SCLC [59]. Successive cohorts of at least 26% response rate in 31 previously untreated patients
three patients were treated at doses from 5 to 40 μg/kg/ who received paclitaxel, carboplatin, and cetuximab
day for 7 days. The dose-limiting toxicity was a capillary [104a]. Robert et al. reported a 29% response rate in 35
leak syndrome that occurred in two of three patients at the patients who were treated with gemcitabine, carboplatin
highest dose. No patient developed clinically significant and cetuximab [87].
neuropathy. One patient achieved a partial response. Some patients with NSCLC have tumors that express
The murine Mab 2A11 binds to gastrin-releasing peptide Her2, the target of trastuzumab (Herceptin®). In one
(GRP), which binds to receptors and stimulates growth study only 24/209 (11%) patients had Her2-positive
of SCLC cells [40]. 2A11 at a dose of 250 mg/m2 over 1 h tumors by immunothistochemistry (IHC), and only 1/24
thrice weekly for 4 weeks was given to 13 previously (4%) of them had a response [18]. In another NSCLC
Robert O. Dillman 687

trial 13/69 had Her2-positive tumors by IHC, and 0/13 low tumor burden stage IV NSCLC were randomized to
responded to trastuzumab [48]. Trials of chemotherapy receive 12.5, 25, or 50 million cells per injection on a
and trastuzumab were also compromised by the low monthly or every other month schedule to a maximum
proportion of patients with HER2-positive tumors. For of 16 injections [73]. There were no significant adverse
example, only 16% to 21% of patients in three NSCLC events associated with the treatment. There was a dose-
trials had HER2-positive tumors, and in two of the trials related survival benefit in patients who received the two
less than 5% of patients had tumors that were Her2 3 higher doses who had a 2-year survival rate of 52%
+ by IHC [26, 47, 116]. Gatzemeier et al. randomized compared to 20% for patients in the lowest dose cohort
101 patients with Her2-positive NSCLC to trastuzumab (p = 0.007). There was response rate of 15% among 61
plus gemcitabine and cisplatin or the chemotherapy patients who had stage IIIB or IV disease, although this
alone, and found no difference in response rate (36% interpretation was confounded by previous therapy.
versus 41%) or PFS (6.1 versus 7.0 months), but only Belagenpumatucel-L is being tested further in a pivotal
six patients had tumors that were 3 + Her2 by IHC, and randomized phase III trial.
five of them did have an objective response to treatment
[26]. Zinner et al. observed a 38% response rate in 21
patients treated with gemcitabine and cisplatin [116]. Summary
Krug et al. observed a 28% response rate among 64
patients including 7/30 in patients treated with docetaxel As of 2008, the anti-VEGF Mab bevacizumab is the
plus trastuzumab and 11/34 in the patients treated only biotherapeutic that has a ever received a commer-
with paclitaxel plus trastuzumab [47]. Response rates cial marketing indication in lung cancer. None of the
did not appear to be higher than anticipated response cytokine or cell therapy approaches are considered
rates for chemotherapy alone, but to determine the true promising at this time, but other Mabs are still being
contribution of trastuzumab would require randomized tested. Some of the results with adoptive cell therapy are
trials. Collectively these studies have been disappointing. provocative, but have not been replicated. IFN-α adds
This does not appear to be a fruitful area for further little if any benefit after chemotherapy, and adds to toxi-
investigation given that so few NSCLC express Her2. cicty when administered with chemotherapy. Doses of
IL-2 that are effective in other malignancies are too
toxic to administer to patients with significant underly-
ing lung disease. The results with retinoids have been
Vaccines disappointing. Randomized trials will determine whether
There is great interest in the potential role of vaccines in any of the current vaccine approaches are worthwhile.
the adjuvant setting of lung cancer, but no products are
commercially available. In a randomized trial in which
95 post operative NSCLC received no additional treat- References
ment or an i.d. dose of an autologous irradiated suspen-
1. Amery WK. Final results of a multicenter placebo-controlled
sion of fresh tumour cells combined with a small dose of levamisole study of resectable lung cancer. Cancer Treat Rep
C parvum, there was no difference in outcome [100]. 1978;62:1677–83.
A vaccine consisting of an anti-idiotype antibody 2. Anthony HM, Mearns AJ, Mason MK, et al. Levamisole and sur-
BEC2, a mouse Mab that mimics the structure of the gery in bronchial carcinoma patients: increase in deaths from car-
diorespiratory failure. Thorax 1979; 34:4–12.
disialoganglioside GD3, was administered with BCG to
3. Ardizzoni A, Salvati F, Rosso R, et al. Combination of chemo-
15 SCLC patients following primary therapy with che- therapy and recombinant alpha-interferon in advanced non-small
motherapy with or without radiation therapy [28]. All cell lung cancer. Multicentric Randomized FONICAP Trial Report.
patients developed anti-BEC2 antibodies and five were The Italian Lung Cancer Task Force. Cancer 1993; 72:2929–35.
found to have antibodies against GD3. A Phase III trial 4. Ardizzoni A, Bonavia M, Viale M, et al. Biologic and clinical
effects of continuous infusion interleukin-2 in patients with non-
is being conducted to evaluate BEC2 plus BCG as adju- small cell lung cancer. Cancer 1994; 73:1353–60.
vant therapy after chemotherapy and irradiation. 5. Arnold A, Ayoub J, Douglas L, et al. Phase II trial of 13-cis-retin-
The most encouraging vaccine in lung cancer at the oic acid plus interferon α in non-small-cell lung cancer. J Natl
moment is belagenpumatucel-L, a transforming growth Cancer Inst 1994;86:306–9.
factor beta-2 (TGF-β) antisense gene-modified alloge- 6. Athanasiadis I, Kies MS, Miller M, et al. Phase II study of all-
trans-retinoic acid and alpha-interferon in patients with advanced
neic tumor cell vaccine designed to inhibit secretion of non-small cell lung cancer. Clin Cancer Res 1995; 1:973–9.
TGF-β at the site of immunization. In a randomized 7. Ayoub J, Jean-Francois R, Cormier Y, et al. Placebo-controlled
phase II trial, 75 NSCLC patients with stage II, III, or trial of 13-cis-retinoic acid activity on retinoic acid receptor-beta
688 Biological therapy of lung cancer

expression in a population at high risk: implications for chemopre- 25. Garaci E, Lopez M, Bonsignore G, et al. Sequential chemoimmu-
vention of lung cancer. J Clin Oncol 1999; 17:3546–52. notherapy for advanced non-small cell lung cancer using cisplatin,
8. Bedikian AY, Patt YZ, Murphy WK, et al. Prospective evaluation etoposide, thymosin-alpha 1 and interferon-alpha 2a. Eur J Cancer
of thymosin fraction V immunotherapy in patients with non-small 1995;31A:2403–5.
cell lung cancer receiving vindesine, doxorubicin, and cisplatin 26. Gatzemeier U, Groth G, Butts C, et al. Randomized phase II trial
(VAP) chemotherapy. Am J Clin Oncol 1984; 7:399–404. of gemcitabine-cisplatin with or without trastuzumab in HER2-
9. Bernstein ZP, Goldrosen MH, Vaickus L, et al. Interleukin-2 with positive non-small-cell lung cancer. Ann Oncol 2004;15:19–27.
ex vivo activated killer cells: therapy of advanced non-small-cell 27. Goncalves A, Camerlo J, Bun H, et al. Phase II study of a combina-
lung cancer. J Immunother 1991; 10:383–7. tion of cisplatin, all-trans-retinoic acid and interferon-alpha in
10. Betticher DC, Anderson H, Ranson M, et al. Carboplatin combined squamous cell carcinoma: clinical results and pharmacokinetics.
with amifostine, a bone marrow protectant, in the treatment of Anticancer Res 2001;21:1431–7.
non-small-cell lung cancer: a randomised phase II study. Br J 28. Grant SC, Kris MG, Houghton AN, Chapman PB. Long survival
Cancer 1995; 72:1551–5. of patients with small cell lung cancer after adjuvant treatment
11. Bitran JD, Green M, Perry M, et al. A phase II study of recombi- with the anti-idiotypic antibody BEC2 plus Bacillus Calmette-
nant interferon-gamma following combination chemotherapy for Guerin. Clin Cancer Res 1999;5:1319–23.
patients with extensive small cell lung cancer. CALGB. Am J Clin 29. Hanna N, Lilenbaum R, Ansari R, et al. Phase II trial of cetuximab
Oncol 1995; 18:67–70. in patients with previously treated non-small-cell lung cancer. J
12. Bjornsson S, Takita H, Kuberka N, et al. Combination chemother- Clin Oncol 2006;24:5253–8.
apy plus methanol extracted residue of bacillus Calmette-Guerin 30. Hennekens CH, Buring JE, Manson JE, et al. Lack of effect of
or Corynebacterium parvum in stage III lung cancer. Cancer Treat long-term supplementation with beta carotene on the incidence of
Rep 1978;62:505–10. malignant neoplasms and cardiovascular disease. N Engl J Med
13. Blumenschein GR, Khuri F, Gatzemeier U, et al. A randomized 1996;334:1145–49.
phase III trial comparing bexarotene-carboplatin-paclitaxel versus 31. Herskovic A, Bauer M, Seydel HG, et al. Post-operative thoracic
carboplatin-paclitaxel in chemotherpay-naïve patients with advanced irradiation with or without levamisole in non- small cell lung can-
or metastatic non-small cell lung cancer. J Clin Oncol 2005;23 cer: results of a Radiation Therapy Oncology Group Study. Int J
(16 Suppl) 621s (abstract #LBA7001). Radiat Oncol Biol Phys 1988;14:37–42.
14. Bradley JD, Scott CB, Paris KJ, et al. A phase III comparison of 32. Holmes EC, Gail M. Surgical adjuvant therapy for stage II and
radiation therapy with or without recombinant beta-interferon for stage III adenocarcinoma and large-cell undifferentiated carci-
poor-risk patients with locally advanced non-small-cell lung cancer noma. J Clin Oncol 1986;4:710–5.
(RTOG 93–04). Int J Radiat Oncol Biol Phys 2002;52:1173–9. 33. Issell BF, Valdivieso M, Hersh EM, et al. Combination chemoim-
15. Castiglione F, Porcile G, Gridelli C. The potential role of amifos- munotherapy for extensive non-oat cell lung cancer. Cancer Treat
tine in the treatment of non small cell lung cancer. Lung Cancer Rep 1978; 62:1059–63.
2000; 29:57–66. 34. Jackson DV, Jr., Paschal BR, Ferree C, et al. Combination chemo-
16. Chaninian AP, Goldberg J, Holland JF, et al. Chemotherapy versus therapy-radiotherapy with and without the methanol- extraction
chemoimmunotherapy with levamisole or Corynebacterium par- residue of Bacillus Calmette-Guerin (MER) in small cell carci-
vum in advanced lung cancer. Cancer Treat Rep 1982;66:1291–7. noma of the lung: a prospective randomized trial of the Piedmont
17. Clamon G, Herndon J, Perry MC, et al. Interleukin-2 activity in Oncology Association. Cancer 1982;50:48–52.
patients with extensive small-cell lung cancer: a phase II trial of 35. Jansen RL, Slingerland R, Goey SH, et al. Interleukin-2 and inter-
Cancer and Leukemia Group B. J Natl Cancer Inst 1993; 85:316–20. feron-alpha in the treatment of patients with advanced non-small-
18. Clamon G, Herndon J, Kern J, et al. Lack of trastuzumab activity cell lung cancer. J Immunother 1992;12:70–3.
in nonsmall cell lung carcinoma with overexpression of erb-B2: 36. Jassem J, Zatloukal P, Ramlau R, et al. A randomized phase III trial
39810: a phase II trial of Cancer and Leukemia Group B. Cancer comparing bexarotene-cisplatin-vinorelbine versus cisplatin-
2005;103:1670–5. vinorelbine in chemotherpay-naive patients with advanced or
19. Cohen MH, Chretien PB, Ihde DC, et al. Thymosin fraction V and metastatic non-small cell lung cancer. J Clin Oncol 2005;23 (16
intensive combination chemotherapy. Prolonging the survival of Suppl) 627s (abstract #LBA7024).
patients with small-cell lung cancer. JAMA 1979; 241:1813–15. 37. Jett JR, Maksymiuk AW, Su JQ et al. Phase III trial of recombinant
20. Davis S, Mietlowski W, Rohwedder JJ et al. Levamisole as an interferon gamma in complete responders with small-cell lung
adjuvant to chemotherapy in extensive bronchogenic carcinoma: a cancer. J Clin Oncol 1994; 12:2321–2326.
Veterans Administration Lung Cancer Group Study. Cancer 38. Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized
1982;50:646–51. phase II trial comparing bevacizumab plus carboplatin and pacli-
21. Dillman RO, Beauregard JC, Zavanelli MI, et al. In vivo immune res- taxel with carboplatin and paclitaxel alone in previously untreated
toration in advanced cancer patients after administration of thymosin locally advanced or metastatic non-small-cell lung cancer. J Clin
fraction 5 or thymosin alpha 1. J Biol Response Mod 1983;2:139–49. Oncol 2004;22:2184–91.
22. Dillman RO, Beauregard J, Royston I, Zavanelli MI. Phase II trial 39. Kataja V, Yap A. Combination of cisplatin and interferon-alpha 2a
of thymosin fraction 5 and thymosin alpha 1. J Biol Response Mod (Roferon-A) in patients with non-small cell lung cancer (NSCLC).
1987; 6:263–7. An open phase II multicentre study. Eur J Cancer 1995;31A:35–40.
23. Edwards FR. Use of BCG as an immunostimulant after resection 40. Kelley MJ, Linnoila RI, Avis IL, et al. Antitumor activity of a
of carcinoma of the lung: a two-year assessment of a trial of 500 monoclonal antibody directed against gastrin- releasing peptide in
patients. Thorax 1979; 34:801–6. patients with small cell lung cancer. Chest 1997;112:256–61.
24. Elias DJ, Hirschowitz L, Kline LE, et al. Phase I clinical compara- 41. Kelly K, Crowley JJ, Bunn PA, Jr., et al. Role of recombinant inter-
tive study of monoclonal antibody KS1/4 and KS1/4-methotrexate feron alfa-2a maintenance in patients with limited-stage small-cell
immunconjugate in patients with non-small cell lung carcinoma. lung cancer responding to concurrent chemoradiation: a Southwest
Cancer Res 1990;50:4154–9. Oncology Group study. J Clin Oncol 1995;13:2924–30.
Robert O. Dillman 689

42. Khuri FR, Fossella FV, Lee JS et al. Phase II trial of recombinant 60. Maasilta P, Holsti LR, Halme M, et al. Natural alpha-interferon in
IFN-alpha2a with etoposide/cisplatin induction and interferon/ combination with hyperfractionated radiotherapy in the treatment
megestrol acetate maintenance in extensive small cell lung cancer. of non-small cell lung cancer. Int J Radiat Oncol Biol Phys 1992;
J Interferon Cytokine Res 1998; 18:241–5. 23:863–8.
43. Kimura H, Yamaguchi Y. Adjuvant immunotherapy with interleukin 61. Macchiarini P, Hardin M, Angeletti CA. Long-term evaluation of
2 and lymphokine-activated killer cells after noncurative resection intrapleural bacillus Calmette-Guerin with or without adjuvant
of primary lung cancer. Lung Cancer 1995;13:31–44. chemotherapy in completely resected stages II and III non-small-
44. Kimura H, Yamaguchi Y. Adjuvant chemo-immunotherapy after cell lung cancer. Am J Clin Oncol 1991; 14:291–7.
curative resection of Stage II and IIIA primary lung cancer. Lung 62. Mandanas R, Einhorn LH, Wheeler B, et al. Carboplatin (CBDCA)
Cancer 1996; 14:301–4. plus alpha interferon in metastatic non-small cell lung cancer. A
45. Komaki R, Lee JS, Milas L, et al. Effects of amifostine on acute Hoosier Oncology Group phase II trial. Am J Clin Oncol 1993;
toxicity from concurrent chemotherapy and radiotherapy for inop- 16:519–21.
erable non-small-cell lung cancer: report of a randomized com- 63. Manegold C, von Pawel J, Zatoukal P, et al. Randomised, double-
parative trial. Int J Radiat Oncol Biol Phys 2004;58:1369–77. blind multicenter phase III study of bevacizumab in combination
46. Krauss S, Comas F, Perez C, et al. Treatment of inoperable non- with cisplatin and gemcitabine in chemotherapy-naïve patients
small cell carcinoma of the lung with radiation therapy, with or with advanced or recurrent non-squamous non-small cell lung can-
without levamisole. A randomized trial of the Southeastern Cancer cer. J Clin Oncol 2007;25:18s (part II) [abstract LBA7514].
Study group. Am J Clin Oncol 1984; 7:405–12. 64. Matthay RA, Mahler DA, Beck GJ, et al. Intratumoral Bacillus
47. Krug LM, Miller VA, Patel J, et al. Randomized phase II study of Calmette-Guerin immunotherapy prior to surgery for carcinoma of
weekly docetaxel plus trastuzumab versus weekly paclitaxel plus the lung: results of a prospective randomized trial. Cancer Res
trastuzumab in patients with previously untreated advanced nons- 1986;46:5963–8.
mall cell lung carcinoma. Cancer 2005;104:2149–55. 65. Mattson K, Niiranen A, Ruotsalainen T, et al. Interferon mainte-
48. Lara PN Jr, Laptalo L, Longmate J, et al. Trastuzumab plus doc- nance therapy for small cell lung cancer: improvement in long-
etaxel in HER2/neu-positive non-small-cell lung cancer: a term survival. J Interferon Cytokine Res 1997;17:103–5.
California Cancer Consortium screening and phase II trial. Clin 66. Maurer LH, Pajak T, Eaton W, et al. Combined modality therapy
Lung Cancer 2004;5:231–6. with radiotherapy, chemotherapy, and immunotherapy in limited
49. Lee JS, Lippman SM, Benner SE, et al. Randomized placebo- small-cell carcinoma of the lung: a Phase III cancer and Leukemia
controlled trial of isotretinoin in chemoprevention of bronchial Group B Study. J Clin Oncol 1985;3:969–76.
squamous metaplasia. J Clin Oncol 1994a; 12:937–45. 67. McCracken JD, Chen T, White J, et al. Combination chemother-
50. Lee YC, Luh SP, Wu RM, Lee CJ. Adjuvant immunotherapy with apy, radiotherapy, and BCG immunotherapy in limited small-cell
intrapleural Streptococcus pyogenes (OK- 432) in lung cancer carcinoma of the lung: a Southwest Oncology Group Study. Cancer
patients after resection. Cancer Immunol Immunother 1994b; 1982;49:2252–8.
39:269–74. 68. McKneally MF, Maver C, Kausel HW. Regional immunotherapy
51. Leong SS, Tan EH, Fong KW, et al. Randomized double-blind trial of lung cancer with intrapleural B.C.G. Lancet 1976a;1:377–9.
of combined modality treatment with or without amifostine in 69. McKneally MF, Maver C, Kausel HW, Alley RD. Regional immu-
unresectable stage III non-small-cell lung cancer. J Clin Oncol notherapy with intrapleural BCG for lung cancer. J Thorac
2003;21:1767–74. Cardiovasc Surg 1976b;72:333–8.
52. Lippman SM, Lee JJ, Karp DD, et al. Randomized phase III inter- 70. Millar JW, Roscoe P, Pearce SJ, et al. Five-year results of a con-
group trial of isotretinoin to prevent second primary tumors in stage trolled study of BCG immunotherapy after surgical resection in
I non-small-cell lung cancer. J Natl Cancer Inst 2001;93:605–18. bronchogenic carcinoma. Thorax 1982;37:57–60.
53. Lissoni P, Tisi E, Barni S, et al. Biological and clinical results of a 71. Movsas B, Scott C, Langer C, et al. Randomized trial of amifostine
neuroimmunotherapy with interleukin-2 and the pineal hormone in locally advanced non-small-cell lung cancer patients receiving
melatonin as a first line treatment in advanced non-small cell lung chemotherapy and hyperfractionated radiation: radiation therapy
cancer. Br J Cancer 1992; 66:155–8. oncology group trial 98–01. J Clin Oncol 2005;23:2145–54.
54. Lissoni P, Meregalli S, Fossati V, et al. A randomized study of 72. National Cancer Institute Lung Cancer Study Group. Surgical
immunotherapy with low-dose subcutaneous interleukin-2 plus adjuvant intrapleural BCG treatment for stage I non-small cell
melatonin vs chemotherapy with cisplatin and etoposide as first- lung cancer. Preliminary report of the National Cancer Institute
line therapy for advanced non-small cell lung cancer. Tumori Lung Cancer Study Group. J Thorac Cardiovasc Surg 1981;82:
1994;80:464–7. 649–57.
55. Low TL, Goldstein AL. Thymosins: structure, function and thera- 73. Nemunaitis J, Dillman RO, Schwarzenberger PO, et al. Phase II
peutic applications. Thymus 1984; 6:27–42. study of belagenpumatucel-L, a transforming growth factor beta-2
56. Lowe J, Iles PB, Shore DF, et al. Intrapleural BCG in operable antisense gene-modified allogeneic tumor cell vaccine in non-
lung cancer. Lancet 1980;1:11–4. small-cell lung cancer. J Clin Oncol 2006;24:4721–30.
57. Ludwig Lung Cancer Study Group (LLCSG). Intrapleural and 74. Nio Y, Nagami H, Tamura K, et al. Multi-institutional randomized
intravenous Corynebacterium parvum in patients with resected clinical study on the comparative effects of intracavital chemo-
stage I and II non-small cell carcinoma of the lung. Cancer therapy alone versus immunotherapy alone versus immunochemo-
Immunol Immunother 1986a;23:1–4. therapy for malignant effusion. Br J Cancer 1999;80:775–85.
58. Ludwig Lung Cancer Study Group (LLCSG). Immunostimulation 75. O’Brien ME, Saini A, Smith IE, et al. A randomized phase II study
with intrapleural BCG as adjuvant therapy in resected non-small of SRL172 (Mycobacterium vaccae) combined with chemother-
cell lung cancer. Cancer 1986b;58:2411–6. apy in patients with advanced inoperable non-small-cell lung can-
59. Lynch TJ, Jr., Lambert JM, Coral F, et al. Immunotoxin therapy of cer and mesothelioma. Br J Cancer 2000;83:853–7.
small-cell lung cancer: a phase I study of N901- blocked ricin. J 76. Oldham RK, Blumenschein G, Schwartzberg L, et al. Combination
Clin Oncol 1997; 15:723–34. biotherapy utilizing interleukin-2 and alpha interferon in patients
690 Biological therapy of lung cancer

with advanced cancer: a National Biotherapy Study Group Trial. 94. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone
Mol Biother 1992;4:4–9. or with bevacizumab for non-small-cell lung cancer. N Engl
77. Omenn GS, Goodman GE, Thornquist MD, et al. Effects of a com- J Med 2006;355:2542–50.
bination of beta carotene and vitamin A on lung cancer and cardio- 95. Sarna G, Figlin R, Callaghan M. α(human leukocyte)-interferon
as treatment for non-small cell carcinoma of the lung: a phase II
vascular disease. N Engl J Med 1996;334:1150–5.
trial. J Biol Response Mod 1983;2:343–7.
78. Pastorino U, Infante M, Maioli M, et al. Adjuvant treatment of 96. Sarna GP, Lowitz BB, Haskell CM, et al. Chemo-immunotherapy
stage I lung cancer with high-dose vitamin A. J Clin Oncol for unresectable bronchogenic carcinoma. Cancer Treat Rep
1993;11:1216–22. 1978; 62:681–7.
79. Perez CA, Bauer M, Emami BN, et al. Thoracic irradiation with or 97. Scher HI, Shank B, Chapman R, et al. Randomized trial of com-
without levamisole (NSC #177023) in unresectable non-small cell bined modality therapy with and without thymosin fraction V in the
carcinoma of the lung: a phase III randomized trial of the RTOG. treatment of small cell lung cancer. Cancer Res 1988; 48:1663–70.
Int J Radiat Oncol Biol Phys 1988; 15:1337–46. 98. Schiller JH, Storer B, Dreicer R, et al. Randomized phase II-III
80. Pines A. A 5-year controlled study of B.C.G. and radiotherapy trial of combination beta and gamma interferons and etoposide
and cisplatin in inoperable non-small cell cancer of the lung.
inoperable lung cancer. Lancet 1976; 1:380–1.
J Natl Cancer Inst 1989;81:1739–43.
81. Prior C, Oroszy S, Oberaigner W, et al. Adjunctive interferon- 99. Silva RR, Bascioni R, Rossini S, et al. A phase II study of mito-
alpha-2c in stage IIIB/IV small-cell lung cancer: a phase III trial. mycin C, vindesine and cisplatin combined with alpha interferon
Eur Respir J 1997;10:392–6. in advanced non-small cell lung cancer. Tumori 1996;82:68–71.
82. Quan WD, Jr., Casal R, Rosenfeld M, Walker PR. Alpha inter- 100. Souter RG, Gill PG, Gunning AJ, Morris PJ. Failure of specific active
feron-2b, leucovorin, and 5-fluorouracil (ALF) in non-small cell immunotherapy in lung cancer. Br J Cancer 1981;44:496–501.
lung cancer. Cancer Biother Radiopharm 1996;11:229–34. 101. Tan Y, Xu M, Wang W, et al. IL-2 gene therapy of advanced lung
83. Ratto GB, Zino P, Mirabelli S, et al. A randomized trial of adoptive cancer patients. Anticancer Res 1996;16:1993–8.
102. Tannehill SP, Mehta MP, Larson M, et al. Effect of amifostine on
immunotherapy with tumor-infiltrating lymphocytes and interleukin-2
toxicities associated with sequential chemotherapy and radiation
versus standard therapy in the postoperative treatment of resected therapy for unresectable non-small-cell lung cancer: results of a
non-small cell lung carcinoma. Cancer 1996; 78:244–51. phase II trial. J Clin Oncol 1997;15:2850–7.
84. Recchia F, De Filippis S, Pompili PL, et al. Carboplatin, vindesine, 103. Tester WJ, Kim KM, Krigel RL, et al. A randomized Phase II
5-fluorouracil-leucovorin and 13-cis retinoic acid in the treatment study of interleukin-2 with and without beta- interferon for
of advanced non-small cell lung cancer. A phase II study. Clin Ter patients with advanced non-small cell lung cancer: an Eastern
1999;150:269–74. Cooperative Oncology Group study (PZ586). Lung Cancer
85. Richards F, Howard V, Shore A, et al. Combination chemotherapy 1999;25:199–206.
with and without the methanol-extracted residue of bacillus 104a. Thienelt CD, Bunn PA Jr, Hanna N, et al. Multicenter phase I/II
study of cetuximab with paclitaxel and carboplatin in untreated
Calmette-Guerin (MER) in extensive non-small-cell lung cancer: a
patients with stage IV non-small-cell lung cancer. J Clin Oncol
prospective randomized study for the Piedmont Oncology 2005;23:8786–93.
Association. Cancer 1981;47:2827–32. 104b. Treat J, Friedland D, Luginbuhl W, et al. Phase II trial of all trans-
86. Rinaldi DA, Lippman SM, Burris HA, et al. Phase II study of 13-cis- retinoic acid in metastatic non-small cell lung cancer. Cancer
retinoic acid and interferon-alpha 2a in patients with advanced Invest 1996;14:1415–20.
squamous cell lung cancer. Anticancer Drugs 1993;4:33–6. 105. Tummarello D, Graziano F, Isidori P, et al. Consolidation biochemo-
87. Robert F, Blumenschein G, Herbst RS, et al. Phase I/IIa study of therapy for patients with advanced non-small cell lung carcinoma
cetuximab with gemcitabine plus carboplatin in patients with che- responding to induction PVM (cisplatin, vinblastine, mitomycin-C)
regimen. A phase II study. Cancer 1996;77:2251–7.
motherapy-naive advanced non-small-cell lung cancer. J Clin
106. Tyagi P. Bexarotene in combination with chemotherapy fails to
Oncol 2005;23:9089–96. prolong survival in patients with advanced non-small-cell lung
88. Robinson E, Bartal A, Cohen Y, et al. Treatment of lung cancer by cancer: results from the SPIRIT I and II trials. Clin Lung Cancer
radiotherapy, chemotherapy, and methanol extraction residue of BCG 2005;7:17–9.
(MER): clinical and immunological studies. Cancer 1977;40:1052–9. 107. van Zandwijk N, Groen HJ, Postmus PE, et al. Role of recombi-
89. Roeslin N, Lang JM, Morand G, et al. Regional immunotherapy in nant interferon-gamma maintenance in responding patients with
resectable squamous cell lung carcinoma. Analysis of a random- small cell lung cancer. A randomised phase III study of the
ized study. Cancer Immunol Immunother 1982;13:174–5. EORTC Lung Cancer Cooperative Group. Eur J Cancer
90. Ruotsalainen TM, Halme M, Tamminen K, et al. Concomitant 1997;33:1759–66.
108. Vokes EE, Haraf DJ, Hoffman PC. Escalating doses of interferon
chemotherapy and IFN-alpha for small cell lung cancer: a ran-
alpha-2A with cisplatin and concomitant radiotherapy: a phase I
domized multicenter phase III study. J Interferon Cytokine Res study. Cancer Chemother Pharmacol 1993;33:203–9.
1999;19:253–9. 109. Vokes EE, Figlin R, Hochster H, et al. A phase II study of recom-
91. Ruotsalainen T, Halme M, Isokangas OP, et al. Interferon-alpha and binant human interleukin-4 for advanced or recurrent non-small
13-cis-retinoic acid as maintenance therapy after high-dose combi- cell lung cancer. Cancer J Sci Am 1998; 4:46–51.
nation chemotherapy with growth factor support for small cell lung 110. White JE, Chen T, Reed R, et al. Limited squamous cell carci-
cancer – a feasibility study. Anticancer Drugs 2000;11:101–8. noma of the lung: a Southwest Oncology Group randomized
92. Sakamoto J, Teramukai S, Watanabe Y, et al. Meta-analysis of adju- study of radiation with or without doxorubicin chemotherapy and
with or without levamisole immunotherapy. Cancer Treat Rep
vant immunochemotherapy using OK-432 in patients with resected
1982;66:1113–20.
non-small-cell lung cancer. J Immunother 2001;24:250–6. 111. Woodruff M, Walbaum P. A phase-II trial of Corynebacterium
93. Salvati F, Rasi G, Portalone L, et al. Combined treatment with parvum as adjuvant to surgery in the treatment of operable lung
thymosin-alpha1 and low-dose interferon-alpha after ifosfamide in cancer. Cancer Immunol Immunother 1983;16:114–6.
non-small cell lung cancer: a phase-II controlled trial. Anticancer 112. Wright PW, Hill LD, Peterson AV, Jr., et al. Preliminary results of
Res 1996;16:1001–4. combined surgery and adjuvant Bacillus Calmette- Guerin plus
Robert O. Dillman 691

levamisole treatment of resectable lung cancer. Cancer Treat Rep 115. Zarogoulidis K, Ziogas E, Papagiannis A, et al. Interferon alpha-
1978;62:1671–5. 2a and combined chemotherapy as first line treatment in SCLC
113. Yano T, Ichinose Y, Yokoyama H, et al. Inhalation therapy using patients: a randomized trial. Lung Cancer 1996;15:197–205.
a streptococcal preparation (OK-432) against bronchioloalveolar 116. Zinner RG, Glisson BS, Fossella FV, et al. Trastuzumab in com-
carcinoma of the lung. Anticancer Res 1999;19:5511–4. bination with cisplatin and gemcitabine in patients with Her2-
114. Yoshida K, Sugiura T, Takifuji N, et al. Randomized phase II trial overexpressing, untreated, advanced non-small cell lung cancer:
of three intrapleural therapy regimens for the management of report of a phase II trial and findings regarding optimal identification
malignant pleural effusion in previously untreated non-small cell of patients with Her2-overexpressing disease. Lung Cancer
lung cancer: JCOG 9515. Lung Cancer 2007;58:362–8. 2004;44:99–110.
21.7 Biological therapy of B and T cell
lymphoproliferative disorders
ROBERT O. DILLMAN

Biotherapy has consistently exhibited significant activity trial confirmed that IFN-α was superior to splenectomy
in lymphoid malignancies. The first report of a complete as initial treatment of the disease [155]. However, in
remission with a monoclonal antibody in 1982 was in a 1991 the purine analog deoxycoformycin (pentostatin)
patient with follicular lymphoma. Interferon alpha (IFN- gained regulatory approval for the treatment of HCL
α) became the first biological approved for the treatment and displaced IFN-α as the treatment of choice because
of malignancy in 1986, because of its activity in hairy was it active in interferon-refractory disease [67, 81]
cell leukemia, and for many years was used in the treat- less toxic, and superior to IFN-α when compared
ment of lymphoma. The first monoclonal antibody directly in randomized trials in previously untreated
approved for the treatment of a malignancy was the anti- patients [77]. In 1993 another purine analog 2-chlorode-
CD20 chimeric Mab rituximab, which has become a oxyadenosine (cladribine), which like pentostatin was
block-bluster drug since its approval in 1997. The first also associated with durable responses in over 90% of
immunotoxin to gain regulatory approval, denileukin previously untreated patients, was also approved for the
diftitox was approved based on activity in T cell lym- treatment of HCL [17, 136].
phoma. The anti-CD52 humanized Mab alemtuzumab The circulating lymphocytes of HCL typically express
was approved based on its activity in CLL. The first two very high levels of CD20. The anti-CD20 chimeric Mab
radiolabeled antibodies approved for cancer treatment rituximab has been used to treat relapsed HCL using both
were the anti-CD20 products Y-90 ibritumomab tiuxetan the 4 and 8 week treatment schedules of 375 mg/m2 per
and I-131 tositumomab which are very active in the week. The 4-week schedule produced responses in 5/10
treatment of B cell lymphoma. Finally, it is widely antici- patients in one study [103], but only in 6/24 patients who had
pated that perhaps one or more of the patient-specific all relapsed after cladbribine [126]. The 8-week schedule
vaccines consisting of idiotype protein may become the first produced responses in 8/15 relapsed patients [163].
tumor specific vaccines to receive regulatory approval as
treatment for follicular lymphoma.
B Cell Lymphoma
Interferon
Hairy Cell Leukemia In early trials single agent activity for IFN-α was dem-
This section starts with this relatively uncommon B cell onstrated in the lymphomas with response rates near
lymphoproliferative disorder because of its significance 50% in indolent lymphomas, and as high as 20% in
in the validation of biotherapy. In 1986, IFN-α became more aggressive lymphomas [185, 190, 192]. Numerous
the first biological to achieve regulatory approval based trials focused on the role of IFN-α in combination with
on its activity in hairy cell leukemia (HCL) [66, 75, 86, chemotherapy or as maintenance therapy following
164]. In one report 75% of 64 patients with HCL, 61 of induction chemotherapy as summarized in Table 1. In a
whom had undergone prior splenectomy, who were U.S. multicenter trial 249 patients with B cell lymphoma
treated with 2 MU/m2 IFN-α2b s.c. thrice weekly dem- were randomized to combination chemotherapy with
onstrated an objective response [75]. In another report cyclophosphamide, doxorubicin, vincristine and predni-
78% of 212 patients with HCL, 166 of whom had under- sone (COPA) with or without IFN-α [156, 157]. There
gone prior splenectomy, who were treated with 2 MU/ was better progression-free survival (PFS) and overall
m2 s.c. IFN-α2b s.c. thrice weekly had an objective survival (OS) in the IFN-α arm, but there was also more
response [164]. These responses proved to be durable toxicity. In a French trial 268 patients with B cell
with 28% in continuous remission beyond 6 years and lymphoma were randomized to combination chemo-
83% alive beyond 6 years [141]. A small randomized therapy with cyclophosphamide, doxorubicin, vindesine,

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 693
© Springer Science + Business Media B.V. 2009
694 Biological therapy of B and T cell lymphoproliferative disorders

Table 1. Randomized trials of interferon-alpha in the treatment of B-cell lymphoma.


Author Year Ref. # of Pts Induction Maintenance Results
Smalley et al. 1992, 2001 NEJM [156,157] Leukemia 249 COPA ± IFN None ↑PFS ↑OS
Peterson et al. 1993 1997 ASCO [133,191] 581 CTX ± IFN ± IFN for CR NSD
Solal-Celigny et al. 1993 NEJM [158] 268 CHVP ± IFN CHVP ± IFN ↑PFS ↑OS
Aviles et al. 1994 Leuk Lymphoma [2] 384 CEOP-Bleo ± IFN for CR ↑EFS ↑OS
Hagenbeek et al. 1998 J Clin Oncol [69] 347 CVP + RT to bulky ± IFN for OR/SD ↑PFS
Unterhalt et al. 1996 Leukemia [168] 498 PmM or CVP ± IFN for CR ↑DFS
Fisher et al. 2000 J Clin Oncol [49] 571 CHOP & ProMACE ± IFN 1 yr for OR NSD
Rohatiner et al. 2001 Br J Cancer [151] 204 CLB ± IFN ± IFN for OR NSD
Neri et al. 2001 J Hematother [125] Stem 151 MXT/Leu CEOP CVP ± IFN 1 yr ↑DFS ↑OS
Cell Res

CTX = cyclophosphamide
CLB = chlorambucil
COPA = cyclophosphamide, vincristine, prednisone, doxorubicin
CHOP = cyclophosphamide, vincristine, doxorubicin,prednisone
CHVP = cyclophosphamide, doxorubicin, vindesine, prednisone
CEOP-Bleo = cyclophosphamide, epirubicin, vincristine, prednisone, bleomycin
CVP = cyclophosphamide, vincristine, prednisone
ProMACE = procarbazine, methotrexate, doxorubicin, cyclophosphamide, etoposide
PmM = prednimustine and mitoxantrone
MXT/Leu = mitoxantrone, chlorambucil
CR = complete response
OR = objective response (partial or complete response)
PFS = progression free survival
EFS = event free survival (neither death nor disease progression)
DFS = disease free survival
OS = overall survival

prednisone (CHVP) with or without IFN, and the same no progressed during chemotherapy, to observation or
treatment was continued as maintenance treatment in IFN-α [69]. IFN-α maintenance therapy was associated
responders [158]. This trial also demonstrated improve- with longer PFS. In another 151 indolent lymphoma
ment in PFS and OS. It should be noted that these two patients were randomized to receive IFN-α or observation
randomized trials were not limited to patients with indolent after chemotherapy [125]. Again there was improvement
lymphoma. The US trial included patients with high-risk in PFS for patients who experienced a CR, and improved
follicular lymphoma as well as patients with intermediate OS for all patients. In a Spanish trial 384 patients with
grade non-follicular lymphoma. The French trial was follicular lymphoma who were in complete remission
conducted in patients with high-risk follicular lym- after six cycles of cyclophosphamide, epirubicin, vincris-
phoma defined by tumor mass >7 cm, B-symptoms, or tine, prednisone and bleomycin (CEOP-Bleo) chemother-
multiple extranodal sites of disease. apy, were randomized to observation or IFN-α maintenance
CALBG randomized 581 indolent lymphoma patients [4]. At a median follow up of almost 10 years, the IFN-α
to cyclophosphamide with or without IFN-α, with a arm had a better event free survival (64% versus 35%,
second randomization to observation or IFN-α for p < .01) and OS (81% versus 57%, p < .001). In contrast
patients who had complete remissions [133, 191]. There to these results, in a trial in which 204 patients were
was no difference based on the initial therapy, but there randomized to chlorambucil with or without IFN-α, and
was some trend toward better PFS for the patients who the responders were randomized to observation or IFN-
achieved a complete response (CR) and subsequently α, there was no difference in outcome [150]. In a U.S.
received maintenance IFN-α. A German trial randomized cooperative group trial there was no survival difference
indolent lymphoma patients who had achieved a CR for 571 patients with low-grade stage III or IV lympho-
with chemotherapy alone to IFN-α or observation [168]. mas who were randomized to CHOP and PROMACE
Maintenance IFN-α was associated with longer disease induction therapy with or without maintenance IFN-α,
free survival. A European cooperative group trial ran- and subsequently 268 responders were randomized to
domized indolent lymphoma patients whose disease had observation or 1 year of IFN-α [49].
Robert O. Dillman 695

A meta-analysis focused on ten randomized trials in transplant arm than the IFN-α maintenance arm (65%
which IFN-α was tested as part of initial therapy in versus 33%, p < .0001) [105]. More recently the inferi-
2005 patients with lymphoma [151]. In seven of these ority of IFN-α compared to other therapies was con-
trials IFN-α was used as part of initial therapy, and in firmed in another trial that compared rituximab plus
seven IFN-α was used as maintenance therapy. In the CHOP to CHOP as induction therapy in patients with
trials in which IFN-α was combined with chemotherapy, follicular lymphoma, and then randomized responders
there was no increase in response rates compared to che- to high-dose chemotherapy or maintenance IFN-α [72].
motherapy alone. However, the use of IFN-α either with or Survival was similar for RCHOP-transplant, RCHOP-
after chemotherapy was associated with better survival. IFN-α, and CHOP-transplant, all of which were supe-
The best results were obtained in four trials in which the rior to the CHOP-IFN-α arm. This trial seems to have
intensity of IFN-α therapy was greater than 36 MU per solidified chemotherapy plus rituximab as the current
month (p = .0004). Better survival was also noted in the standard initial therapy for patients with follicular lym-
five trials in which doxorubicin or mtioxantrone were phoma and suggests there is no general benefit for the
used in the combination chemotherapy (p = .0008). addition of IFN-α or high dose chemotherapy for
Based on these trials, prior to the introduction of patients who respond to RCHOP. Thus, controversies
rituximab, it appeared that IFN-α added nothing to che- regarding the role of IFN-α in B-cell lymphoma have
motherapy in the initial treatment of patients with low- been muted by the introduction of rituximab, which as a
risk indolent lymphoma, but the use of maintenance single agent had much higher response rates and a much
IFN-α in responding patients was associated with pro- milder toxicity profile than interferon.
longed PFS, but not OS. The converse appeared to be
true in high-risk, low-grade lymphoma. In that disease
setting it appeared that concurrent administration of
Interleukin-2
IFN-α with “CHOP-like” regimens was associated with The published experience in lymphoma with interleukin-2
a progression free and perhaps overall survival advantage, (IL-2) alone, or with adoptive cellular therapy, is sum-
but maintenance IFN-α did not appear to add benefit in marized in Table 2. In three small pilot studies, IL-2 +
that setting. lymphokine activated killer (LAK) cells were associ-
Two additional trials have been completed in lym- ated with response rates of 0%, 8%, and 50% [6, 113,
phoma patients who had responded to induction therapy, 172]. Five studies of IL-2 alone using high-dose bolus,
and then were randomized to consolidation with either low-dose bolus, or intermediate-dose continuous infu-
intensive chemotherapy and stem cell rescue, or IFN-α. sion intravenous [CIV] IL-2 were associated with
In patients with mantle cell lymphoma who were in CR response rates of 0%, 4%, 20% and 22% [1, 41, 65,
after chemotherapy the median PFS was much better in 172]. Most of these studies had a mix of all types of
the transplant arm than the IFN-α arm (39 versus 17 lymphoma patients. Interestingly, in one French trial the
months, p = .011) [40]. Similarly, in 244 patients with response rate for intermediate-dose CIV IL-2 was 22%
follicular lymphoma who were in remission following for intermediate lymphomas compared to only 4% in
chemotherapy, the 5-year PFS was twice as high in the indolent lymphoma [65]. In a cooperative group clinical

Table 2. Interleukin-2-based treatment of B-cell lymphoma


Lead author Year Ref. Regimen # Pts Response rate
Allison 1989 J Clin Oncol [1] LD bolus 9 22%
Margolin 1991 J Immunother [113] HD bolus & hybrid bolus/CIV + LAK 15 0%
Bernstein 1991 J Immunother [6] ID CIV + LAK 12 8%
Weber 1992 J Clin Oncol [172] HD bolus + LAK 8 50%
Weber 1992 J Clin Oncol [172] HD bolus 11 0%
Duggan 1992 J Immunother [41] LD bolus 20 20%
Gisselbrecht 1994 Blood [65] HD CIV for low-grade 24 4%
Gisselbrecht 1994 Blood [65] HD CIV for intermediate-grade 23 22%

HD = high dose
ID = intermediate dose
CIV = continuous intravenous
LAK = lymphokine activated killer cells
IFN = interferon
696 Biological therapy of B and T cell lymphoproliferative disorders

trial patients were randomized to a combination of low- relapsed after a previous response to rituximab [29, 85].
dose bolus IL-2 with or without IFN-β [41]. Toxicity In four trials in previously untreated follicular lymphomas
was substantial, and there appeared to be no advantage rituximab produced response rates ranging from 67% to
of the addition of IFN-β since the response rates were 78% [21, 64, 78, 179]. In small lymphocytic lymphoma
4/20 for IL-2 alone and 3/21 for IL-2 plus IFN-β. three trials in previously treated patients yielded response
Because of the success of rituximab in lymphoma, and rates of only 14% [54, 116, 137], but the response rate
the toxicity of higher dose IL-2 regimens, outpatient s.c. was 65% in such patients who had previously received
IL-2 was combined with rituximab for the treatment of chemotherapy and who received maintenance rituximab
relapsed follicular lymphoma with reported response [78]. In marginal zone lymphoma response rates were
rates of 10% in a trial of 54 patients [95], 26% in a trial 85% in previously untreated patients with extranodal
of 34 patients [186], and 55% in a trial of 20 patients MALT disease compared only 45% in such patients who
[61]. An industry sponsored randomized phase 2 trial of had relapsed after prior chemotherapy [22], and 77% in
8 weeks of s.c. IL-2 with 4 weeks of standard rituximab patients who had relapsed with gastric MALT [114]. In
for patients who had relapsed after prior chemotherapy lymphoplasmacytic lymphoma, four trials in previously
was terminated because of poor accrual. treated patients reported response rates ranging from
Because of the toxicity associated with high dose sched- 16% to 50% [37, 63, 166] while three trials with untreated
ules of IL-2, the limited single agent activity, and the patients reported response rates ranging from 35% to
emergence of other agents for the treatment of lymphoma, 65% with the best results obtained with extended rather
there has been little interest in pursuing trials with IL-2 in than only 4 weeks of therapy [37, 63, 167]. In the limited
recent years in the lymphomas other than as low doses for experience reported for single agent rituximab in mantle
immune reconstitution purposes following myelosuppres- cell lymphoma responses were about 30% regardless of
sive therapy, such as in the transplant setting. whether patients were previously untreated or had
relapsed after prior therapy [19, 54]. There are only two
published reports regarding single-agent rituximab in
Monoclonal Antibodies large B cell lymphoma, both of which were conducted in
In November 1997 Rituximab [Rituxan®] became the patients who had relapsed after prior therapy, and both
first monoclonal antibody (Mab) approved with a mar- recorded a response rate of 37% [19, 165]. In practice
keting indication for a malignant disease. Rituximab has rituximab is almost always used in combination with
revolutionized the treatment of B cell lymphoproliferative chemotherapy in aggressive lymphomas, but is often
disorders and is now standard therapy. The clinical trial used alone as initial therapy of patients with follicular
experience with rituximab is summarized in Table 3. In lymphoma or marginal zone lymphomas.
nine trials of relapsed, previously treated follicular lym- Rituximab has been safely combined or sequenced
phomas rituximab produced response rates ranging from with various types of chemotherapy with numerous
46% to 76% [3, 8, 15, 28, 54, 64, 116, 137, 178]. reports of high response rates and durable remissions.
Responses were evident within 2 months in most patients, These are summarized in Chapter 10 of this text. In large
and persisted for over a year from the date of first treatment. randomized trials rituximab plus chemotherapy has con-
Response rates of over 40% were also noted in patients sistently produced superior outcomes compared to the
with bulky B-cell lymphoma [31] and patients who same chemotherapy alone. In patients with previously

Table 3. Response rates for rituximab as a single agent


Lymphoma histology Setting # trials # patients Response range Median response rate
Follicular lymphoma Untreated 4 180 67–78% 73%
Follicular lymphoma Relapsed/refractory 9 547 46–76% 58%
Small lymphocytic Untreated 1 23 65% 65%
Small lymphocytic Relapsed/refractory 3 68 13–14% 14%
Marginal zone Untreated 1 24 87% 87%
Marginal zone Relapsed/refractory 2 37 45–77% 61%
Lymphoplasmacytic Untreated 4 78 35–65% 50%
Lymphoplasmacytic Relapsed/refractory 3 92 16–50% 33%
Mantle Cell Untreated 1 34 29% 29%
Mantle Cell Relapsed/refractory 2 54 30–33% 32%
Large B cell Relapsed/refractory 2 87 37% 37%
Robert O. Dillman 697

untreated follicular lymphomas response rates were higher complete response rate (27% versus 4%), was a striking
for RCHOP versus CHOP [72], RCVP versus CVP difference in the time to disease progression, medians of
[112], and RMCP versus MCP [71]. In previously 31 months versus 8 months (p = .007), but no difference
untreated indolent lymphoma response rates were higher in time to become “rituximab refractory” which was
for RFND versus FND [117]. In previously untreated about 30 months in both arms, nor in overall survival at
lymphoplasmacytoid lymphomas response rates were a median follow up of 42 months [70]. In large B cell
higher for RCHOP versus CHOP [11]. In relapsed folli- lymphoma the one randomized trial that addressed this
cular lymphoma response rates were higher for RCHOP issue found there was no advantage in terms of PFS or
versus CHOP [169] and for RFCM versus FCM [56]. In OS derived by giving four weekly doses of rituximab
relapsed mantle cell there was a higher response rate for every 6 months for 2 years after responding to RCHOP
RFCM versus FCM [55]. In the trials in which it is an induction chemotherapy [68]. In patients with relapsed
endpoint that has been assessed, PFS has also been longer mantle cell lymphoma who responded to RFCM as sec-
in those patients who received rituximab with their che- ond line therapy, compared to observation there was
motherapy [71, 112, 169]. Survival advantages have not also prolongation of PFS for patients who received two
been established in any of these trials, and probably will additional 4 week courses of rituximab 3 and 9 months
not be because of the clinical benefit associated with after completing RFCM [56]. There is no specific data
second and third-line therapies in these diseases. regarding this issue in small lymphocytic lymphoma,
In patients with large B cell lymphoma the addition of however, one would expect that any sort of additional
rituximab has not only typically increased response rates rituximab would prolong PFS based on the higher
and complete response rates, but more importantly, it has response rates achieved with prolonged rituximab in
also produced longer progression free and overall sur- that disorder, and the results in follicular lymphoma.
vival and is probably curative therapy for more than half Two radiolabeled murine Mabs have been approved
of all patients. All four large randomized trials in which for the treatment of B cell lymphoma. These are both
RCHOP was compared to CHOP as initial therapy for anti-CD20 antibodies: Y-90 ibritumomab tiuxetan
patients with large B cell lymphoma, RCHOP has been (Zevalin®) which was approved in February 2002, and
associated with a superior PFS and OS with a substantial I-131 tositumomab (Bexxar®) which was approved in
reduction in the risk of death [20, 47, 68, 134, 135]. In June 2003 [32]. In both products the original mouse
the one relatively small randomized trial in mantle cell antibodies ibritumomab tositumomab were kept as
that has been published, RCHOP was superior to CHOP murine proteins in order to decrease half-life in the cir-
in terms of response rate (94% versus 75%) complete culation to reduce non-specific total body irradiation.
response rate (34% versus 7%), with a 50% longer median Ibritumomab is the same antibody that was modified to
PFS (21 versus 14 months), but with no difference in create the chimeric antibody rituximab; therefore, it has
survival [106]. exactly the same binding to CD20 as rituximab. The
As discussed in Chapter 10, the role of maintenance clinical trial results for these two products are summa-
rituximab remains controversial. In indolent lympho- rized in Table 4. As expected, in phase I trials the dose
mas four randomized trials utilizing a variety different limiting toxicities of these agents were cytopenias sec-
maintenance schedules, have shown that administration ondary to bone marrow suppression with nadirs 5 to 8
of additional doses or courses of rituximab clearly pro- weeks after treatment, but both produced objective
longs progression free survival after initial treatment response rates of 65–70% with durabilities of 9 to 12
with rituximab alone in untreated patients [64], after months [87, 176]. Both of these products have produced
rituximab alone in patients who have relapsed after prior response rates of about 40% with durabilities of about
chemotherapy [64, 70], and after chemotherapy and 1-year in patients with transformed B-cell lymphomas
rituximab in patients who had relapsed after prior that had been heavily treated with prior chemotherapy.
chemotherapy [56, 169]. However, none of these trials Both products have produced response rates of about
have established a survival benefit. The only published 80% with a median duration of response of 11 to 12
randomized trial that has tried to address the issue of months in patients with follicular lymphoma who had
duration of benefit from rituximab (or time to becoming failed prior chemotherapy, but were not refractory to
rituximab refractory, is of uncertain relevance because it Rituximab [27, 89, 178]. Both products produced higher
was conducted in patients who had relapsed after che- response rates that an unconjugated Mab in randomized
motherapy, and they were then treated with rituximab trials, as compared to tositumomab for the I-131 product,
alone. However, the additional rituximab therapy was and to rituximab for the Y-90 product [30, 178]. Both
associated with a higher response rate 52% versus 35%, products produced response rates in over 70% of patients
698 Biological therapy of B and T cell lymphoproliferative disorders

Table 4. Radioimmunotherapy of B cell lymphoma: radiolabeled anti-CD20 antibodies


131-I tositumomab (Bexxar) 90-Y-ibritumomab tiuxetan (Zevalin)
Dose Limiting Toxicity Bone marrow suppression Bone marrow suppression
Therapeutic Dose 75 cGy total body dose 0.4 mCi/kg (max 32 mCi)
Phase I response rate 71% 67%
Response rate in relapsed follicular 81% 86%
lymphoma
Response rate in rituximab refractory 70% 74%
lymphoma
Response rate in transformed lymphoma 39% 56%
RIT versus unlabeled Moab 67% versus 28% (tositumomab) 80% versus 56% (rituximab)

with indolent B-cell lymphomas that had become refrac- of these products will be as consolidation treatment after
tory to Rituximab, defined as either no response or induction therapy [107, 138, 183], or as part of marrow
progression with 6 months after a response [82, 177]. ablative therapy and hematopoietic stem cell rescue
Long term follow up has confirmed the remissions in [121, 124, 170, 171].
these various settings are usually quite durable [50, 76,
175, 180]. In a young cohort (median age 49 years) of
76 patients with previously untreated follicular lym-
Immunotoxins
phoma, treatment with I-131 tositumomab produced a Denileukin diftitox, an immunotoxin consisting of IL-2
response rate of 95% with a 75% complete response fused to diphtheria toxin, is approved for treatment of
rate, and 60% molecular complete response rate [88]. At CD-25 expressing cutaneous T-cell lymphomas (CTCL).
a median follow-up of greater than 5 years, the 5-year The IL-2 receptor is also expressed on many B cell
progression-free survival rate was 59%, and the median malignancies; so denileukin diftitox has been tested in
progression-free survival was projected to be 6.1 years. patients with relapsed indolent lymphoma. In 45 patients
Seventy percent of the 57 complete responders were with progressive B cell lymphoma, all of whom had
still in remission 4 to 8 years following therapy. been treated with rituximab in the past, there was a 24%
Despite these tremendous results, these two radiola- response rate with a median duration of 7 months [24].
beled products have struggled in the marketplace A second phase II trial was aborted because of slow
because of the reluctance of medical oncologists to refer accrual after 35 of a planned 77 patients had been
their patients for this treatment. One reason for this is enrolled [99]. Objective responses were observed in
the limited numbers of sites able and willing to provide 3/29 evaluable patients, 1/21 with follicular lymphoma
therapy, so that often patients have to travel long dis- and 2/8 with small lymphocytic lymphoma. In both of
tances seeking such treatment. The treatment itself these trials response rates were similar regardless of
requires some complex coordinated planning. Many whether the lymphoma tested positive or negative for
hospitals have kept these products off their formularies the IL-2 receptor.
because of the high single cost of the radioisotope and The combination of denileukin diftitox and rituximab
the financial risks associated with fixed payments. Since produced a response rate of 32% in 38 evaluable
these treatments have to be delivered by physicians with patients, 30 (80%) were rituximab-refractory [25]. The
radiation credentials, medical oncologists do not share overall response rate (ORR) was 32% and median dura-
in the profit margin associated with such treatment in tion of response was 8 months. Of note 6/11 patients
the way that they do with the administration of chemo- with rituximab-refractory follicular lymphoma had an
therapy and other intravenous products. Because there objective response.
are so many systemic agents available for lymphoma,
physicians can easily rationalize that they should try all
other therapies before considering referral for radioim-
Vaccines
munotherapy. Despite the high response rates, to date As of early 2008 there were no vaccines approved for
there are no trials directly comparing radioimmunother- use in any B cell malignancy, although three products,
apy plus rituximab to chemotherapy plus rituximab, MyVaxID, FavID, and BiovaxID were being evaluated
although it is likely efficacy would be similar and qual- in pivotal regulatory trials [51, 145, 184]. Initial investi-
ity of life superior in the radioimmunotherapy arm. At gation established that vaccination with an individual
this time it appears that the most acceptable application patient’s idiotype protein could induce an endogenous
Robert O. Dillman 699

idiotype response [100]. In a trial of 41 patients with longer PFS. It is possible the dendritic cells pulsed with
indolent B cell lymphoma, vaccination with an idiotype- idiotype protein may be a more powerful vaccination
keyhole limpet hemocyanin (KLH) product produced approach [147, 193].
anti-idiotype responses in 49% of patients [83]. For the
subset of 32 patients who were in their first remission,
PFS was much longer in those patients who exhibited an
immune response compared to those who did not (7.9 T Cell Lymphoproliferative
versus 1.3 years, p = .0001). The vast majority of Malignancies
patients in these trials had follicular lymphoma, in part
because of the inherent difficulty in isolating the idiotype Even though T cell malignancies account for less than
from other B cell malignancies [149]. In a trial of 33 10% of the malignant lymphoproliferative disorders,
consecutive patients with relapsed follicular lymphoma, they have been the object of many trials of biological
who had relapsed after an initial complete response and therapy. Most of the trials or biologics in T cell lym-
were induced into a second remission with CHOP, an phoma have been in mycosis fungoides cutaneous T cell
idiotype vaccine was able to be prepared for 83% [74]. lymphoma (CTCL) with predominantly patients who
Overall 80% of the 25 treated patients exhibited some were in the Sezary syndrome stage of disease which fea-
sort of a cellular idiotype-specific response. All 20 tures a characteristic desquamative rash and circulating
responders had a longer disease free interval after their malignant T cells. The single-agent activities of different
second remission than their first. A cellular idiotype- biologicals in the T cell lymphoproliferative diseases are
specific response was detected in 18/25 (72%) while a summarized in Table 6.
humoral idiotype-specific response was demonstrated in
13 patients (52%). In another trial of an idiotype-KLH
Interferon
product co-administered with GM-CSF, objective tumor
responses were noted in 4/32 previously treated indo- As a single agent interferon alpha produced response
lent lymphoma patients [144]. Encouraging results such rates ranging from 40% to 60% in patients with CTLC
as these have attracted commercial interest, and at pres- with most of the responses observed in patients with
ent there are at least anti-idiotype vaccine products that mycosis fugoides [10, 39, 131]. Many trials were con-
have entered into phase III trials, all of which are being ducted with IFN-α in combination with other therapeutic
conducted in patients with follicular lymphoma. All modalities. A response rate of 83% was reported for the
three utilize the idiotype paraprotein of the malignant B combination of IFN-α and psolaren with ultraviolet light
cell clone as the immunogen to produce an endogenous A (PUVA) and (IFN-α2a) in 63 symptomatic early-stage
anti-idiotype response as summarized in Table 5. It has CTCL patients, with most of the responses being CRs
been announced that the Genitope trial did not meet the [18]. In another trial of IFN-α and PUVA in 25 patients
necessary regulatory endpoints, but those patients who with early-stage CTCL, 91% of patients had an objective
were able to mount an anti-idiotype response did have a response with 76% achieving a CR [152]. In two trials

Table 5. Anti-idiotype vaccines in follicular lymphoma


MyVaxID (Genitope) FavID (Favrille) BiovaxID (Biovest)
Vaccine type Id-KLH/GM-CSF Id-KLH/GM-CSF Id-KLH/GM-CSF
Production Method Recombinant DNA Recombinant DNA Hybridoma rescue
Disease Setting Untreated Untreated or Relapsed Untreated
Induction therapy CVP Rituximab PACE or RCHOP
Eligibility CR or PR CR or PR or SD CR only
Accrual Finished Finished In progress

Id = idiotype
KLH = keyhole limpet hemocyanin
GM-CSF = granulocyte-macrophage colony stimulating factor
CVP = cyclophosphamide, vincristine, prednisone
PACE = platinum, doxorubicin, cyclophosphamide, and etoposide
RCHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone
CR = complete response
PR = partial response
700 Biological therapy of B and T cell lymphoproliferative disorders

Table 6. Biotherapy of T cell lymphoma with single agent biologicals


Modality Class Biological agent Reference Disease Patients Response rate
Cytokine Interferon-α [10] CTCL 20 45%
Cytokine Interferon-α [131] CTCL 22 59%
Cytokine Interferon-α [39] CTCL 45 38%
Cytokine Interleukin-2 [65] CTCL 7 71%
Cytokine Interleukin-2 [140] CTCL 22 18%
Immunotoxin Denileukin diftitox [130] CTCL 71 30%
Immunotoxin Denileukin diftitox [104, 153] CTCL 35 37%
Immunotoxin Denileukin diftitox [25] PTCL 27 48%
Mab antiCD4 [97] CTCL 8 62%
Mab antiCD5 [35] CTCL 10 40%
Mab Y90 – antiCD5 [60] CTCL 8 38%
Mab Alemtuzumab [108] PTCL 8 50%
Mab Alemtuzumab [45] PTCL 14 36%
Mab Alemtuzumab [5] CTCL 14 86%
Mab Alemtuzumab [110] CTCL 22 55%
Mab Alemtuzumab [93] CTCL 8 38%
Retinoid Isotretinoin [94] CTCL 25 44%
Retinoid Isotretinoin [120] CTCL 39 59%
Retinoid Isotretinoin [16] PTCL 12 50%
Retinoid Etretinate [120] CTCL 29 67%
Retinoid Bexarotene [42] CTCL 94 49%

CTCL = cutaneous T cell lymphoma


PTCL = peripheral T cell lymphoma

s.c. IFN-α was combined with extracorporeal photo- anthracycline-resistant peripheral T-cell lymphoma
chemotherapy (photopheresis) using oral 8-methoxypso- (PTCL) who were treated with thrice weekly s.c. IFN-
ralen as the photosensitizer. A response rate of 90% was α2a and daily oral 13-cis retinoic acid (isotretinoin) [84].
observed in 39 patients, most of whom had early-stage A response rate of 100% was reported for 28 patients
CTCL, with 24 achieving a CR [98]. In the second trial, with Sezary syndrome who were treated with extracor-
which was limited to 14 patients with stage II CTCL, a poreal photochemotherapy and two or more of various
response rate of only 50% was observed [181]. IFN-α biologicals including IFN-α, retinoids, and GM-CSF
has combined with a variety of retinoids to treat patients [146]. One small randomized trial compared thrice
with T cell lymphomas. In an early report all seven weekly 9 MU IFNα-2a plus PUVA to 9 MU IFNα-2a
patients treated with IFN-α and etretinate had a response plus acitretin in 98 patients with stage I and II CTCL,
[161]. In another small study there was an 83% CR rate only 82 of whom were considered evaluable for response
in 12 patients with CTCL who received IFN-α and acit- [159]. IFNα-2a plus PUVA was not only less toxic, but
retin [2]. In a more complex trial 45 patients, 13 of whom was associated with more rapid response, and a complete
had stage I Sezary syndrome and the other 32 mycosis response rate of 70% compared to only 38% for IFNα-2a
fungoides, were treated with 3 months of daily s.c. IFN-α plus acitretin.
alone, and responders then continued to received thrice IFNα has also been combined with chemotherapy in
weekly s.c. IFN-α, while non-responders were treated the treatment of patients with T cell lymphoma. The
with thrice weekly s.c. IFN-α plus etretinate [39]. After purine analogs fludarabine and pentostatin have sub-
1 year 28 patients (62%) were in remission. IFN-α alone stantial single-agent activity in CTCL. A response rate
produced a response in 17 patients (38%). Among the 28 of 41% was observed in a trial of 41 CTCL patients,
patients who had not responded after 3 months of IFN-α, only six of whom had not received prior therapy, and 29
11 (39%) eventually achieved a response after changing of whom had not responded to prior therapy, who were
to IFN-α plus etretinate. The combination of s.c. IFN-α treated with pentostatin 4 mg/m2 i.v. days 1 through 3
and the oral synthetic retinoid bexarotene produced a and IFNα 10 MU/m2 i.m. on day 22, and 50 MU/m2 i.m.
response rate of 39% among 18 CTCL patients, which 23 through 26 [58]. A response rate of 51% was observed
appeared no better than either agent used alone [160]. A in 35 patients, 10 of whom had received pentostatin pre-
response rate of 31% was noted for 17 patients with viously and 21 of whom had not responded to prior
Robert O. Dillman 701

therapy, who were treated with fludarabine 25 mg/m2 i.v. kin diftitox yielded a response rate of 67% among 14
days 1 to 5 every 28 days with IFNα 5 MU/m2 s.c. thrice patients with relapsed or refractory CTCL [57].
weekly for up to eight cycles [59].
Immunotoxins
Interleukin-2 Denileukin diftitox (Ontak™) is a genetically engineered
There has been limited experience with IL-2 in the T fusion protein that combines the active chain of diphthe-
cell malignancies. In one trial that utilized continuous ria toxin to the cytokine IL-2 which binds to the CD25
i.v. infusion of relatively high doses of IL-2, responses receptor full-length sequence for interleukin-2 (IL-2).
were noted in 5/7 patients [65]. In a recent trial of s.c. This immunotoxin has therapeutic potential for any
IL-2 a response rate of only 18% was reported for 22 malignancy, including B cell lymphomas and Hodgkin’s
heavily pretreated CTCL patients [140]. disease, in which there is over-expression of CD25, or
more specifically, the subunit of the IL-2 receptor.
Denileukin diftitox was approved by the US FDA based
Retinoids on its activity in cutaneous T cell lymphoma where
The vitamin analogs collectively known as the retinoids, response rates 30% to 40% were achieved in trials [104,
have been shown to modulate proliferation and differen- 130, 153]. Response rates in peripheral T cell lymphomas
tiation in premalignant and malignant cells, and also may be even higher [26] This product may be useful in
have immunologic effects. The retinoid response is any CD25 overexpressing malignancy, which includes
mediated by nuclear receptors, which have been charac- some cases of B-cell lymphoma and Hodgkin’s disease
terized as retinoic acid receptors (RARs) and retinoid in addition to most T cell malignancies.
“X” receptors (RXRs). Because of the dramatic effects
of retinoids in skin disorders including acne and aging
of the skin, there has been intense investigation of retin-
Monoclonal Antibodies
oids in the treatment of CTCL, especially in early stage The humanized anti-CD52 Mab Alemtuzumab
mycosis fungoides and Sezary syndrome, and also in (Campath®) was approved in May 2001 based on data
PTCL. As summarized in Table 6, as single-agents retin- submitted for the treatment of patients with CLL that
oids have produced response rates ranging from 50% to had recurred or been refractory to the purine analog flu-
70% in CTCL, depending on the product and the spe- darabine. As shown in Table 6, alemtuzumab has exhib-
cific CTCL patient population [42, 43, 94, 120], and ited significant activity as a single agent in both CTCL
50% in PTCL [16]. Substantial anti-tumor activity was with where response rates ranged from 38% to 86% [5,
demonstrated with cis-retinoic acid (isotretinoin) and 45, 110], and in PTCL where response rates ranged from
related retinoids, but none of these agents were ever 36% to 50% in two small studies [93, 108]. Alemtuzumab
submitted for regulatory approval for marketing these has also been combined with chemotherapy in the treat-
agents in T cell malignancies. ment of PTCL with very encouraging results. In an
The retinoid X receptor-selective agent LGD1069 was Italian trial the combination of CHOP chemotherapy
given orally by mouth to 52 patients in an dose escalation and s.c. alemtuzumab yielded a response rate of 75% in
trial in which some tumor response were observed at higher 24 patients with 17 of the 18 responses recorded as CRs
doses [119]. This product became bexarotene (Targretin [62]. In a Korean trial in 20 PTCL patients the combina-
capsules; Ligand Pharmaceuticals Incorporated, San tion of CHOP chemotherapy and i.v. alemtuzumab was
Diego, Calif), which received regulatory approval based associated with a response rate of 80% with 13 of the 16
on the activity of this agent in early-stage cutaneous T-cell responses being CRs [96].
lymphoma at doses of 300 mg/m2 per day [42, 43].
Hypertriglyceridemia and hypothyroidism were the major
adverse events noted. High response rates have also been
Vaccines
observed with bexarotene gel [9, 139]. Interestingly, a sin- A vaccine consisting of dendritic cells loaded with the
gle institution retrospective comparison of all-trans retinoic lysate from CTCL cells and KLH was given as weekly
acid (RAR-specific) and bexarotene (RXR-specific) in intranodal injections in ten patients with CTCL [111].
patients with relapsed mycosis fungoides/Sézary syndrome Tumor-specific delayed-type hypersensitivity (DTH)
detected no substantial differences between the products in reactions to tumor lysate were observed in 3/8 patients
terms of duration of disease control, although the response so tested. There was a 50% objective response rate
rate was higher for bexarotene (21% versus 12%) [140]. including one CR and four PRs with all of the response
The combination of bexarotene and immunotoxin denileu- occurring in patients who had a low tumor burden.
702 Biological therapy of B and T cell lymphoproliferative disorders

rituximab with cyclophosphamide and a purine analog,


Summary quickly attracted the interest of investigators, and enthu-
Although T cell malignancies are relatively rare, they have siasm for pursuing IFN-α in CLL waned.
been the target of many biological therapies. Interferon-
alpha, the retinoid bexarotene, the immunotoxin denileu-
kin diftitox, and the Mab alemtuzumab are all routinely
Monoclonal Antibodies
used in the treatment of these malignancies both as single A summary of clinical trials of Mab in CLL are shown
agents, and increasingly in combination therapies. in Table 7. Some of the earliest trials with Mabs were
conducted in patients with CLL. The anti-CD5 murine
Mab T101 bound to CLL cells and decreased circulating
lymphocyte numbers, but at the doses given, and with
Chronic Lymphocytic Leukemia the murine construct, was not able to induce sustained
responses [33–35, 53]. Trials were never carried out at
Interferon
higher doses or with humanized constructs.
Early U.S. trials using leukocyte-derived interferon and In May 2001 the anti-CD52 monoclonal antibody
recombinant interferon in small numbers of patients alemtuzumab (Campath) became the second Mab
with chronic lymphocytic leukemia (CLL) were associ- approved by the U.S. FDA for a hematologic malignancy
ated with non-durable response rates of 11% to 15% when it was granted regulatory approval based on its
[52]. However a Greek trial reported objective responses activity in patients with CLL whose disease had recurred
in 10/26 (38%) [7], and a British trial reported responses or been refractory to the purine analog fludarabine [91].
in 8/18 (44%), although all were transient [118]. Other Trials conducted in the fludarabine resistant or refractory
trials focused on IFN-α as maintenance therapy follow- setting were associated with response rates ranging from
ing chemotherapy. A small randomized trial of Italian 45 33% to 55% with a median of 40% [23, 91, 123, 132,
patients showed a significantly longer duration of 142, 143]. In 115 patients who had progressive disease
response and fewer infections in patients who received after multiple course of prior chemotherapy, the response
IFN-α maintenance therapy [46]. However investiga- was still 23% [48] In the untreated setting alemtuzumab
tors who conducted a single-arm U.S. trial of mainte- produced responses of 83% to 87% when given by the
nance therapy in 31 patients who had responded to i.v. or s.c. route [109, 187]. In a European registration
fludarabine, concluded that adding IFN-α did not trial 297 previously untreated CLL patients were ran-
enhance the degree of chemotherapy-induced remis- domized to oral chlorambucil 40 mg/m2 monthly versus
sion, and that the time to progression was similar to a standard i.v. alemtuzumab [80]. The Mab produced
historical control group [127]. In a German trial in superior response rate, 83% versus 55% (p < .0001) and
which 44 high-risk patients were randomized to obser-
vation or IFN-α, there was no difference in PFS or OS
Table 7. Single-agent activity of monoclonal antibodies in CLL
[102]. In the largest trial of IFN-α in CLL, 133 previ-
ously untreated patients were randomized to receive flu- Antibody Reference Setting Patients Response
rate
darabine and prednisone with or without interferon, and
then 78 responders were randomized to observation ver- Anti-CD5 [35] Relapsed 10 0%
Anti-CD5 [53] Relapsed 13 0%
sus maintenance interferon [115]. The initial response Alemtuzumab [109] Untreated 38 87%
rates were similar (84% and 86%) suggesting that IFN-α Alemtuzumab [80] Untreated 149 83%
added nothing over chemotherapy alone. There was a Alemtuzumab [123] Relapsed 91 55%
longer response duration in patients in patients who Alemtuzumab [23] Relapsed 16 50%
received maintenance therapy, however, this difference Alemtuzumab [142] Relapsed 136 40%
Alemtuzumab [132] Relapsed 29 38%
could be explained by an imbalance in the distribution Alemtuzumab [91] Relapsed 93 33%
of patients who were in CR at that the time they entered Alemtuzumab [143] Relapsed 24 33%
the maintenance phase of the study. Although these were Alemtuzumab [48] Relapsed 115 23%
all relatively small studies by the standards of modern Rituximab [162] Untreated 19 90%
clinical trial design, the failure of IFN-α to consistently Rituximab [78] Untreated 26 70%
Rituximab [79] Untreated 43 58%
achieve durable disease control, and the toxicity associ- Rituximab [12] Relapsed 33 45%
ated with IFN-α, dampened enthusiasm for larger studies. Rituximab [128] Relapsed 40 36%
Furthermore, the high durable response rates achieved Rituximab [188] Relapsed 23 35%
with combinations of rituximab with fludarabine, or Rituximab [73] Relapsed 28 25%
Robert O. Dillman 703

PFS which led to a marketing indication as initial ther- community setting [36]. In a trial of young patients with
apy, even though in the U.S. purine-analog based therapy relapsed CLL, cladribine, cyclophosphamide, and ritux-
is considered the treatment of choice. imab (CCR) produced a response rates of 58% [148].
Fludarabine and alemtuzumab combination therapy as In one randomized phase II trial, concurrent fludarabine
the initial treatment of patients with CLL produced plus rituximab was compared to the sequence of fludara-
responses in 83% of 36 patients, with 11 of the 30 bine followed by rituximab in 104 patients [13]. The
responses being CR [44]. Combining GM-CSF with response rate was higher for concurrent therapy (90% ver-
alemtuzumab in 14 patients with relapsed disease did not sus 77%) as was the complete response rate (47% vs
appear to increase the response rate (36%) or decrease 28%). A retrospective comparison of these patients to a
the risk of infection [189]. Because of the additive toxic- similar population of patients treated with fludarabine
ity and immuosuppression associated with combining alone at the same dose in an earlier randomized trial sug-
alemtuzumab with chemotherapy, most of the recent gested that patients treated with the combination of flu-
emphasis has been on using alemtuzumab after chemo- darabine plus rituximab had much better outcomes,
therapy in an effort to eradicate minimal residual disease including a higher response rate (84% versus 63%), higher
in responding CLL patients. Thrice weekly doses of 10 complete response rate (38% versus 20%) better progres-
to 30 mg have been administered i.v. or s.c. for 4 to 12 sion free survival after 2 years (67% versus 45%), and bet-
weeks as a consolidative therapy [122, 129, 173]. These ter overall survival at 2 years (93% versus 81%) [14].
studies have shown that consolidation with alemtuzumab
does increase clinical, phenotypic, and molecular com-
plete response rates, but there is an increased risk of
opportunistic infections, even if anti-viral, anti-bacterial,
Summary
and anti-Pneumocystis prophylaxis is used. Biotherapy now plays a crucial and ever increasing role in
Although rituximab does not have regulatory approval the management of both B-cell and T cell lymphoprolif-
for marketing as a treatment for CLL, it is widely used erative malignancies. Because of its ease of delivery,
in combination with chemotherapy. As shown in Table 7, anti-tumor activity and relatively low toxicity rate,
in patients with previously treated, relapsed CLL, as a rituximab has become the most important single agent
single agent rituximab was associated with response in the treatment of indolent B cell lymphoma., and com-
rates ranging from 25% to 45% with a median of 35%. bined with chemotherapy, has become the standard of
over a wide range of doses and by various schedules of treatment for more aggressive B cell lymphomas.
administration [12, 73, 128, 188]. Response rates were Rituximab alone or in combination with chemotherapy
much higher in patients who had not been previously has become the initial treatment of choice for indolent
treated with a range from 58% to 90% with the highest lymphomas, and CHOP + Rituximab has become the
rate occurring in patients with early stage disease who treatment of choice for large B cell lymphoma. Despite
were considered to be high risk. [78, 79, 162]. evidence-based data supporting the use of interferon-α
The popularity of rituximab in CLL is due to the abil- in lymphoma, because of its toxicity and need for
ity to combine it with chemotherapy with no apparent chronic treatment, this biological never gained wide-
increase in toxicity. Rituximab combined with purine spread acceptance in the treatment of lymphoma in the
analogs has produced response rates of about 90% in U.S. IL-2 was never an important agent in these disor-
untreated patients treated with rituximab and fludara- ders because of the need for hospitalization and toxicity
bine [12, 154], and 33% in previously treated patients associated with higher doses, and the lack of clinical
who received pentostatin plus rituximab [38]. The com- activity at lower doses. Two biological agents, the
binations of purine analogs plus cyclophosphamide plus immunotoxin denileukin diftitox and the retinoid bex-
rituximab have been associated with very high response arotene were both received regulatory approval based
rates and durable complete remissions in previously on studies conducted in cutaneous T cell lymphoma.
untreated patients. In single institution trials the combi- The use of denileukin difitox is limited by its immuno-
nation of fludarabine, cyclophosphamide, and rituximab genicity. The antiCD52 Mab alemtuzumab is being used
(FCR) has produced response rates of 77% in the increasingly in combination with chemotherapy for the
relapsed setting [174], and 95% as initial therapy [92]. T cell malignancies, but its immune inhibition and tox-
The combination of pentostatin, cyclophosphamide, and icity profile make it a less attractive option in B cell
rituximab (PCR) has produced response rates of 75% to lymphoma because of the availability of rituximab.
90% in previously untreated patients [36, 90, 101], but Vaccine approaches are interesting and promising, but
only 33% in previously treated elderly patients in the at this time there is no commercial product.
704 Biological therapy of B and T cell lymphoproliferative disorders

19. Coiffier B, Haioun C, Ketterer N, et al. Rituximab (anti-CD20


References monoclonal antibody) for the treatment of patients with relapsing
1. Allison MA, Jones SE, McGuffey P. Phase II trial of outpatient or refractory aggressive lymphoma: a multicenter phase II study.
interleukin-2 in malignant lymphoma, chronic lymphocytic leuke- Blood 1998;92:1927–1932.
mia, and selected solid tumors. J Clin Oncol 1989; 7:75–80. 20. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus
2. Aviles A, Guzman R, Garcia EL, Maqueo JC. Biological modifiers rituximab compared with CHOP alone in elderly patients with dif-
(etretinate (changed from etetrinate) and alfa 2a) in the treatment of fuse large-B-cell lymphoma. N Engl J Med 2002;346:235–42.
refractory cutaneous T-cell lymphoma. Cancer Biother Radiopharm 21. Colombat P, Salles G. Brousse, et al. Rituximab (anti-CD20 mono-
1996;11:21–4. clonal antibody) as single first-line therapy for patients with folli-
3. Aviles A, Leon MI, Diaz-Maqueo JC, et al. Rituximab in the treat- cular lymphoma with a low tumor burden: clinical and molecular
ment of refractory follicular lymphoma – six doses are better than evaluation. Blood 2001;97:101–6.
four. J Hematother Stem Cell Res 2001;10:313–6. 22. Conconi A, Martinelli G, Thieblemont C, et al. Clinical activity of
4. Aviles A, Neri N, Huerta-Guzman J, et al. Interferon alpha 2b as rituximab in extranodal marginal zone B-cell lymphoma of MALT
maintenance therapy improves outcome in follicular lymphoma. type. Blood. 2003;102:2741–5.
Leuk Lymphoma. 2004;4:2247–51. 23. Cortelezzi A, Pasquini MC, Sarina B, et al. A pilot study of low-
5. Bernengo MG, Quaglino P, Comessatti A, et al. Low-dose intermittent dose subcutaneous alemtuzumab therapy for patients with chemo-
alemtuzumab in the treatment of Sézary syndrome: clinical and immu- therapy-refractory chronic lymphocytic leukemia. Haematologica
nologic findings in 14 patients. Haematologica 2007;92:784–94. 2005;90:410–2.
6. Bernstein ZP, Vaickus L, Friedman N, et al. Interleukin-2 lym- 24. Dang NH, Hagemeister FB, Pro B, et al. Phase II study of denileu-
phokine-activated killer cell therapy of non-Hodgkin’s lymphoma kin diftitox for relapsed/refractory B-Cell non-Hodgkin’s lym-
and Hodgkin’s disease. J Immunother 1991;10:141–146. phoma. J Clin Oncol 2004;22:4095–102.
7. Boussiotis VA, Pangalis GA. Interferon alfa-2b therapy in untreated 25. Dang NH, Fayad L, McLaughlin P, et al. Phase II trial of the com-
early stage, B-chronic lymphocytic leukaemia patients: one-year bination of denileukin diftitox and rituximab for relapsed/refractory
follow-up. Br J Haematol 1991;79 Suppl 1:30–3. B-cell non-Hodgkin lymphoma. Br J Haematol 2007;138:502–5.
8. Bremer K. Semi-extended, six weekly rituximab infusions in pre- 26. Dang NH, Pro B, Hagemeister FB, et al. Phase II trial of denileu-
treated advanced low-grade B cell non-Hodgkin’s lymphoma: a phase kin diftitox for relapsed/refractory T-cell non-Hodgkin lymphoma.
II study. Anticancer Drugs 2003;14:809–15. Br J Haematol 2007;136:439–47.
9. Breneman D, Duvic M, Kuzel T et al. Phase 1 and 2 trial of bexaro- 27. Davies AJ, Rohatiner AZ, Howell S, et al. Tositumomab and iodine
tene gel for skin-directed treatment of patients with cutaneous I 131 tositumomab for recurrent indolent and transformed B-cell
T-cell lymphoma. Arch Dermatol 2002;138:325–32. non-Hodgkin’s lymphoma. J Clin Oncol 2004;22:1469–79.
10. Bunn PA, Jr., Ihde DC, Foon KA. The role of recombinant inter- 28. Davis TA, Czerwinski DK, Levy R. Therapy of B-cell lymphoma
feron alfa-2a in the therapy of cutaneous T-cell lymphomas. Cancer with anti-CD20 antibodies can result in the loss of CD20 antigen
1986; 57:1689–1695. expression. Clin Cancer Res 1999;5:611–5.
11. Buske C, Dreyling MH, Eimermacher H et al. Combined immuno- 29. Davis TA, Grillo-Lopez AJ, White CA, et al. Rituximab anti-CD20
chemotherapy (R-CHOP) results in superior response rates and monoclonal antibody therapy in non-Hodgkin’s lymphoma: safety
time to treatment failure in first line of patients with lymphoplas- and efficacy of re-treatment. J Clin Oncol 2000;18:3135–43.
macytoid/immunocytoma. Blood 2004;104:50a [abstract 162]. 30. Davis TA, Kaminski MS, Leonard JP, et al. The radioisotope con-
12. Byrd JC, Murphy T, Howard RS, et al. Rituximab using a thrice tributes significantly to the activity of radioimmunotherapy. Clin
weekly dosing schedule in B-cell chronic lymphocytic leukemia Cancer Res 2004;10:7792–8.
and small lymphocytic lymphoma demonstrates clinical activity 31. Davis T, White CA, Grillo-Lopez A, et al. Single-agent monoclo-
and acceptable toxicity. J Clin Oncol 2001;19:2153–64. nal antibody efficacy in bulky non-Hodgkin’s lymphoma: results
13. Byrd JC, Peterson B, Morrison VA, et al. Randomized phase 2 of a phase II trial of rituximab. J Clin Oncol 1999;17:1851–1857.
study of fludarabine with concurrent versus sequential treatment 32. Dillman RO. Radiolabeled anti-CD20 monoclonal antibodies for
with rituximab in symptomatic, untreated patients with B-cell the treatment of B-cell lymphoma. J Clin Oncol 2002;20:3545–57.
chronic lymphocytic leukemia: results from Cancer and Leukemia 33. Dillman RO, Shawler DL, Sobol RE, et al. Murine monoclonal
Group B 9712 (CALGB 9712). Blood 2003;101:6–14. antibody therapy in two patients with chronic lymphocytic leuke-
14. Byrd JC, Rai K, Peterson BL, et al. Addition of rituximab to flu- mia. Blood 1982; 59:1036–45.
darabine may prolong progression-free survival and overall sur- 34. Dillman RO, Shawler DL, Dillman JB, et al. Therapy of chronic
vival in patients with previously untreated chronic lymphocytic lymphocytic leukemia and cutaneous T-cell lymphoma with T101
leukemia: an updated retrospective comparative analysis of monoclonal antibody. J Clin Oncol 1984; 2:881–91.
CALGB 9712 and CALGB 9011. Blood 2005;105:49–53. 35. Dillman RO, Beauregard J, Shawler DL et al. Continuous infusion of
15. Case DC Jr, Ebrahim K, Hedlund J, et al. Rituxiab in relapsed fol- T101 monoclonal antibody in chronic lymphocytic leukemia and
licular lymphoma. Blood 2002;100:292b [abstract #4704]. cutaneous T-cell lymphoma. J Biol Response Mod 1986; 5:394–410.
16. Cheng AL, Su IJ, Chen CC, et al. Use of retinoic acids in the treat- 36. Dillman RO, Schreeder MT, Hon JK, et al. Community-based
ment of peripheral T-cell lymphoma: a pilot study. J Clin Oncol phase II trial of pentostatin, cyclophosphamide, and rituximab
1994; 12:1185–1192. (PCR) biochemotherapy in chronic lymphocytic leukemia and
17. Cheson BD, Sorenson JM, Vena DA, et al. Treatment of hairy cell small lymphocytic lymphoma. Cancer Biother Radiopharm 2007;
leukemia with 2-chlorodeoxyadenosine via the Group C protocol 22:185–93.
mechanism of the National Cancer Institute: a report of 979 37. Dimopoulos MA, Zervas C, Zomas A, et al. Treatment of
patients. J Clin Oncol 1998;16:3007–15. Waldenstrom’s macroglobulinemia with rituximab. J Clin Oncol
18. Chiarion-Sileni V, Bononi A, Fornasa CV et al. Phase II trial of 2002;20:2327–33.
interferon-alpha-2a plus psolaren with ultraviolet light A in patients 38. Drapkin R, Di Bella NJ, Cuasay LC, et al. Phase II multicenter trial
with cutaneous T-cell lymphoma. Cancer 2002;95:569–75. of pentostatin and rituximab in patients with previously treated or
Robert O. Dillman 705

untreated chronic lymphocytic leukemia. Blood 2002;100:803a previously treated mantle-cell lymphoma, immunocytoma, and
[abstract 3171]. small B-cell lymphocytic lymphoma. J Clin Oncol 2000;18:
39. Dreno B, Claudy A, Meynadier J, et al. The treatment of 45 patients 317–24.
with cutaneous T-cell lymphoma with low doses of interferon- 55. Forstpointner R, Dreyling M, Repp R, et al. The addition of ritux-
alpha 2a and etretinate. Br J Dermatol 1991;125:456–459. imab to a combination of fludarabine, cyclophosphamide, mitox-
40. Dreyling M, Lenz G, Hoster E, et al. Early consolidation by antrone (FCM) significantly increases the response rate and
myeloablative radiochemotherapy followed by autologous stem prolongs survival as compared with FCM alone in patients with
cell transplantation in first remission significantly prolongs pro- relapsed and refractory follicular and mantle cell lymphomas:
gression-free survival in mantle-cell lymphoma: results of a pro- results of a prospective randomized study of the German Low-
spective randomized trial of the European MCL Network. Blood Grade Lymphoma Study Group. Blood. 2004;104:3064–71.
2005;105:2677–84. 56. Forstpointner R, Unterhalt M, Dreyling M, et al. Maintenance
41. Duggan DB, Santarelli MT, Zamkoff K, et al. A phase II study of therapy with rituximab leads to a significant prolongation of
recombinant interleukin-2 with or without recombinant interferon- response duration after salvage therapy with a combination of
beta in non-Hodgkin’s lymphoma. A study of the Cancer and rituximab, fludarabine, cyclophosphamide, and mitoxantrone
Leukemia Group B. J Immunother 1992; 12:115–22. (R-FCM) in patients with recurring and refractory follicular and
42. Duvic M, Hymes K, Heald P, et al. Bexarotene is effective and safe mantle cell lymphomas: Results of a prospective randomized study
for treatment of refractory advanced-stage cutaneous T-cell lym- of the German Low Grade Lymphoma Study Group (GLSG).
phoma: multinational phase II-III trial results. J Clin Oncol Blood 2006;108:4003–8.
2001a;19:2456–71. 57. Foss F, Demierre MF, DiVenuti G. A phase-1 trial of bexarotene
43. Duvic M, Martin AG, Kim Y, et al. Phase 2 and 3 clinical trial of and denileukin diftitox in patients with relapsed or refractory cuta-
oral bexarotene (Targretin capsules) for the treatment of refractory neous T-cell lymphoma. Blood 2005;106:454–7.
or persistent early-stage cutaneous T-cell lymphoma. Arch 58. Foss FM, Ihde DC, Breneman DL, et al. Phase II study of pentosta-
Dermatol 2001b; 137:581–93. tin and intermittent high-dose recombinant interferon alfa-2a in
44. Elter T, Borchmann P, Schulz H, et al. Fludarabine in combination advanced mycosis fungoides/Sézary syndrome. J Clin Oncol
with alemtuzumab is effective and feasible in patients with 1992;10:1907–13.
relapsed or refractory B-cell chronic lymphocytic leukemia: results 59. Foss FM, Ihde DC, Linnoila IR, et al. Phase II trial of fludarabine
of a phase II trial. J Clin Oncol 2005;23:7024–31. phosphate and interferon alfa-2a in advanced mycosis fungoides/
45. Englad G, Hagberg H, Erlanson M, et al. A pilot study of alemtu- Sézary syndrome. J Clin Oncol 1994;12:2051–9.
zumab (anti-CD52 monoclonal antibody) therapy for patients with 60. Foss FM, Raubitscheck A, Mulshine JL, et al. Phase I study of the
relapsed or chemotherapy-refractory peripheral T-cell lymphomas. pharmacokinetics of a radioimmunoconjugate, 90Y-T101, in
Blood 2004;103:2920–4. patients with CD5-expressing leukemia and lymphoma. Clin
46. Ferrara F, Rametta V, Mele G, et al. Recombinant interferon-alpha Cancer Res 1998;4:2691–700.
2A as maintenance treatment for patients with advanced stage 61. Friedberg JW, Neuberg D, Gribben JG, et al. Combination immu-
chronic lymphocytic leukemia responding to chemotherapy. Am J notherapy with rituximab and interleukin 2 in patients with
Hematol 1992;41:45–9. relapsed or refractory follicular non-Hodgkin’s lymphoma. Br J
47. Feugier P, Van Hoof A, Sebban C, et al. Long-term results of the Haematol 2002;117:828–34.
R-CHOP study in the treatment of elderly patients with diffuse large 62. Gallamini A, Zaja F, Patti C, et al. Alemtuzumab (Campath-1H)
B-cell lymphoma: a study by the Groupe d’Etude des Lymphomes and CHOP chemotherapy as first-line treatment of peripheral
de l’. Adulte. J Clin Oncol. 2005 Jun 20;23(18):4117–26. T-cell lymphoma: results of a GITIL (Gruppo Italiano Terapie
48. Fiegl M, Falkner A, Hopfinger G, et al. Routine clinical use of Innovative nei Linfomi) prospective multicenter trial. Blood
alemtuzumab in patients with heavily pretreated B-cell chronic 2007;110:2316–23.
lymphocytic leukemia: a nation-wide retrospective study in 63. Gertz MA, Rue M, Blood E, et al. Multicenter phase 2 trial of
Austria. Cancer 2006;107:2408–16. rituximab for Waldenstrom macroglobulinemia (WM): an Eastern
49. Fisher RI, Dana BW, LeBlanc M, et al. Interferon alpha consolida- Cooperative Oncology Group Study (E3A98). Leuk Lymphoma
tion after intensive chemotherapy does not prolong the progres- 2004;45:2047–55.
sion-free survival of patients with low-grade non-Hodgkin’s 64. Ghielmini M, Schmitz SF, Cogliatti SB, et al. Prolonged treatment
lymphoma: results of the Southwest Oncology Group randomized with rituximab in patients with follicular lymphoma significantly
phase III study 8809. J Clin Oncol 2000;18:2010–6. increases event-free survival and response duration compared with
50. Fisher RI, Kaminski MS, Wahl RL, et al. Tositumomab and the standard weekly × 4 schedule. Blood 2004;103:4416–23.
iodine-131 tositumomab produces durable complete remissions in a 65. Gisselbrecht C, Maraninchi D, Pico JL, et al. Interleukin-2 treat-
subset of heavily pretreated patients with low-grade and transformed ment in lymphoma: a phase II multicenter study. Blood 1994;
non-Hodgkin’s lymphomas. J Clin Oncol 2005;23:7565–7573. 83:2081–5.
51. Flowers CR. BiovaxID idiotype vaccination: active immunother- 66. Golomb HM, Ratain MJ, Fefer A, et al. Randomized study of the
apy for follicular lymphoma. Expert Rev Vaccines 2007;6:307–17. duration of treatment with interferon alfa-2B in patients with hairy
52. Foon KA, Bunn PA Jr. Alpha-interferon treatment of cutaneous T cell leukemia. J Natl Cancer Inst 1988;80:369–73.
cell lymphoma and chronic lymphocytic leukemia. Semin Oncol 67. Golomb HM, Dodge R, Mick R, et al. Pentostatin treatment for
1986;13(4 Suppl 5):35–9. hairy cell leukemia patients who failed initial therapy with recom-
53. Foon KA, Schroff RW, Bunn PA, et al. Effects of monoclonal anti- binant alpha-interferon: a report of CALGB study 8515. Leukemia
body therapy in patients with chronic lymphocytic leukemia. 1994;8:2037–40.
Blood 1984; 64:1085–1093. 68. Habermann TM, Weller EA, Morrison VA, et al. Rituximab-CHOP
54. Foran JM, Rohatiner AZ, Cunningham D, et al. European phase versus CHOP alone or with maintenance rituximab in older
II study of rituximab (chimeric anti-CD20 monoclonal antibody) patients with diffuse large B-cell lymphoma. J Clin Oncol
for patients with newly diagnosed mantle-cell lymphoma and 2006;24:3121–7.
706 Biological therapy of B and T cell lymphoproliferative disorders

69. Hagenbeek A, Carde P, Meerwaldt JH, et al. Maintenance of 84. Huang CL, Lin ZZ, Su IJ, et al. Combination of 13-cis retinoic acid
remission with human recombinant interferon alfa-2a in patients and interferon-alpha in the treatment of recurrent or refractory
with stages III and IV low-grade malignant non-Hodgkin’s lym- peripheral T-cell lymphoma. Leuk Lymphoma 2002;43:1415–20.
phoma. European Organization for Research and Treatment of 85. Igarashi T, Ohtsu T, Fujii H, et al. Re-treatment of relapsed indo-
Cancer Lymphoma Cooperative Group. J Clin Oncol 1998; lent B-cell lymphoma with rituximab. Int J Hematol
16:41–47. 2001;73:213–21.
70. Hainsworth JD, Litchy S, Shaffer DW, et al. Maximizing therapeu- 86. Jacobs AD, Champlin RE, Golde DW. Recombinant alpha-2-inter-
tic benefit of rituximab: maintenance therapy versus re-treatment feron for hairy cell leukemia. Blood 1985;65:1017–20.
at progression in patients with indolent non-Hodgkin’s lymphoma 87. Kaminski MS, Zasadny KR, Francis IR, et al. Iodine-131-anti-B1
– a randomized phase II trial of the Minnie Pearl Cancer Research radioimmunotherapy for B-cell lymphoma. J Clin Oncol 1996;14:
Network. J Clin Oncol 2005;23:1088–95. 1974–81.
71. Herold M, Hass A, Srock S, et al. Rituximab added to first-line 88. Kaminski MS, Tuck M, Estes J, et al. 131I-tositumomab therapy as
mitoxantrone, chlorambucil, and prednisolone chemotherapy fol- initial treatment for follicular lymphoma. N Engl J Med 2005;352:
lowed by interferon maintenance prolongs survival in patients with 441–9.
advanced follicular lymphoma: an East German Study Group 89. Kaminski MS, Zelenetz AD, Press OW, et al. Pivotal study of
Hematology and Oncology Study. J Clin Oncol 2007;25: Iodine I 131 tositumomab for chemotherapy-refractory low-grade
1986–92. or transformed low-grade B-cell lymphomas. J Clin Oncol
72. Hiddemann W, Kneba M, Dreyling M, et al. Frontline therapy with 2001;19:3918–28.
rituximab added to the combination of cyclophosphamide, doxo- 90. Kay NE, Geyer SM, Call TG, et al. Combination chemotherapy with
rubicin, vincristine, and prednisone (CHOP) significantly improves pentostatin, cyclophosphamide, and rituximab shows significant
the outcome for patients with advanced-stage follicular lymphoma activity with low accompanying toxicity in previously untreated B
compared with therapy with CHOP alone: results of a prospective chronic lymphocytic leukemia. Blood 2007;109:405–411.
randomized study of the German Low-Grade Lymphoma Study 91. Keating MJ, Flinn I, Jain V, et al. Therapeutic role of alemtuzumab
Group. Blood 2005;106:3725–32. (Campath-1H) in patients who have failed fludarabine: results of a
73. Huhn D, von Schilling C, Wilhelm M, et al Rituximab therapy of large international study. Blood 2002;99:3554–61.
patients with B-cell chronic lymphocytic leukemia. Blood 92. Keating MJ, O’Brien S, Albitar M, et al. Early results of a
2001;98:1326–31. chemoimmunotherapy regimen of fludarabine, cyclophosphamide,
74. Inoges S, Rodriguez-Calvillo M, Zabalegui N, et al. Clinical ben- and rituximab as initial therapy for chronic lymphocytic leukemia.
efit associated with idiotypic vaccination in patients with follicular J Clin Oncol 2005;23:4079–88.
lymphoma. J Natl Cancer Inst 2006;98:1292–301. 93. Kennedy GA, Seymour JF, Wolf M, et al. Treatment of patients
75. Golomb HM, Jacobs A, Fefer A et al. Alpha-2 interferon therapy with advanced mycosis fungoides and Sézary syndrome with
of hairy-cell leukemia: a multicenter study of 64 patients. J Clin alemtuzumab. Eur J Haematol 2003;71:250–6.
Oncol 1986;4:900–5. 94. Kessler JF, Jones SE, Levine N, et al. Isotretinoin and cutaneous
76. Gordon LI, Witzig T, Molina A et al. Yttrium 90-labeled ibritu- helper T-cell lymphoma (mycosis fungoides). Arch Dermatol
momab tiuxetan radioimmunotherapy produces high response 1987; 123:201–204.
rates and durable remissions in patients with previously treated 95. Khan KD, Emmanouilides C, Benson DM Jr, et al. A phase 2
B-cell lymphoma. Clin Lymphoma 2004;5:98–101. study of rituximab in combination with recombinant interleukin-2
77. Grever M, Kopecky K, Foucar MK, et al. Randomized comparison for rituximab-refractory indolent non-Hodgkin’s lymphoma. Clin
of pentostatin versus interferon alfa-2a in previously untreated Cancer Res 2006;12:7046–53.
patients with hairy cell leukemia: an intergroup study. J Clin Oncol 96. Kim JG, Sohn SK, Chae YS, et al. Alemtuzumab plus CHOP as front-
1995;13:974–82. line chemotherapy for patients with peripheral T-cell lymphomas: a
78. Hainsworth JD, Litchy S, Burris HA 3rd, et al. Rituximab as first- phase II study. Cancer Chemother Pharmacol 2007;60:129–34.
line and maintenance therapy for patients with indolent non-Hodg- 97. Knox S, Hoppe RT, Maloney D, et al. Treatment of cutaneous
kin’s lymphoma. J Clin Oncol 2002;20:4261–7. T-cell lymphoma with chimeric anti-CD4 monoclonal antibody.
79. Hainsworth JD, Litchy S, Barton JH, et al. Single-agent rituximab Blood 1996; 87:893–899.
as first-line and maintenance treatment for patients with chronic 98. Kuzel TM, Roenig HH Jr, Samuelson E, et al. Effectiveness of
lymphocytic leukemia or small lymphocytic lymphoma: a phase II interferon alfa-2a combined with phototherapy for mycosis fun-
trial of the Minnie Pearl Cancer Research Network. J Clin Oncol goides and the Sézary syndrome. J Clin Oncol 1995;13:257–63.
2003;21:1746–51. 99. Kuzel TM, Li S, Eklund J, et al. Phase II study of denileukin
80. Hillmen P, Skotnicki AB, Robak T et al. Alemtuzumab compared diftitox for previously treated indolent non-Hodgkin lymphoma:
with chlorambucil as first-line therapy for chronic lymphocytic final results of E1497. Leuk Lymphoma 2007;48:2397–402.
leukemia. J Clin Oncol 2007;25:5616–23. 100. Kwak LW, Campbell MJ, Czerwinski DK, et al. Induction of
81. Ho AD, Thaler J, Mandelli F, et al. Response to pentostatin in immune responses in patients with B-cell lymphoma against the
hairy-cell leukemia refractory to interferon-alpha. The European surface-immunoglobulin idiotype expressed by their tumors. N
Organization for Research and Treatment of Cancer Leukemia Engl J Med 1992; 327:1209–15.
Cooperative Group. J Clin Oncol 1989; 7:1533–38. 101. Lamanna N, Kalaycio M, Maslak P, et al. Pentostatin, cyclophos-
82. Horning SJ, Younes A, Jain V, et al. Efficacy and safety of tositu- phamide, and rituximab is an active, well-tolerated regimen for
momab and iodine-131 tositumomab (Bexxar) in B-cell lym- patients with previously treated chronic lymphocytic leukemia.
phoma, progressive after rituximab. J Clin Oncol 2005;23: J Clin Oncol 2006;24:1575.
712–9. 102. Langenmayer I, Nerl C, Knauf W, et al. Interferon-alpha 2b (IFN
83. Hsu FJ, Caspar CB, Czerwinski D, et al. Tumor-specific idiotype alpha) for early-phase chronic lymphocytic leukaemia with high
vaccines in the treatment of patients with B-cell lymphoma – long- risk for disease progression: results of a randomized multicentre
term results of a clinical trial. Blood 1997;89:3129–35. study. Br J Haematol 1996;94:362–9.
Robert O. Dillman 707

103. Lauria F, Lenoci M, Annino L, et al. Efficacy of anti-CD20 interferon maintenance. Proc Am Soc Clin Oncol 2003;22:564
monoclonal antibodies (Mabthera) in patients with progressed [abstract 2269]
hairy cell leukemia. Haematologica 2001;86:1046–50. 118. McSweeney EN, Giles FJ, Worman CP, et al. Recombinant inter-
104. LeMaistre CF, Saleh MN, Kuzel TM, et al. Phase I trial of a feron alfa 2a in the treatment of patients with early stage B
ligand fusion-protein (DAB389IL-2) in lymphomas expressing chronic lymphocytic leukaemia. Br J Haematol 1993;85:77–83.
the receptor for interleukin-2. Blood 1998; 91:399–405. 119. Miller VA, Benedetti FM, Rigas JR, et al. Initial clinical trial of a
105. Lenz G, Dreyling M, Schiegnitz E et al. Myeloablative radiochemo- selective retinoid X receptor ligand, LGD1069. J Clin Oncol
therapy followed by autologous stem cell transplantation in first 1997;15:790–5.
remission prolongs progression-free survival in follicular lym- 120. Molin L, Thomsen K, Volden G, et al. Oral retinoids in mycosis
phoma: results of a prospective, randomized trial of the German fungoides and Sezary syndrome: a comparison of isotretinoin
Low-Grade Lymphoma Study Group. Blood 2004;104:2667–74. and etretinate. A study from the Scandinavian Mycosis Fungoides
106. Lenz G, Dreyling M, Hoster E, et al. Immunochemotherapy with Group. Acta Derm Venereol 1987; 67:232–6.
rituximab and cyclophosphamide, doxorubicin, vincristine, and 121. Mones JV, Coleman M, Kostakoglu L, et al. Dose-attenuated
prednisone significantly improves response and time to treatment radioimmunotherapy with tositumomab and iodine 131 tositu-
failure, but not long-term outcome in patients with previously momab in patients with recurrent non-Hodgkin’s lymphoma
untreated mantle cell lymphoma: results of a prospective ran- (NHL) and extensive bone marrow involvement. Leuk Lymphoma
domized trial of the German Low Grade Lymphoma Study Group 2007;48:342–8.
(GLSG). J Clin Oncol 2005;23:1984–92. 122. Montillo M, Tedeschi A, Miqueleiz S, et al. Alemtuzumab as
107. Leonard JP, Coleman M, Kostakoglu L, et al. Abbreviated consolidation after a response to fludarabine is effective in purging
chemotherapy with fludarabine followed by tositumomab and residual disease in patients with chronic lymphocytic leukemia.
iodine I 131 tositumomab for untreated follicular lymphoma. J Clin Oncol 2006;24:2337–42.
J Clin Oncol 2005;23:5696–704. 123. Moreton P, Kennedy B, Lucas G, et al. Eradication of minimal
108. Lundin J, Osterborg A, Brittinger G, et al. CAMPATH-1H mono- residual disease in B-cell chronic lymphocytic leukemia after
clonal antibody in therapy for previously treated low-grade non- alemtuzumab therapy is associated with prolonged survival.
Hodgkin’s lymphomas: a phase II multicenter study. European J Clin Oncol 2005;23:2971–9.
Study Group of CAMPATH-1H Treatment in Low-Grade Non- 124. Nademanee A, Forman S, Molina A, et al. A phase 1/2 trial of
Hodgkin’s Lymphoma. J Clin Oncol 1998; 16:3257–3263. high-dose yttrium-90-ibritumomab tiuxetan in combination with
109. Lundin J, Kimby E, Bjorkholm M, et al. Phase II trial of subcuta- high-dose etoposide and cyclophosphamide followed by autolo-
neous anti-CD52 monoclonal antibody alemtuzumab (Campath-1H) gous stem cell transplantation in patients with poor-risk or
as first-line treatment for patients with B-cell chronic lymphocytic relapsed non-Hodgkin lymphoma. Blood 2005;106:2896–902.
leukemia (B-CLL). Blood 2002;100:768–73. 125. Neri N, Aviles A, Cleto S, et al. Chemotherapy plus interferon-
110. Lundin J, Hagberg H, Repp R, et al. Phase 2 study of alemtu- alpha2b versus chemotherapy in the treatment of follicular lym-
zumab (anti-CD52 monoclonal antibody) in patients with phoma. J Hematother Stem Cell Res 2001; 10:669–674.
advanced mycosis fungoides/Sezary syndrome. Blood 126. Nieva J, Bethel K, Saven A. Phase 2 study of rituximab in the
2003;101:4267–72. treatment of cladribine-failed patients with hairy cell leukemia.
111. Maier T, Tun-Kyi A, Tassis A, et al. Vaccination of patients with Blood 2003;102:810–3.
cutaneous T-cell lymphoma using intranodal injection of autolo- 127. O’Brien S, Kantarjian H, Beran M, et al. Interferon maintenance
gous tumor-lysate-pulsed dendritic cells. Blood 2003;102: therapy for patients with chronic lymphocytic leukemia in remis-
2338–44. sion after fludarabine therapy. Blood 1995;86:1298–300.
112. Marcus R, Imrie K, Belch A, et al. CVP chemotherapy plus ritux- 128. O’Brien SM, Kantarjian H, Thomas DA, et al. Rituximab dose-
imab compared with CVP as first-line treatment for advanced escalation trial in chronic lymphocytic leukemia. J Clin Oncol
follicular lymphoma. Blood 2005;105:1417–23. 2001;19:2165–70.
113. Margolin KA, Aronson FR, Sznol M, et al. Phase II trial of high- 129. O’Brien SM, Kantarjian HM, Thomas DA, et al. Alemtuzumab
dose interleukin-2 and lymphokine-activated killer cells in as treatment for residual disease after chemotherapy in patients
Hodgkin’s disease and non-Hodgkin’s lymphoma. J Immunother with chronic lymphocytic leukemia. Cancer 2003;98:2657–63.
1991;10:214–220. 130. Olsen E, Duvic M, Frankel A, et al. Pivotal phase III trial of two
114. Martinelli G, Laszlo D, Ferreri AJ, et al. Clinical activity of ritux- dose levels of denileukin diftitox for the treatment of cutaneous
imab in gastric marginal zone non-Hodgkin’s lymphoma resistant T-cell lymphoma. J Clin Oncol 2001; 19:376–88.
to or not eligible for anti-Helicobacter pylori therapy. J Clin Oncol 131. Olsen EA, Rosen ST, Vollmer RT, et al. Interferon alfa-2a in the
2005;23:1979–83. treatment of cutaneous T cell lymphoma. J Am Acad Dermatol
115. Mauro FR, Zinzani P, Zaja F et al. Fludarabine + prednisone +/- 1989;20:395–407.
alpha-interferon followed or not by alpha-interferon maintenance 132. Osterborg A, Dyer MJS, Bunjes D, et al. Phase II multicenter
therapy for previously untreated patients with chronic lympho- study of human CD52 antibody in previously untreated chronic
cytic leukemia: long term results of a randomized study. lymphocytic leukemia. J Clin Oncol 1997;15:1567–74.
Haematologica 2003;88:1348–57. 133. Peterson B, Petroni G, Oken M. Cyclophosphamide + interferon
116. McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chi- alpha-2b in follicular low-grade lymphomas: a preliminary report
meric anti-CD20 monoclonal antibody therapy for relapsed indo- on an intergroup trial (CALGB 8691 and EST 7486). Proc Am
lent lymphoma: half of patients respond to a four-dose treatment Soc Clin Oncol 1993; 12:1240 (abstract).
program. J Clin Oncol 1998;16:2825–33. 134. Pfreundschuh H, Schubert J, Ziepert M, et al. Six versus eight
117. McLaughlin P, Hagemeister FB, Romaguera J, et al. Stage IV cycles of bi-weekly CHOP-14 with or without rituximab in
indolent lymphoma: a randomized study of concurrent vs. sequen- elderly patients with aggressive CD20 + B-cell lymphomas: a
tial use of FND chemtotherapy (fludarabine, mitoxantrone, dex- randomised controlled trial (RICOVER-60). Lancet Oncol
amethasone) and rituximab monoclonal antibody therapy, with 2008;9:105–16.
708 Biological therapy of B and T cell lymphoproliferative disorders

135. Pfreundschuh M, Trumper L, Osterborg A, et al. CHOP-like che- 152. Rupoli S, Barulli S, Guiducci B, et al. Low dose interferon-alpha2b
motherapy plus rituximab versus CHOP-like chemotherapy alone combined with PUVA is an effective treatment of early stage
in young patients with good-prognosis diffuse large-B-cell lym- mycosis fungoides: results of a multicenter study. Cutaneous-T Cell
phoma: a randomised controlled trial by the MabThera International Lymphoma Multicenter Study Group. Haematologica 1999;84:
Trial (MinT) Group. Lancet Oncol 2006;7:379–91. 809–13.
136. Piro LD, Ellison DJ, Saven A. The Scripps Clinic experience with 153. Saleh MN, LeMaistre CF, Kuzel TM, et al. Antitumor activity of
2-chlorodeoxyadenosine in the treatment of hairy cell leukemia. DAB389IL-2 fusion toxin in mycosis fungoides. J Am Acad
Leuk Lymphoma 1994;14 Suppl 1:121–5. Dermatol 1998;39:63–73.
137. Piro LD, White CA, Grillo-Lopez AJ, et al. Extended rituximab 154. Shulz H, Klein SK, Rehwald U, et al. Phase 2 study of a combined
(anti-CD20 monoclonal antibody therapy in patients with immunochemotherapy using rituximab and fludarabine in patients
relapsed low-grade or follicular non-Hodgkins lymphoma. Ann with chronic lymphocytic leukemia. Blood 2002;100:3115–20.
Oncol 1999;10:655–61. 155. Smalley RV, Connors J, Tuttle RL, et al. Splenectomy vs. alpha
138. Press OW, Unger JM, Braziel RM, et al. Phase II trial of CHOP interferon: a randomized study in patients with previously
chemotherapy followed by tositumomab/iodine I-131 tositu- untreated hairy cell leukemia. Am J Hematol 1992a;41:13–8.
momab for previously untreated follicular non-Hodgkin’s lym- 156. Smalley RV, Andersen JW, Hawkins MJ, et al. Interferon alfa
phoma: five-year follow-up of Southwest Oncology Group combined with cytotoxic chemotherapy for patients with non-
Protocol S9911. J Clin Oncol 2006;24:4143–9. Hodgkin’s lymphoma. N Engl J Med 1992b; 327:1336–1341.
139. Prince HM, McCormack C, Ryan G, et al. Bexarotene capsules 157. Smalley RV, Weller E, Hawkins MJ, et al. Final analysis of the
and gel for previously treated patients with cutaneous T-cell lym- ECOG I-COPA trial (E6484) in patients with non-Hodgkin’s lym-
phoma: results of the Australian patients treated on phase II trials. phoma treated with interferon alfa (IFN-alpha2a) plus an anthracy-
Australas J Dermatol 2001;42:91–7. cline-based induction regimen. Leukemia 2001; 15:1118–1122.
140. Querfeld C, Rosen ST, Guitart J, et al. Comparison of selective 158. Solal-Celigny P, Lepage E, Brousse N, et al. Recombinant inter-
retinoic acid receptor- and retinoic X receptor-mediated efficacy, feron alfa-2b combined with a regimen containing doxorubicin in
tolerance, and survival in cutaneous t-cell lymphoma. J Am Acad patients with advanced follicular lymphoma. Groupe d’Etude des
Dermatol 2004;51:25–32. Lymphomes de l’Adulte. N Engl J Med 1993; 329:1608–1614.
141. Rai K, Davey F, Peterson B, et al. Recombinant alpha-2b-inter- 159. Stadler R, Otte HG, Luger T, et al. Prospective randomized mul-
feron in therapy of previously untreated hairy cell leukemia: ticenter clinical trial on the use of interferon-2a plus acitretin
long-term follow-up results of study by Cancer and Leukemia versus interferon-2a plus PUVA in patients with cutaneous T-cell
Group B. Leukemia 1995;9:1116–20. lymphoma stages I and II. Blood 1998;92:3578–81.
142. Rai K, Mercier RJ, Cooper MR, et al. Campath-1H is an effective 160. Straus DJ, Duvic M, Kuzel T, et al. Results of a phase II trial of
salvage therapy for fludarabine failing CLL patients: results of a oral bexarotene (Targretin) combined with interferon alfa-2b
phase II trial. Blood 2001;96:163a [abstract 703]. (Intron-A) for patients with cutaneous T-cell lymphoma. Cancer
143. Rai KR, Freter CE, Mercier RJ, et al. Alemtuzumab in previously 2007;109:1799–803.
treated chronic lymphocytic leukemia patients who also had 161. Thestrup-Pedersen K, Hammer R, Kaltoft K, et al. Treatment of
received fludarabine. J Clin Oncol 2002;20:3891–7. mycosis fungoides with recombinant interferon-alpha 2a alone and
144. Redfern CH, Guthrie TH, Bessudo A, et al. Phase II trial of idio- in combination with etretinate. Br J Dermatol 1988;118:811–8.
type vaccination in previously treated patients with indolent 162. Thomas DA, O’Brien S, Giles FJ, et al. Single agent Rituxan in
non-Hodgkin’s lymphoma resulting in durable clinical responses. early stage chronic lymphocytoic leukemia. Blood 2001;98:364a
J Clin Oncol 2006;24:3107–12. [abstract 1533].
145. Reinis M. Drug evaluation: FavId, a patient-specific idiotypic 163. Thomas DA, O’Brien S, Bueso-Ramos C, et al. Rituximab in relapsed
vaccine for non-Hodgkin’s lymphoma. Curr Opin Mol Ther or refractory hairy cell leukemia. Blood 2003;102:3906–11.
2007;9:291–8. 164. Thompson JA, Fefer A. Interferon in the treatment of hairy cell
146. Richardson SK, Lin JH, Vittorio CC, et al. High clinical response leukemia. Cancer 1987;59(3 Suppl):605–9.
rate with multimodality immunomodulatory therapy for Sézary 165. Tobinai K, Igarashi T, Itoh K, et al. Japanese multicenter phase II and
syndrome. Clin Lymphoma Myeloma 2006;7:226–32. pharmacokinetic study of rituximab in relapsed or refractory patients
147. Ritchie D, Hermans I, Yang J, et al. Autologous dendritic cells with aggressive B-cell lymphoma. Ann Oncol 2004;15:821–30.
pulsed with eluted peptide as immunotherapy for advanced B-cell 166. Treon SP, Agus DB, Link B, et al. CD-20 directed antibody-
malignancies. Leuk Lymphoma 2006;47:675–82. mediated immunotherapy induces responses and facilitates
148. Robak T, Smolewski P, Cebula B, et al. Rituximab combined hematologic recovery in patients with Waldenstrom’s macroglob-
with cladribine or with cladribine and cyclophosphamide in ulinemia. J Immunother 2001; 24:272–279.
heavily pretreated patients with indolent lymphoproliferative dis- 167. Treon SP, Emmanouilides C, Kimby E, et al. Extended rituximab
orders and mantle cell lymphoma. Cancer 2006;107:1542–50. therapy in Waldenstrom’s macroglobulinemia. Ann Oncol 2005;16:
149. Rodgriquez-Calvillo M, Inoges S, Lopez-Diaz de Cerio A, et al. 132–8.
Variations in “rescuability” of immunoglobulin molecules from 168. Unterhalt M, Herrmann R, Tiemann M, et al. Prednimustine,
different forms of human lymphoma: implications for anti-idiotype mitoxantrone (PmM) vs cyclophosphamide, vincristine, predni-
vaccine development. Crit Rev Oncol Hematol 2004;52:1–7. sone (COP) for the treatment of advanced low-grade non-Hodg-
150. Rohatiner A, Radford J, Deakin D, et al. A randomized controlled kin’s lymphoma. German Low-Grade Lymphoma Study Group.
trial to evaluate the role of interferon as initial and maintenance Leukemia 1996;10:836–43.
therapy in patients with follicular lymphoma. Br J Cancer 2001; 169. Van Oers MH, Klasa R, Marcus RE, et al. Rituximab mainte-
85:29–35. nance improves clinical outcome of relapsed/resistant follicular
151. Rohatiner AZ, Gregory WM, Peterson B, et al. Meta-analysis to non-Hodgkin lymphoma in patients both with and without ritux-
evaluate the role of interferon in follicular lymphoma. J Clin imab during induction: results of a prospective randomized phase
Oncol 2005;23:2215–23. 3 intergroup trial. Blood 2006;108:3295–301.
Robert O. Dillman 709

170. Vose JM, Bierman PJ, Enke C, et al. Phase I trial of iodine-131 phoma treated with yttrium 90 ibritumomab tiuxetan. Cancer
tositumomab with high-dose chemotherapy and autologous stem- 2007;109:1804–10.
cell transplantation for relapsed non-Hodgkin’s lymphoma. J 181. Wollina U, Looks A, Meyer J, et al. Treatment of stage II cutane-
Clin Oncol 2005;23:461–7. ous T-cell lymphoma with interferon alfa-2a and extracorporeal
171. Vose JM, Bierman PJ, Loberiza FR Jr, et al. Phase I trial of (90) photochemotherapy: a prospective controlled trial. J Am Acad
Y-ibritumomab tiuxetan in patients with relapsed B-cell non- Dermatol 2001;44:253–60.
Hodgkin’s lymphoma following high-dose chemotherapy and 182. Zinzani PL, Alinari L, Tani M, et al. Preliminary observations of
autologous stem cell transplantation. Leuk Lymphoma a phase II study of reduced-dose alemtuzumab treatment in
2007;48:683–90. patients with pretreated T-cell lymphoma. Haematologica
172. Weber JS, Yang JC, Topalian SL, et al. The use of interleukin-2 2005;90:702–3.
and lymphokine-activated killer cells for the treatment of patients 183. Zinzani PL, Tani M, Fanti S, et al. A phase 2 trial of fludarabine
with non-Hodgkin’s lymphoma. J Clin Oncol 1992; 10:33–40. and mitoxantrone chemotherapy followed by yttrium-90 ibritu-
173. Wendtner CM, Ritgen M, Schweighofer CD, et al. Consolidation momab tiuxetan for patients with previously untreated, indolent,
with alemtuzumab in patients with chronic lymphocytic leuke- nonfollicular, non-Hodgkin lymphoma. Cancer 2008;112:856–62.
mia (CLL) in first remission – experience on safety and efficacy 184. Kunkel LA. Idiotype vaccines in the treatment of B-cell non-
within a randomized multicenter phase III trial of the German Hodgkin’s lymphoma. Cancer Invest 2004;22:97–105.
CLL Study Group (GCLLSG). Leukemia 2004;18:1093–101. 185. Canellos GP. Interferon in the treatment of malignant lymphoma.
174. Wierda W, O’Brien S, Wen S, et al. Chemoimmunotherapy with Semin Oncol 1985;12(4 Suppl 5):25–9.
fludarabine, cyclophosphamide, and rituximab for relapsed and 186. Gluck WL, Hurst D, Yuen A, et al. Phase I studies of interleukin
refractory chronic lymphocytic leukemia. J Clin Oncol 2005;23: (IL)-2 and rituximab in B-cell non-hodgkin’s lymphoma: IL-2
4070–8. mediated natural killer cell expansion correlations with clinical
175. Wiseman GA, Witzig TE. Yttrium-90 (90Y) ibritumomab tiux- response. Clin Cancer Res 2004;10:2253–64.
etan (Zevalin) induces long-term durable responses in patients 187. Hillmen P, Skotnicki AB, Robak T, et al. Alemtuzumab com-
with relapsed or refractory B-Cell non-Hodgkin’s lymphoma. pared with chlorambucil as first-line therapy for chronic lympho-
Cancer Biother Radiopharm 2005;20:185–8. cytic leukemia. J Clin Oncol 2007;25:5616–23.
176. Witzig TE, White CA, Wiseman GA et al. Phase I/II trial of 188. Itala M, Geisler CH, Kimby E, et al. Standard-dose anti-CD20
IDEC-Y2B8 radioimmunotherapy for treatment of relapsed or antibody rituximab has efficacy in chronic lymphocytic leukae-
refractory CD20 + B-cell non-Hodgkin’s lymphoma. J Clin Oncol mia: results from a Nordic multicentre study. Eur J Haematol
1999;17:3793–803. 2002;69:129–34.
177. Witzig TE, Flinn IW, Gordon LI et al. Treatment with ibritu- 189. Lin TS, Flinn IW, Lucas MS, et al. Filgrastim and alemtuzumab
momab tiuxetan radioimmunotherapy in patients with rituximab- (Campath-1H) for refractory chronic lymphocytic leukemia.
refractory follicular non-Hodgkin’s lymphoma. J Clin Oncol Leukemia 2005a;19:1207–10.
2002a;20:3262–9. 190. O’Connell MJ, Colgan JP, Oken MM et al. Clinical trial of
178. Witzig TE, Gordon LI, Cabanillas F, et al. Randomized con- recombinant leukocyte A interferon as initial therapy for favor-
trolled trial of yttrium- 90-labeled ibritumomab tiuxetan radio- able histology non-Hodgkin’s lymphomas and chronic lympho-
immunotherapy versus rituximab immuno- therapy for patients cytic leukemia. An Eastern Cooperative Oncology Group pilot
with relapsed or refractory low-grade, follicular, or transformed study. J Clin Oncol 1986;4:128–36.
B-cell non-Hodgkin’s lymphoma. J Clin Oncol 2002b;20: 191. Peterson BA, Petroni GR, Oken MM, et al. Proc Am Soc Clin
2453–63. Oncol 1997;16:14a (abstract.)
179. Witzig TE, Vukov AM, Habermannn TM, et al. Rituximab ther- 192. Sherwin SA, Knost JA, Fein S et al. A multiple-dose phase I trial
apy for patients with newly diagnosed, advanced-stage, follicular of recombinant leukocyte A interferon in cancer patients. JAMA
grade I non-Hodgkin’s lymphoma: a phase II trial in the North 1982;248:2461–6.
Central Cancer Treatment Group. J Clin Oncol 2005;23:1103–8. 193. Timmerman JM, Czerwinski DK, Davis TA et al. Idiotype-pulsed
180. Witzig TE, Molina A, Gordon LI, et al. Long-term responses in dendritic cell vaccination for B-cell lymphoma: clinical and
patients with recurring or refractory B-cell non-Hodgkin lym- immune responses in 35 patients. Blood 2002;99:1517–26.
21.8 Biological therapy of multiple myeloma
ROBERT K. OLDHAM

Interferon (nonsecreted) could be attempted. No major clinical


studies of antibody against other plasma cell membrane
Several studies have demonstrated response rates of targets have been reported.
about 20% in patients with refractory multiple myeloma CD20 is rarely expressed on plasma cells, but has
treated with recombinant alpha-interferon [1, 3, 7]. An been reported to be present on malignant cells from up
attempt to augment this response experience by using to 20% of patients with multiple myeloma [11].
high dose induction schedules, often with the addition of Rituximab has rarely produced objective responses in
Prednisone, did not result in enhanced response [1]. multiple myeloma even in patients whose plasma cells
Anecdotal observations have suggested possible syner- were felt to overexpress CD20. Using the standard dos-
gism between the interferons and cytotoxic drugs [2, 5]. ing of 375 mg/m2, Treon et al saw objective responses in
Because of response rates of 50% in untreated patients only 1/19 patients [11], and Hussein et al reported
[8], studies were done to evaluate the combination of responses only 2/21 patients prior to starting chemo-
interferon with standard chemotherapeutic regimens as therapy five weeks later [10]. There has been a much
initial therapy for multiple myeloma. Preliminary inter- greater unpublished experience in the community prac-
pretation suggested that the duration of initial response tice of medicine with anecdotal reports of occasional
may be extended with the addition of alpha-interferon excellent responses in refractory myeloma patients
[6]. Follow-up studies were not confirmatory. whose plasma cell overexpressed CD20. Another strat-
The availability of very active new agents such as egy is to use rituximab as an adjuvant treatment after an
Velcade has precluded further studies with Interferon com- intial response to chemotherapy, in an effort to suppress
binations. In addition, thalidomide and reilamide, with the B cell clone that leads to the malignant plasma cells.
their immunomodulating capacities, are active in multiple Based on this same rationale, rituximab may be more
myeloma [9]. Given the high degree of activity both with active against smoldering myeloma than myeloma with
chemotherapy and immunomodulators, as well as the use a high proliferative index.
of autologous bone marrow transplants, Interferon is now
little studied in multiple myeloma [4, 9].
With the advent of Velcade and revlamide as targeted
therapy for multiple myeloma [4, 9], there has been little References
subsequent activity with other forms of biotherapy in this dis- 1. Case DC, Jr., Sonneborn HL, Paul SD et al. Phase II study of rDNA
order. However, given the activity of revlamide as an immu- alpha-2 interferon (INTRON A) in patients with multiple myeloma
nomodular, we may soon see other drugs working through utilizing an escalating induction phase. Cancer Treat Rep 1986;
70:1251–1254.
the immune system to treat this disseminated disease.
2. Clark RH, Dimitrov NV, Axelson JA, and Charmella LJ. Leukocyte
interferon as a biological response modifier in lymphoproliferative
diseases resistant to standard therapy. Blood 1983; 62:188a

Antibodies (abstract).
3. Cooper MR. Interferons in the treatment of multiple myeloma.
Semin Oncol 1986; 13:13–20.
Plasma cells produce myeloma protein (antibody), giving
4. Dimopoulos M, Spencer A, Attal M, et al. Lenalidomide plus dex-
ready availability of a secreted protein target for antibody amethasone for relapsed or refractory multiple myeloma. N Engl
induction. However, this very characteristic makes it Med 2007;357:2123–2132.
unlikely that antibody therapy will be useful in myeloma, 5. Ferraresi R. Enhanced response to chemotherapy after treatment
at least not with the myeloma protein as target, since with DNA alpha-2 interferon. Am Soc Hematol 1983; 62:212a.
6. Mandell F, Tribalto M. Recombinant alpha-2b interferon as main-
the therapeutic antibody would be totally absorbed in tenance therapy in responding multiple myeloma patients. Blood
the vascular and extracellular pool. Anti-CEA studies 1987; 70:247a, 318.
in colon cancer suggest this is not a rational approach to 7. Ohno R, Kimura K. Treatment of multiple myeloma with recombi-
follow, but targeting myeloma cell membrane proteins nant interferon alfa-2a. Cancer 1986; 57:1685–1688.

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 711
© Springer Science + Business Media B.V. 2009
712 Biological therapy of multiple myeloma

8. Quesada JR, Alexanian R, Hawkins M et al. Treatment of multiple 10. Hussein MA, Karam MA, McLain DA, et al. Biologic and clinical
myeloma with recombinant alpha-interferon. Blood 1986; evaluation of rituxan in the management of newly diagnosed mul-
67:275–278. tiple myeloma patients. Blood 1999;94:313a [abstract 1400].
9. Weber DM, Chen C, Niesbizky R, et al. Lenalidomide plus dexam- 11. Treon SP, Pilarski LM, Belch AR, et al. CD20-directed serotherapy
ethasone for relapsed multiple myeloma in North America. N Engl in patients with multiple myeloma: biologic considerations and
J Med 2007;357:2133–2142. therapeutic applications. J Immunother. 2002;25:72-81.
21.9 Biological therapy of squamous cell cancers
of the head and neck
ROBERT O. DILLMAN

Introduction patients. In another randomized prospective study of


patients with advanced squamous cell carcinoma of the
This category includes squamous cell cancers that originate in head and neck, treatment with methotrexate plus BCG
the mouth, oropharynx, nasopharynx, and larynx. Many was no better than methotrexate alone in terms of response
of the cancers are associated with smoking and other rate or survival [62].
forms of tobacco abuse that are local irritants and mutants, There were two small trials with relatively positive
and some are caused by human papilloma virus, and other results. One was another small randomized phase II trial,
viruses. Primary therapy typically includes surgery and/or in which 34 patients with recurrent squamous cell cancer
radiation therapy, and increasingly chemotherapy as part of the head and neck were randomized to receive the five-
of organ sparing approaches that minimize the morbidity drug chemotherapy regimen BACON (bleomycin, dox-
associated with extensive surgery in this area. orbucin, CCNU, vincristine and nitrogen mustard) alone
(n = 14) or with BCG by scarification (n = 20) [41]. The
patients treated with BACON plus BCG experienced a
Non-specific Immune Stimulants longer survival (P = 0.014) than those treated with
BACON alone. The other positive trial was in the neoad-
Bacillus Calmette Guerin (BCG) juvant setting. Following neoadjuvant methotrexate and
There were no trials that tested single-agent activity of definitive local therapy, 52 patients with locally advanced
BCG, but several trials addressed the issue of whether squamous cell cancer were randomized to adjuvant meth-
BCG augmented methotrexate or other chemotherapy otrexate alone (n = 27) or with an adjuvant treatment with
regimens in the treatment of advanced squamous cell a BCG vaccine (n = 25) consisting of 2 to 4 million Tice
cancers of the head and neck. In a large, non-randomized strain BCG organisms given i.d. in alternating sides of the
phase II trial, 100 patients with advanced, recurrent, or neck every 2 weeks for six doses, then every 4 weeks for
metastatic squamous cell carcinoma of the head and neck nine doses [50]. At a median follow-up of about 3.5 years,
were treated with the combination of chemotherapy and 52% of the BCG vaccine-treated group remained disease
BCG [49]. The objective response rate was 35%, and the free compared to only 26% of the controls. Similarly,
median duration of response was 17 weeks, both of which 68% of the BCG vaccine-treated group were alive at 3.5
were considered similar to historical results obtained with years compared to only 41% of the controls. Based on
chemotherapy alone. In a small randomized phase II trial, this trial BCG showed promise as adjuvant immunother-
38 patients with advanced, inoperable squamous cell car- apy in ear, nose, and throat cancers.
cinoma of the head and neck were randomized to receive
methotrexate alone or with BCG [35]. The response rates, Levamisole
3/19 in the methotrexate alone arm and 4/19 for metho-
trexate plus BCG, were similar as was the duration of Several small randomized trials were conducted in head
response and survival. In another small randomized phase and neck cancer with the antihelminth levamisole.
II trial, 23 patients with advanced recurrent head and neck A randomized double-blind study compared levamisole
carcinoma were randomized to receive either high dose (n = 31) with placebo (n = 34) as adjuvant treatment
methotrexate with calcium leucovorin rescue (HDMTX) following surgery of patients with squamous cancer
(n = 12) or HDMTX in combination with BCG (n = 11) of the head and neck [61]. There was no difference in
[6]. There were three objective tumor responses of simi- progression free survival (PFS) or overall survival (OS).
lar brief duration in both groups. Although this trial was The largest randomized trial ever conducted with levam-
too small to be conclusive, it cast doubt on whether BCG isole in head and neck cancer patients, actually sug-
added anything to HDMTX chemotherapy for such gested a detrimental effect. After treatment of a primary

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 713
© Springer Science + Business Media B.V. 2009
714 Biological therapy of squamous cell cancers of the head and neck

squamous cell carcinoma of the head and neck, 134 the tumor-bearing lymph nodes of the neck, or into the
patients were randomized to receive placebo (n = 65) or cervical node region in patients who lacked palpable
levamisole, 150 mg/day orally for 3 consecutive days lymph nodes, before and after radiation therapy [10].
every other week [33]. Patients with stage I and II disease After 2.5 years of follow up, the study was terminated
who were treated with levamisole had a significantly when it became evident that there was no difference dis-
higher incidence of recurrence than the placebo-treated ease free survival. In a large trial 209 operable patients
patients (p = 0.02). with squamous cell head and neck cancer were random-
Some other small studies were more encouraging. In ized to surgery alone or pre-operative intratumoral
a small randomized study of 24 patients with squamous immunotherapy with C parvum followed by post-operative
cell cancer of the larynx or hypopharynx, 12 patients s.c. C parvum for 2 years [31]. Only 176 were considered
received placebo and 12 received levamisole 50 mg t.i.d. fully evaluable, but there was no difference disease free
for 3 consecutive days every 2 weeks following surgery or overall survival.
and/or radiation [30]. After 20 months of follow up, the
recurrence rate was only 20% in the levamisole group
compared to 50% in the control group. In another trial,
OK-432
following conventional radiotherapy, 82 patients with The streptococcal preparation OK-432 has been exten-
T1 or T2 N0 M0 squamous cell carcinoma of the oral sively studied in Japan in various malignancies including
cavity were randomized to receive either placebo or epidermoid cancers of the head and neck. During 1984
levamisole at a dose of 150 mg daily for 3 consecutive to 1989 120 newly diagnosed patients of laryngeal
days every 2 weeks [34]. After a median of 3 years of squamous cell carcinoma were registered at ten differ-
follow up, 44% of patients who received levamisole ent institutions, stratified by stages of disease, treated
were still disease-free compared to only 32% in the pla- with radiation therapy and 5FU chemotherapy with or
cebo group. There also was a faster recovery from radi- without surgery, with or without chemotherapy, and
ation-induced leukopenia and lymphopenia in the randomized to no further therapy or immunotherapy
levamisole group. In another small randomized study, with OK-432 [23]. There was no apparent advantage for
34 patients without distant metastases were randomized those patients who received OK-432. The 5-year dis-
to observation and 31 to receive futraful and uracil ease free survival rate for 109 evaluable patients was
(UFT) plus levamisole as adjuvant treatment for stage 76% in the OK-432 arm and 75% in the control arm.
III and IV squamous cell carcinomas of oral cavity, The 5-year overall survival rate was 84% in the OK-432
oropharynx, hypopharynx and larynx following primary arm and 78% in the control arm.
therapy [24]. The 5-year disease free survival rate
favored the UFT-levamisole arm (57% versus 39%), but
was not statistically significant (p = 0.21). There was
Thymic Hormones
also a trend for a lower rate of distant metastasis (10% It has long been recognized that the thymus gland is
versus 32%, p = 0.06). The contribution of levamisole important in the development of cell-mediated immu-
beyond UFT is unclear. nity and that many thymic hormones have stimulating
affects on T lymphocytes. Clinical experiments have
been performed with crude thymus extracts, isolated
Cornybacterium Parvum (C parvum) purified products, and synthesized peptides. Several
Three randomized trials failed to demonstrate any ben- thymic preparations have been of clinical interest,
efit for C parvum in the treatment of epidermoid cancers including thymosin fractions 5, thymosin-α1, prothy-
of the head and neck. In a small randomized trial, mosin, thymulin, thymopoietin, thymostimulin (TP-1),
patients with advanced squamous cancer of the head and thymic humoral factor [46]. In vitro thymostimulin
and neck were randomized to treatment with weekly restores a number of immunologic defects noted in the
methotrexate alone or methotrexate with C parvum monocytes and dendritic cells of patients with head and
given s.c. weekly as bilateral doses in sites around the neck cancers. In a small study, 18 patients with squamous
neck [63]. The addition of C. parvum did not improve cell cancer of the head and neck were treated with thy-
the response rate, response duration or survival. In a mostimulin at one of three dosages (0.5, 1.0, or 2.0 mg/
randomized trial involving 57 patients with previously kg) prior to surgery, and then their exicised tumors were
untreated squamous cell carcinoma of the head and examined and results compared to those from 16 con-
neck, 29 received radiation alone and 28 patients temporary controls who had not received treatment with
received C. parvum administered i.v. and locally into the thymic hormone [21]. Preoperative treatment with
Robert O. Dillman 715

thymostimulin enhanced T-cell infiltration. Anti-tumor a 30% response rate in 18 evaluable patients who had an
effects were not determined. There have been no publi- 8.5 month median survival [3]. IFN-α2b, cisplatin and
cations of follow up studies. 5FU were combined to treat 14 patients with previously
treated squamous cell head and neck cancers [2].
Hematologic toxicity caused treatment delays in 9/14
patients, which led to early closure of the trial, but 8/14
Interferons (54%) of the patients had an objective response rate.
Interferon-α
Interferon-alpha (IFN-α) was studied in several trials in Interferon-gamma
squamous cell cancer of the head and neck. In a phase II Interferon-gamma (IFN-γ) was given at a dose of
trial, 71 patients with recurrent or metastatic squamous 0.25 mg/m2 as a 24-h infusion i.v. weekly for 4 weeks to
cell carcinoma of the head and neck were randomized to eight patients with locally advanced, but resectable head
receive recombinant IFN-α2b at a relatively low dose (6 and neck cancer [42]. There were minimal side effects,
MU/m2 daily × three every 4 weeks) or a relatively high and three patients had clinically measurable responses
dose (12 MU thrice weekly) [59]. As shown in Table 1, prior to surgery.
the response rate was 5%, with a response rate of 1/32
for evaluable patients who received the lower dose, and
2/29 for those who received the higher dose. Median
survival was about 6 months in both arms. Interleukins
Other trials combined IFN-α with radiation therapy
and/or chemotherapy. In a small study, 22 patients with
Interleukin-2
operable head and neck cancer were randomized to Local injections of IL-2 might enhance an immune
receive radiotherapy alone prior to surgery, or radiother- response in lymph nodes that drain head and neck can-
apy with natural leukocyte IFN-α at a dose of 6 MU i.m. cers. In one study, 20 patients with recurrent, inoperable
daily for 4 weeks prior to surgery, and then thrice weekly head and neck squamous cell carcinoma received peri-
for 2 months [54]. Objective responses were observed in lymphatic injections of natural IL-2 for 10 days [11].
4/10 patients who received IFN-α and radiotherapy, Irrespective of the site of the recurrence, injections were
compared to 2/12 among those who only underwent always given 1.5 cm below the insertion of the sterno-
radiation therapy, but there was no difference in survival. cleidomastoid muscle on the mastoid. Injections were
The study was discontinued because of worse toxicity in performed on the ipsilateral side if the lymphatic chain
the IFN-α cohort. IFN-α was combined with 5-FU and was still present, or on the contralateral side if the patient
cisplatin to treat patients with recurrent or metastatic had previously undergone an extensive neck dissection.
squamous cell carcinoma of the head and neck who had Thirteen patients had brief response to such treatment,
not received prior chemotherapy [20]. The response rate and there was no benefit from repeated treatment. The
was 25% among 50 eligible patients and the median sur- same study investigators treated 31 patients with recur-
vival was 5 months. The authors concluded that IFN-α rent head and neck squamous cell carcinoma with ten
added to toxicity without conveying a benefit to the che- daily doses of 500 or 500,000 U of IL-2 as injections
motherapy. In 20 patients with recurrent and/or meta- 1.5 cm from the insertion of the sternocleidomastoid
static squamous cell carcinoma of the head and neck, the muscle on the mastoid [12]. There were no toxic effects.
same combination of chemotherapy and IFN-α produced Objective, but non-durable tumor responses were noted
in 4/16 patients who received the 500 U dose, but the
response rate was 0/15 at the higher dose. In a U.S. dose
Table 1. Single-agent activity of various biologicals in escalation trial of local therapy, IL-2 was injected per-
patients with head and neck cancer ilesionally in divided doses in each of four quadrants
Modality Biological Lead Response and bilaterally into the superior jugular lymph nodes in
class agent author Patients rate (%) 36 patients with unresectable squamous cell carcinoma
Non-specific Isotretinoin [25] 19 16 of the head and neck [58]. The maximum tolerated dose
Cytokine Interferon-α [59] 61 5 was determined to be 2 MU/day. There were two objective
Cytokine Interleukin-2 [12] 31 13 responses for a response rate of 6%.
(IL-2) In a European multicenter trial, 202 patients with
Mab Cetuximab [57] 109 13 advanced, but potentially resectable cancers of the oral
716 Biological therapy of squamous cell cancers of the head and neck

cavity or oropharynx, were randomized to undergo sur- were treated by peritumoral injection of this cytokine
gery and radiation therapy alone or in combination with combination, in addition to oral zinc sulfate, oral indo-
perilymphatic recombinant IL-2 [14]. A dose of 5,000 U methacin, i.v. cyclophosphamide. [15]. Four patients
IL-2 was injected around the ipsilateral cervical lymph reportedly had an objective response. In a four-center
node chain daily for 10 days before surgery, and after phase I/II dose-escalation clinical trial, 54 patients with
surgery the same IL-2 dose was injected into the con- locally advanced, node-negative primary oral squamous
tralateral cervical lymph node chain 5 consecutive days cell carcinoma were treated with a leukocyte multikine
monthly for 1 year. Treatment was well-tolerated and interleukin product prior to surgical resection [51].
did not interfere with the treatment plan. Those patients Paraffin-embedded tumor samples obtained at surgical
who received IL-2 had a longer disease free survival resection of the residual tumor showed what the authors
(p < 0.01) and overall survival (p < 0.03). interpreted as increased intraepithelial T cell infiltration.
A preparation of polyethylene glycol-modified IL-2 Because of the trial design, it was not possible to tell
(PEG-IL-2) provides a slower release of IL-2 from sites whether significant anti-tumor effects occurred.
of injection, and therefore a more continuous exposure
to the IL-2. PEG-IL-2 was given to 19 patients with
recurrent head and neck squamous cell carcinoma as
Interleukin-12
intratumoral injections of 200,000 U of (PEG-IL-2) two Interleukin-12 (IL-12) was injected intratumorally prior
or three times a week for 4 weeks [29]. Temporary to surgery once weekly, two or three times, at either 100
regional swelling and redness developed in ten patients, or 300 ng/kg, in ten previously untreated patients with
and nine patients had systemic eosinophilia. One patient head and neck squamous cell carcinoma [55]. The injec-
had an objective response. tions resulted in a greater infiltration of natural killer
Several trials combined IL-2 and IFN-α. In a phase II cells and CD20+ B cells than were seen in samples from
study 11 patients with recurrent head and neck cancer 20 patients who had not received IL-12. Because of the
were treated with continuous i.v. recombinant human trial design, it was not possible to tell whether signifi-
IL-2 and i.m. or s.c. IFN-α2a [53]. Two patients (18%) cant anti-tumor effects occurred.
achieved a partial response, but toxicity was substantial.
In another phase II trial, a response rate of 36% was
reported for 14 patients with advanced head and neck
cancer, even though toxicity was such that only three
Retinoids and Vitamins
patients were able to complete three cycles of therapy There is a long history of interest in the effects of vita-
[45]. The degree of natural killer cell activation corre- mins and especially vitamin A analogs, for the treatment
lated with response. and prevention of head and neck cancers. Much of the
Other trials combined IL-2 with chemotherapy for the focus has been on 13-cis-retinoic acid (isotretinoin,
treatment of squamous cell head and neck cancer. In a CRA). As summarized in Table 1, an objective response
non-randomized phase II study, 23 patients with rate of 16% was observed in 19 evaluable patients who
advanced (stage III or IV) head and neck squamous cell were treated with isotretinoin as part of a multi-institu-
carcinoma received chemotherapy with cisplatin, 5FU, tional, randomized phase II trial that included 40 patients
and vinorelbine, or the same combination with IL-2 at a with advanced head and neck squamous cell carcinoma
dose of 9 MIU s.c. daily from day 9 to 13 and from day who were randomized to treatment with either metho-
16 to 20 of every cycle [27]. The response rate was 63% trexate or isotretinoin [25]. In this trial there were no
for the chemotherapy alone, and 100% with the addition objective responses in patients treated with methotrex-
of IL-2. In a randomized, phase III, multicenter clinical ate chemotherapy.
trial, only 33 patients with advanced head and neck Because of in vitro evidence of synergy, and some
squamous-cell carcinoma were randomized to 5FU and evidence of single-agent activity, several small trials
cisplatin alone, or combined with s.c. IL-2 at a dose of combined 13-cis-retinoic acid (isotretinoin, CRA) with
4.5 MIU/day on days 8–12 and 15–19 of each treatment IFN-α. A response rate of 13% was reported in a trial of
cycle [28]. Response rates were similar between the 16 patients with unresectable recurrent head and neck
arms, 12/17 in the chemotherapy alone arm, and 10/16 carcinomas who were treated with CRA 40 mg p.o. daily
in the IL-2 containing arm. plus IFN-α 3 MU s.c. every other day [32]. A response
Multikine is a combination of natural interleukins rate of only 5% was reported in another phase II trial of
derived from leukocytes. In a pilot study, 12 previously this regimen in 21 patients with recurrent squamous cell
untreated patients with various head and neck cancers carcinoma of the head and neck [60]. In another trial, no
Robert O. Dillman 717

responses were observed in ten patients with advanced toxicity, hypertriglyceridemia and hypercholester-
squamous cell carcinoma of the lung and of the head olemia. These authors concluded that CRA was ineffec-
and neck who were treated with high dose 13-cis-retinoic tive as chemoprevention in patients with radically
acid (2 mg/kg/day) and IFN-α [44]. Collectively, the treated HNSCC. In an Australian trial, 151 patients head
results of these three trials were disappointing, with a and neck squamous cell carcinoma, who had undergone
cumulative recorded objective responses of only 3/47 curative treatment, were randomized to 3 years of treat-
(6%) of patients. ment with isotretinoin at a high dose (1.0 mg/kg/day) or
The retinoids have produced impressive results in the a moderate dose (0.5 mg/kg/day) or placebo [36]. There
prevention of squamous cell cancer of the head and was no difference in the rate of second primary cancers
neck. Patients with squamous cell cancers of the head of the head and neck, lung, or bladder, or time to recur-
and neck remain at high risk for both recurrent and sec- rence of disease. These authors concluded that the use
ond primary tumors after initial therapy. After comple- of CRA as prophylaxis against a second cancer in head
tion of surgery or radiotherapy (or both), 103 patients and neck cancer patients is not indicated.
who were disease-free after primary treatment for The largest trial conducted to date was a phase III
squamous-cell cancers of the larynx, pharynx, or oral randomized trial of 3 years of low-dose isotretinoin
cavity were randomized to receive 12 months of either (30 mg/day) versus placebo in 1,190 patients with early-
isotretinoin (13-cis-retinoic acid) (50 to 100 mg/m2/day) stage (stage I or II) squamous cell cancer of the head
or placebo [19]. There was no reduction in the rate of and neck [22]. After 4 years of follow up, it was deter-
recurrence of the original cancer, but after a median mined that isotretinoin did not statistically significantly
follow-up of 32 months, only 4% of patients in the isot- reduce the rate of second primary tumors or increase
retinoin group had second primary tumors, as compared survival. This study reaffirmed that active smoking was
with 24% in the placebo group (p = 0.005). the greatest risk for subsequently being diagnosed with
Previously high-dose isotretinoin therapy had been additional smoking related cancers.
shown to be an effective treatment for leukoplakia, a Higher doses of cis-retinoic acid and interferon are
precursor of squamous cell cancer in the mouth. In a associated with some toxicity which might be mitigated
follow up study, 70 patients with leukoplakia underwent by concomitant administration of vitamin E; so, all three
induction therapy with a high dose of isotretinoin agents have combined as a chemoprevention therapy
(1.5 mg/kg/day) for 3 months, then patients with after treatment of advanced head and neck cancer. After
responses or stable lesions were randomly assigned to definitive local treatment with surgery, radiotherapy, or
maintenance therapy with either beta carotene (30 mg/ both, 45 patients with locally advanced squamous cell
day) or a low dose of isotretinoin (0.5 mg/kg/day) for 9 head and neck cancer were treated with 13-cRA (50 mg/
months [26]. Of 66 evaluable patients, 55% responded m2/day orally, IFN-α 3MIU/m2 s.c. thrice weekly, and
to the high-dose CRA, and 59 went on to randomization alpha-tocopherol (1,200 IU/day orally) for 12 months
between beta carotene and low-dose CRA. Of the 53 with 85% completing the planned therapy [48]. The
patients who could be evaluated, 92% in the low-dose 2-year disease free survival (DFS) rate was 81%, and
isotretinoin group and 45% in the beta carotene group overall survival (OS) 91%. In a similar trial CRA, IFN-
had sustained disease control (p < 0.001). α2a, and vitamin E were used to treat 45 patients who
Trials conducted more recently outside the United had locally advanced (III or IV) squamous cell carci-
States failed to confirm these impressive results. During noma of the head and neck that had been treated with
1992 to 1995, 272 Italian patients who had undergone surgical resection, radiation, or both [47]. The three-
radical treatment for advanced (stage III and IV) drug bioadjuvant chemopreventive treatment was con-
squamous cell head and neck cancer were randomized tinued for 12 months. The 5-year PFS was 80% and
to no further treatment (n = 126) or to receive 1 year of 5-year OS 81%, which the authors felt was much better
CRA at a dose of 0.5 mg/kg/day p.o. (n = 126) [52]. than the 40% 5-year survival that is considered the his-
Because of difficulties in getting IFN-α, a third arm of torical standard.
CRA plus IFN-α was discontinued after enrolling only Other combinations of biologicals have been tested
15 patients. After 3 years of follow up, the 5-year actu- as chemoprevention strategies for these patients. In a
arial OS rates were similar for both arms at 59% and large European trial, from 1988 to 1994, 2,592 patients
57%, as were DFS rates at 66% and 63%. Adverse (60% with head and neck cancer and 40% with lung
effects were mostly grade I, and occurred in 69% of cancer) were randomized to receive one of four treat-
treated patients. As in other trials, adverse events ment arms: retinyl palmitate (0.3 MIU daily for 1 year
included cytopenia, mucositis, conjunctivitis, cutaneous followed by 0.15 MIU for an additional year),
718 Biological therapy of squamous cell cancers of the head and neck

N-acetylcysteine (600 mg daily for 2 years), the combi- with virus-modified autologous tumor cells prepared from
nation of retinyl palmitate and N-acetylcysteine for 2 short-term tumor cultures [18]. Delayed type hypersen-
years, or no additional treatment [56]. In terms of risk, sitivity reaction to unmodified tumor cells was demon-
94% had smoked tobacco at sometime in their lives and strated, but anti-tumor effects could not be measured
25% continued to smoke after the cancer diagnosis. because of the trial design.
After a median follow-up of 4 years, 916 (35%) patients
had experienced a recurrence, second primary tumor, or
death. There was no statistically significant difference in
event-free or overall survival. or event-free survival for
Adoptive Cell Therapy
any of the 2 × 2 analyses for the impact of retinyl palmi- There has been only limited investigation of adoptive
tate or N-acetylcysteine. There was a lower incidence of cell therapy in patients with head and neck cancer. In
second primary tumors in the no intervention arm that one study, irradiated autologous tumor cells were
was not statistically significant. admixed with BCG and injected i.d. in six patients with
Retinoids have been combined with chemotherapy advanced head and neck cancers, then vaccine-primed
agents and biologicals other than IFN-α. Ifosfamide, lymphocytes were isolated from regional lymph nodes,
cisplatin, and CRA (0.5 mg/kg) were given in combina- expanded in vitro with IL-2 and anti-CD3 Mab, then
tion to treat 52 patients with squamous cell carcinoma infused into patients along with i.v. IL-2 [9]. Specific
of the head and neck, who had either locally advanced cytoxicity against autologous tumor was demonstrated
disease, that could not be managed by surgery or radia- in four patients. There were no significant tumor
tion therapy, locoregional recurrence or distant metasta- responses after transfer of the activated lymphocytes. In
ses [38]. Treatment was well-tolerated. The response another approach, three patients with recurrent head and
rate was 72%, with a median PFS of 10 months and OS neck cancer underwent leukapheresis to collect periph-
of 13 months. Cisplatin, IFN-α (6 MIU/day i.m.), and eral blood mononuclear cells (PBMC) which were then
CRA 1 (mg/kg/day) were given in combination for 12 incubated in vitro with the bifunctional monoclonal
weeks to 23 previously treated patients who had devel- antibody (Mab) catumaxomab, which has one arm that
oped advanced or metastatic head and neck squamous binds to the epithelial cell adhesion molecule (Epcam)
cell carcinoma [16]. There were four objective responses. and the other arm binds to the T cell antigen CD3 [40].
In a trial that enrolled 54 patients, 42 who had exhibited Intravenous administration of catumaxomab is compli-
clinical benefit (complete or partial response, disease cated by the release of cytokines when the Mab binds to
stability) from docetaxel, ifosfamide, and cisplatin che- circulating T cells; so in this trial the strategy was to
motherapy as treatment for recurrent or metastatic activate the T cells in vitro where the release of cytok-
squamous cell carcinoma of the head and neck, low- ines would take place ex vivo, then infused the cells i.v.
dose s.c. IL-2 and oral CRA were given as maintenance in hopes that the catumaxomab would target the cells to
immunotherapy [39]. Median PFS was 11 months and tumor. Assays showed that the PBMNC indeed did
median OS was 22 months. Progressive increases in the release substantial amounts of interferon gamma and
numbers of lymphocytes and natural killer cells were tumor necrosis factor alpha in vitro. Patients received
documented, as well as a decline in levels of vascular escalating doses from 10,000 to 10 million cells per
endothelial growth factor (VEGF). kilogram, with 1 million per kilogram being the maxi-
mum tolerated dose. One patient achieved a CR that
lasted more than 2 years.
Vaccines
The vaccines that were recently approved that prevent
infection with human papilloma virus (HPV) will pre-
Antibody Therapy
vent HPV- squamous cell head and neck cancers, which Cetuximab has been extensively evaluated in squamous
account for a small subset of all head and neck cancers. cell cancers of the head and neck. Vermorken et al. gave
In terms of the more traditional vaccine approach, there single-agent cetuximab at the standard dose and sched-
have been relatively few studies in patients with head ule to 109 patients who had recurrent and/or metastatic
and neck cancer. disease that progressed during platinum chemotherapy
In a pilot study, following radiation therapy 20 patients [57]. Acneiform skin rash, which occurred in 49%, was
with squamous cell cancer of the head and neck were the most common toxicity There was one death due to
preconditioned with IL-2 and subsequently vaccinated an infusion-related allergic reaction. The 13% response
Robert O. Dillman 719

rate was similar to that of single-agent cetuximab in col- The humanized anti-EGFR Mab h-R3 was given at
orectal cancer. four dose levels weekly for 6 weeks in combination
Phase I trials suggested that cetuximab could be with radiotherapy, to 24 patients with advanced head
safely given with radiation therapy and might enhance and neck cancer [13]. In this trial there were some infu-
therapeutic benefit [43]. Bonner et al. conducted a mul- sion reactions, but no dermatologic or allergic toxicities
tinational trial comparing cetuximab plus radiotherapy were noted. The overall survival of patients was consid-
to radiotherapy alone in 424 patients with locoregion- ered encouraging.
ally advanced disease, that showed a benefit for the
addition of cetuximab, including a 26% reduction in
death, with an increase in survival from 29 to 49 months
(p = 0.03) [4]. There was no increase in toxicity other
Summary
than the acneiform rash. The role of biotherapy in squamous cell cancer of the
Pfister et al. treated 22 patients with locoregionally head and neck is very limited at this time. Enthusiasm
advanced disease with cisplatin, cetuximab, and 70 Gy for retinoid therapy alone, or in combination with inter-
radiotherapy [37]. This trial was closed early because of feron has waned with the publication of recent random-
several adverse events from infection and cardiac events. ized trials. The results associated with regional and
Grade 3 or 4 cetuximab-related toxicities included acnei- intranodal injections are interesting, but have not cre-
form rash in 10% and hypersensitivity in 5%. With a ated commercial interest. In the near future, monoclonal
median follow-up of 52 months, the 3-year overall sur- antibody combined with chemotherapy may emerge as a
vival rate is 76%, the 3-year progression-free survival rate new standard of care for systemic disease.
is 56%, and the 3-year locoregional control rate is 71%.
Herbst et al. treated 130 patients who did not have an
objective response or relapsed within 90 days of com-
pleting two cycles of cisplatin/paclitaxel or cisplatin/
References
fluorouracil, with standard cetuximab and cisplatin (75 1. Baselga J, Trigo JM, Bourhis J, et al. Phase II multicenter study of
the antiepidermal growth factor receptor monoclonal antibody
or 100 mg/m2 i.v.) every 3 weeks [17]. The most com- cetuximab in combination with platinum-based chemotherapy in
mon toxicities were anemia, acneiform skin rash, leuko- patients with platinum-refractory metastatic and/or recurrent
penia, fatigue/malaise, and nausea/vomiting. Seven squamous cell carcinoma of the head and neck. J Clin Oncol
patients (5%) experienced a grade 3 or 4 hypersensitiv- 2005;23:5568–77.
ity reaction to cetuximab. Baselga et al. treated 96 2. Benasso M, Merlano M, Blengio F, et al. Concomitant alpha-interferon
and chemotherapy in advanced squamous cell carcinoma of the head
patients who were refractory to cisplatin, with standard and neck. Am J Clin Oncol 1993;16:465–8.
cetuximab and cisplatin at the same dose and schedule, 3. Bensmaine ME, Azli N, Domenge C, et al. Phase I-II trial of recom-
during which progressive disease had occurred [1]. binant interferon alpha-2b with cisplatin and 5-fluorouracil in recur-
Acneiform rash was the most common toxicity. rent and/or metastatic carcinoma of head and neck. Am J Clin Oncol
1996;19:249–54.
Burtness et al. randomized 117 patients who had
4. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab
recurrent or metastatic disease to receive cisplatin every for squamous-cell carcinoma of the head and neck. N Engl J Med
4 weeks with weekly cetuximab or placebo [7]. Response 2006;354:567–78.
rate was higher with the addition of cetuximab, but PFS 5. Bourhis J, Rivera F, Mesia R, et al. Phase I/II study of cetuximab in
and OS were not improved. Survival was better for combination with cisplatin or carboplatin and fluorouracil in
patients with recurrent or metastatic squamous cell carcinoma of
those patients who developed the skin rash.
the head and neck. J Clin Oncol 2006;24:2866–72.
Bourhis et al. treated 53 patients who had recurrent or 6. Buechler M, Mukherji B, Chasin W, Nathanson L. High dose meth-
metastatic disease with a combination of cetuximab, otrexate with and without BCG therapy in advanced head and neck
cisplatin or carboplatin, and escalating doses of 5FU as malignancy. Cancer 1979;43:1095–100.
initial therapy [5]. Dermatologic toxicity was the most 7. Burtness B, Goldwasser MA, Flood W, et al. Phase III randomized
trial of cisplatin plus placebo compared with cisplatin plus cetuximab
common adverse event, but the most common grade 3 in metastatic/recurrent head and neck cancer: an Eastern Cooperative
or 4 adverse events were leucopenia (38%), asthenia Oncology Group study. J Clin Oncol 2005;23:8646–54.
(25%), thrombocytopenia (15%), and vomiting (14%). 8. Chan AT, Hsu MM, Goh BC, et al. Multicenter, phase II study of
Chan et al. treated 59 patients who had recurrent or cetuximab in combination with carboplatin in patients with recurrent
metastatic nasopharyngeal cancer that had recurred after or metastatic nasopharyngeal carcinoma. J Clin Oncol 2005;23:
3568–76.
cisplatin, with cetuximab and carboplatin [8]. Six 9. Chang AE, Li Q, Jiang G, et al. Generation of vaccine-primed lym-
patients (10%) experienced serious treatment-related phocytes for the treatment of head and neck cancer. Head Neck
adverse events during cetuximab. 2003;25:198–209.
720 Biological therapy of squamous cell cancers of the head and neck

10. Cheng VS, Suit HD, Wang CC, et al. Clinical trial of 27. Mantovani G, Bianchi A, Curreli L, et al. Neo-adjuvant chemo-
Corynebacterium parvum (intra-lymph-node and intravenous) and therapy +/- immunotherapy with s.c. IL 2 in advanced squamous
radiation therapy in the treatment of head and neck carcinoma. cell carcinoma of the head and neck: a pilot study. Biotherapy
Cancer 1982;49:239–44. 1994;8:91–8.
11. Cortesina G, De Stefani A, Galeazzi E et al. Interleukin-2 injected 28. Mantovani G, Gebbia V, Airoldi M, et al. Neo-adjuvant chemo-
around tumor-draining lymph nodes in head and neck cancer. Head immuno therapy of advanced squamous-cell head and neck carci-
Neck 1991;13:125–31. noma: a multicenter, phase III, randomized study comparing
12. Cortesina G, De Stefani A, Galeazzi E, et al. Temporary regression cisplatin + 5-fluorouracil (5-FU) with cisplatin + 5-FU + recombi-
of recurrent squamous cell carcinoma of the head and neck is nant interleukin 2. Cancer Immunol Immunother 1998;47:149–56.
achieved with a low but not with a high dose of recombinant inter- 29. Mattijssen V, De Mulder PH, De Graeff A, et al. Intratumoral
leukin 2 injected perilymphatically. Br J Cancer 1994;69:572–6. PEG-interleukin-2 therapy in patients with locoregionally recur-
13. Crombet T, Osorio M, Cruz T, et al. Use of the humanized anti- rent head and neck squamous-cell carcinoma. Ann Oncol
epidermal growth factor receptor monoclonal antibody h-R3 in 1994;5:957–60.
combination with radiotherapy in the treatment of locally advanced 30. Mussche RA, Kluyskens P. Prognosis of primarily treated local-
head and neck cancer patients. J Clin Oncol 2004;22:1646–54. ized laryngeal carcinoma ameliorated through levamisole treat-
14. De Stefani A, Forni G, Rogona R, et al. Improved survival with ment: a randomized pilot study. Oncology 1980;37:329–35.
perilymphatic interleukin 2 in patients with resectable squamous 31. Neifeld JP, Terz JJ, Kaplan AM, Lawrence W Jr. Adjuvant
cell carcinoma of the oral cavity and oropharynx. Cancer Corynebacterium parvum immunotherapy for squamous cell epi-
2002;95:90–7. theliomas of the oral cavity, pharynx, and larynx. J Surg Oncol
15. Feinmesser R, Hardy B, Sadov R, et al. Report of a clinical trial in 1985;28:137–45.
12 patients with head and neck cancer treated intratumorally and 32. Nikolaou AC, Fountzilas G, Daniilidis I. Treatment of unresect-
peritumorally with multikine. Arch Otolaryngol Head Neck Surg able recurrent head and neck carcinoma with 13-cis-retinoic acid
2003;129:874–81. and interferon-alpha. A phase II study. J Laryngol Otol
16. Gravis G, Pech-Gourgh F, Viens P, et al. Phase II study of a com- 1996;110:857–61.
bination of low-dose cisplatin with 13-cis-retinoic acid and inter- 33. Olivari AJ, Glait HM, Guardo A, et al. Levamisole in squamous
feron-alpha in patients with advanced head and neck squamous cell carcinoma of the head and neck. Cancer Treat Rep
cell carcinoma. Anticancer Drugs 1999;10:369–74. 1979;63:983–90.
17. Herbst RS, Arquette M, Shin DM, et al. Phase II multicenter study 34. Padmanabhan TK, Balaram P, Vasudevan DM. Role of levamisole
of the epidermal growth factor receptor antibody cetuximab and immunotherapy as an adjuvant to radiotherapy in oral cancer. I. A
cisplatin for recurrent and refractory squamous cell carcinoma of three-year clinical follow up. Neoplasma 1987;34:627–32.
the head and neck. J Clin Oncol 2005a;23:5578–87. 35. Papac R, Minor DR, Rudnick S, et al. Controlled trial of metho-
18. Herold-Mende C, Karcher J, Dyckhoff G, Schirrmacher V. trexate and Bacillus Calmette-Guérin therapy for advanced head
Antitumor immunization of head and neck squamous cell carci- and neck cancer. Cancer Res 1978;38:3150–3.
noma patients with a virus-modified autologous tumor cell vac- 36. Perry CF, Stevens M, Rabie I, et al. Chemoprevention of head and
cine. Adv Otorhinolaryngol 2005;62:173–83. neck cancer with retinoids: a negative result. Arch Otolaryngol
19. Hong WK, Lippman SM, Itri LM, et al. Prevention of second pri- Head Neck Surg 2005;131:198–203.
mary tumors with isotretinoin in squamous-cell carcinoma of the 37. Pfister DG, Su YB, Kraus DH, et al. Concurrent cetuximab, cispla-
head and neck. N Engl J Med 1990;323:795–801. tin, and concomitant boost radiotherapy for locoregionally
20. Hussain M, Benedetti J, Smith RE, et al. Evaluation of 96-hour advanced, squamous cell head and neck cancer: a pilot phase II
infusion fluorouracil plus cisplatin in combination with alpha study of a new combined-modality paradigm. J Clin Oncol
interferon for patients with advanced squamous cell carcinoma of 2006;24:1072–8.
the head and neck: a Southwest Oncology Group study. Cancer 38. Recchia F, Lalli A, Lombardo M, et al. Ifosfamide, cisplatin, and
1995;76:1233–7. 13-Cis retinoic acid for patients with advanced or recurrent
21. Kerrebijn JD, Simons PJ, Balm AJ, et al. Thymostimulin enhance- squamous cell carcinoma of the head and neck: a phase I-II study.
ment of T-cell infiltration into head and neck squamous cell carci- Cancer 2001;92:814–21.
noma. Head Neck 1996;18:335–42. 39. Recchia F, Candeloro G, Di Staso M, et al. Maintenance immuno-
22. Khuri FR, Lee JJ, Lippman SM, et al. Randomized phase III trial therapy in recurrent or metastatic squamous cell carcinoma of the
of low-dose isotretinoin for prevention of second primary tumors head and neck. J Immunother 2008;31:413–9.
in stage I and II head and neck cancer patients. J Natl Cancer Inst 40. Riechelmann H, Wiesneth M, Schauwecker P, et al. Adoptive ther-
2006;98:441–50. apy of head and neck squamous cell carcinoma with antibody
23. Kimura T, Suzuki K, Motai H, et al. Final report of a randomized coated immune cells: a pilot clinical trial. Cancer Immunol
controlled study with streptococcal preparation OK-432 as a sup- Immunother 2007;56:1397–406.
plementary immunopotentiator for laryngeal cancer. Acta 41. Richman SP, Livingston RB, Gutterman JU, et al. Chemotherapy
Otolaryngol Suppl 1996;525:135–41. versus chemoimmunotherapy of head and neck cancer: report of a
24. Lam P, Yuen AP, Ho CM, et al. Prospective randomized study of randomized study. Cancer Treat Rep 1976;60:535–9.
post-operative chemotherapy with levamisole and UFT for head 42. Richtsmeier WJ, Koch WM, McGuire WP, et al. Phase I-II study
and neck carcinoma. Eur J Surg Oncol 2001;27:750–3. of advanced head and neck squamous cell carcinoma patients
25. Lippman SM, Kessler JF, Al-Sarraf M, et al. Treatment of advanced treated with recombinant human interferon gamma. Arch
squamous cell carcinoma of the head and neck with isotretinoin: a Otolaryngol Head Neck Surg 1990;116:1271–7.
phase II randomized trial. Invest New Drugs 1988;6:51–6. 43. Robert F, Blumenschein G, Herbst RS, et al. Phase I/IIa study of
26. Lippman SM, Batsakis JG, Toth BB, et al. Comparison of low- cetuximab with gemcitabine plus carboplatin in patients with che-
dose isotretinoin with beta carotene to prevent oral carcinogenesis. motherapy-naive advanced non-small-cell lung cancer. J Clin
N Engl J Med 1993;328:15–20. Oncol 2005;23:9089–96.
Robert O. Dillman 721

44. Roth AD, Abele R, Alberto P. 13-cis-retinoic acid plus interferon- 55. van Herpen CM, van der Laak JA, de Vries IJ, et al. Intratumoral
alpha: a phase II clinical study in squamous cell carcinoma of the recombinant human interleukin-12 administration in head and
lung and the head and neck. Oncology 1994;51:84–6. neck squamous cell carcinoma patients modifies locoregional
45. Schantz SP, Dimery I, Lippman SM, et al. A phase II study of inter- lymph node architecture and induces natural killer cell infiltration
leukin-2 and interferon-alpha in head and neck cancer. Invest New in the primary tumor. Clin Cancer Res 2005;11:1899–909.
Drugs 1992;10:217–23. 56. van Sándwich N, Dalesio O, Pastorino U, et al. EUROSCAN, a
46. Schulof RS. Thymic peptide hormones: basic properties and randomized trial of vitamin A and N-acetylcysteine in patients
clinical applications in cancer. Crit Rev Oncol Hematol with head and neck cancer or lung cancer. For the European
1985;3:309–76. Organization for Research and Treatment of Cancer Head and
47. Seixas-Silva JA Jr, Richards T, Khuri FR, et al. Phase 2 bioadju- Neck and Lung Cancer Cooperative Groups. J Natl Cancer Inst
vant study of interferon alfa-2a, isotretinoin, and vitamin E in 2000;92:977–86.
locally advanced squamous cell carcinoma of the head and neck: 57. Vermorken JB, Trigo J, Hitt R, et al. Open-label, uncontrolled,
long-term follow-up. Arch Otolaryngol Head Neck Surg multicenter phase II study to evaluate the efficacy and toxicity of
2005;131:304–7. cetuximab as a single agent in patients with recurrent and/or
48. Shin DM, Khuri FR, Murphy B, et al. Combined interferon-alfa, metastatic squamous cell carcinoma of the head and neck who
13-cis-retinoic acid, and alpha-tocopherol in locally advanced failed to respond to platinum-based therapy. J Clin Oncol
head and neck squamous cell carcinoma: novel bioadjuvant phase 2007;25:2171–7.
II trial. J Clin Oncol 2001;19:3010–7 58. Vlock DR, Snyderman CH, Johnson JT, et al. Phase Ib trial of the
49. Suen JY, Richman SP, Livingston RB, et al. Results of BCG adju- effect of peritumoral and intranodal injections of interleukin-2 in
vant immunotherapy in 100 patients with epidermoid carcinoma of patients with advanced squamous cell carcinoma of the head and
the head and neck. Am J Surg 1977;134:474–8. neck: an Eastern Cooperative Oncology Group trial. J Immunother
50. Taylor SG 4th, Sisson GA, Bytell DE, Raynor WJ Jr. A random- Emphasis Tumor Immunol 1994;15:134–9.
ized trial of adjuvant BCG immunotherapy in head and neck can- 59. Vlock DR, Andersen J, Kalish LA, et al. Phase II trial of inter-
cer. Arch Otolaryngol 1983;109:544–9. feron-alpha in locally recurrent or metastatic squamous cell carci-
51. Timar J, Forster-Horvath C, Lukitis J, et al. The effect of leukocyte noma of the head and neck: immunological and clinical correlates.
interleukin injection (Multikine) treatment on the peritumoral and J Immunother Emphasis Tumor Immunol 1996;19:433–42.
intratumoral subpopulation of mononuclear cells and on tumor 60. Voravud N, Lippman SM, Weber RS, et al. Phase II trial of
epithelia: a possible new approach to augmenting sensitivity to 13-cis-retinoic acid plus interferon-alpha in recurrent head and
radiation therapy and chemotherapy in oral cancer – a multicenter neck cancer. Invest New Drugs 1993;11:57–60.
phase I/II clinical Trial. Laryngoscope 2003;113:2206–17. 61. Wanebo HJ, Hilal EY, Pinsky CM, et al. Randomized trial of
52. Toma S, Bonelli L, Sartoris A, et al. 13-cis retinoic acid in head levamisole in patients with squamous cancer of the head and neck:
and neck cancer chemoprevention: results of a randomized trial a preliminary report. Cancer Treat Rep 1978;62:1663–9.
from the Italian Head and Neck Chemoprevention Study Group. 62. Woods JE, DeSanto LW, Ritts RE Jr. A controlled study of com-
Oncol Rep 2004;11:1297–305. bined methotrexate, BCG, and INH therapy for squamous cell
53. Urba SG, Forastiere AA, Wolf GT, Amrein PC. Intensive recombinant carcinoma of the head and neck. Surg Clin North Am 1977;57:
interleukin-2 and alpha-interferon therapy in patients with advanced 769–78.
head and neck squamous carcinoma. Cancer 1993;71:2326–31. 63. Vogl SE, Schoenfeld DA, Kaplan BH, et al. Methotrexate alone or
54. Valavaara R, Kortekangas AE, Nordman E, et al. Interferon com- with regional subcutaneous Corynebacterium parvum in the treat-
bined with irradiation in the treatment of operable head and neck ment of recurrent and metastatic squamous cancer of the head and
carcinoma. A pilot study. Acta Oncol 1992;31:429–31. neck. Cancer 1982;50:2295–300
21.10 Biological therapy of glioblastoma
ROBERT O. DILLMAN

Current Status of Therapy External Beam Radiation therapy (EBRT) is a stan-


dard component of the initial treatment of GBM, prefer-
for Glioblastomas ably following surgical resection, but is also used as
primary therapy in patients who are not surgical candi-
Glioblastoma multiforme (GBM) or glioblastoma is a
dates [30, 46]. Improvements in the delivery of radia-
collection of morphologically and genetically diverse
tion therapy have decreased toxicity, but partial brain
poorly differentiated neoplasms which collectively are
radiation to 6,000 cGy delivered over 5–6 weeks, which
the most common and most primary adult brain tumor
remains the standard of care for newly diagnosed GBM,
[50, 52]. More than 90% of GBMs are rapidly progres-
and the use of hyperfractionation radiation therapy have
sive primary tumors without clinical or histological
had little effect on survival [82]. Stereotactic radiosur-
evidence of a less malignant precursor lesion, typically
gery and gamma knife radiosurgery have enabled more
appearing in the elderly. Primary GBM are genetically
specific delivery of even higher doses of radiation [51].
characterized by loss of heterozygosity 10q (70% of
Although a randomized trial failed to show a survival
cases), EGFR amplification (36%), p16(INK4a) dele-
advantage for the addition of a radiosurgery boost in the
tion (31%), and PTEN mutations (25%). Secondary
initial management of GBM [72], more advanced uses
GBMs progress from lower grade astrocytomas that are
of this technique to areas around the edge of the radio-
diagnosed in younger patients. These are associated
graphically defined tumor are being explored.
with TP53 mutations and a G:C–>A:T mutation at
Adding chemotherapy to surgery and radiation ther-
CpG sites (60%). When these lower grade lesions
apy does improve the survival of patients with newly
progress to GBM, they acquire additional mutations
diagnosed GBM. A meta-analysis of randomized trials
including loss of heterozygosity 10q (70%). Primary
of adjunctive chemotherapy determined that the 2-year
GBM are associated with a much worse prognosis than
survival for 884 patients treated with radiation therapy
secondary GBM.
alone was only 16% compared to 23% for 1,538 patients
Despite recent therapeutic advances, GBM remains
who received radiation and chemotherapy [29]. Recently,
highly lethal when treated using standard measures [62,
concomitant and adjuvant chemoradiotherapy with oral
66, 75]. The refractoriness of GBM probably relates, in
temozolomide has become widely accepted as the stan-
part, to microscopic local extension which is beyond the
dard treatment for newly diagnosed GBM based on a
areas treated by surgery and radiation therapy [49].
randomized trial that showed improved median survival
Also, there is a limited therapeutic index between brain
from 12 months to 15 months, and the 2-year overall
and tumor which limits radiation therapy. Tumor cell
survival from 8% to 26% [74, 75] meta-analysis of 16
heterogeneity [6], and the limitations of the blood brain
randomized trials comparing chemotherapy to no che-
barrier [76], conspire to impair the effectiveness of most
motherapy showed a survival advantage for the use of
systemically administered therapies.
chemotherapy, but the impact of temozolomide was
Surgery is still the mainstay of standard therapy,
much greater than that of local or systemic carmustine
although a substantial number of patients are not
therapy [71].
candidates for surgery either because of the anatomic
location of the tumor or because of comorbid medical
conditions. Historically, following surgery alone, the
median survival for GBM was only 35 weeks, with a Biological Therapy
1-year survival of 33%, and a 2-year survival of 0%
[41, 70]. The ability to perform a near-total resection
of Glioblastoma
has been enhanced by microsurgical techniques with The potential clinical role of immunotherapy in the
navigational guidance, but GBM is rarely completely management of glioblastomas is under investigation.
resected, which is why surgery alone seldom, if ever, is The early forays with biological therapy, were not
curative [10, 42, 84]. associated with improved outcomes [37, 81]. There has

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 723
© Springer Science + Business Media B.V. 2009
724 Biological therapy of glioblastoma

recently been a rekindling of enthusiasm for vaccine Interferon-alpha


approaches, anti-angiogenesis agents, gene therapy
strategies, newer monoclonal antibodies, and intracra- Intratumor injections of leukocyte interferon were not
nial lymphocyte therapy, but these approaches are still associated with significant responses in 17 patients with
under development. malignant gliomas, 12 primary and five recurrent [40].
Most trials have utilized i.v. or s.c. routes to deliver
IFN-α systemically. In a trial in which IFN-α was given
Non-specific Immune Stimulators for 8 weeks, three patients with glioblastoma exhibited
a clinical response [54]. There appeared to be a possible
There was limited investigation of the early non-specific relationship between IFN-α gene status and tumor
immune stimulants such as bacillus Calmette-Guerin response. No responses were noted in 18 patients with
(BCG), Corynbacterium parvum, or levamisole. In one recurrent glioma who received thrice weekly s.c. IFN-α
small study five patients with malignant gliomas who with oral tamoxifen (Chang et al. 1998). In 29 patients
had undergone BCG inoculation were injected with recurrent anaplastic astrocytoma and glioblastoma,
intratumorally with PPD in an effort to induce an the combination of IFN-α with eflornithine, an irrevers-
intratumoral delayed type hypersensitivity reaction [2]. ible inhibitor of ornithine decarboxylase was associated
There was a subjective increase in inflammation based with no objective tumor responses [15]. As in other
on biopsies obtained before and after the PPD injections, tumor types, IFN-α was combined with chemotherapy
but there was no evidence of an antitumor effect. Other for patients with malignant gliomas. In one phase II trial
studies combined these agents with other treatment. In the combination of BCNU and IFN-α produced objec-
one trial BCG, and in another levamisole, were combined tive response in 7/21 (33%) [13]. In another phase II
with nitrosourea chemotherapy following surgery and/or trial in 35 patients with recurrent malignant glioma, the
radiotherapy, but neither agent appeared to enhance the combination of IFN-α and BCNU chemotherapy pro-
anti-tumor effect [93]. No survival benefit was obtained duced an objective response rate of 29% [14]. This
in a trial in which 25 patients with malignant glioma encouraging results led to a trial in which 383 malignant
were randomized to receive radiation therapy alone or glioma patients were treated with radiation therapy and
with oral levamisole [94]. BCNU, then 275 who had experienced disease progres-
sion at the end of therapy, were randomized to receive
more BCNU alone or in combination with IFN-α [16].
Interferons There was increased toxicity associated with the use of
As summarized in Table 1, limited anti-tumor activity IFN-α but no improvement in outcome.
has been documented in clinical trials in malignant Several trials have explored the use of IFN-α in com-
gliomas exploring the activity of lymphoblastoid inter- bination with radiation therapy. A retrospective review
feron [56], interferon alpha [Chang et al. 1998, 54], or of 175 patients with malignant gliomas suggested that
interferon beta [78, 90, 92]. the highest complete remission rates were in patients

Table 1. Single-agent activity of various biologicals in patients with malignant gliomas


Modality class Biological agent Lead author Patients Response rate (%)
Retinoid 13-cis retinoic acid (CRA) [69] 82 4
Retinoid 13-CRA and celecoxib [43] 25 0
Retinoid All-trans retinoic acid [96] 34 3
Retinoid All-trans retinoic acid [97] 30 10
Cytokine Interferon-α + tamoxifen [95] 18 0
Cytokine Interferon-α + eflornithine [15] 29 0
Cytokine Interferon-α [54] 30 10
Cytokine Lymphoblastoid interferon [56] 14 0
Cytokine Interferon-β [92] 14 0
Cytokine Interferon-β [78] 13 0
Cytokine Interferon-β [3] 21 19
Cytokine Interferon-β [92] 65 28
Monoclonal antibody EMD 55,900 (Mab 425) anti-EGFR [73] 16 0

EGFR = epidermal growth factor receptor.


Robert O. Dillman 725

who received the combination of radiation therapy, and clinical activity. No activity was noted in a small
nitrosourea, and IFN-α [87]. In 19 patients with primary study of 13 patients treated with a different IFN-β prep-
glioblastoma, only two of whom were able to undergo a aration (Fiblaferon) [78]. No anti-tumor effects were
near total resection, the combination of radiation ther- observed in six patients with glioblastoma multiforme
apy and thrice weekly 3–5 MIU s.c. of IFN-α was asso- who were treated with local injection of IFN-β through
ciated with a median survival of 7.5 months [22]. In an Ommaya reservoir [8]. In another trial, 55 of 109
vitro experiments suggested that combining 13-cis patients with newly diagnosed supratentorial glioblas-
retinoic acid with IFN-α was associated with even toma had stable disease following 60 Gy radiation ther-
greater radiosensitizing effects against glioblastoma cell apy, and were then treated with adjuvant IFN-β [19].
lines [45]. A subsequent trial of these agents was com- Their median survival of 13.4 months was encouraging
bined with radiation therapy in 40 patients, only four of based on comparisons to historical controls in the
whom underwent near total resection, and resulted in a Radiation Therapy Oncology Group glioma historical
median survival of 9.3 months with a 1-year survival of database. Japanese studies have combined IFN-β with
42% [23]. Several patients experienced severe radiation other agents resulting in encouraging survival results
necrosis, suggesting that enhanced radiosensitization [4, 80], but randomized trials are need to see whether
had been achieved in vivo. IFN-β adds anything to current regimens that utilize
Systemic IFN-α was given concurrently with intral- temozolomide.
esional interleukin-2 (IL-2) in five patients with recur-
rent malignant glioma [48]. IL-2 was given by this route
because of the induction of cerebral edema by high
Interferon-gamma (IFN-γ)
doses of systemic IL-2. As part of this study four patients In a small randomized trial, 31 patients with high-grade
had received IL-2 alone and cerebral edema was gliomas were randomized to receive intralesional
observed in two patients who received 50,000 IU intral- recombinant IFN-γ prior to and after radiation therapy,
esionally. In the combination study, systemic IFN-α or standard RT after surgical debulking [31]. Intratumoral
was given at dose of 3 MIU to 50 MIU i.v. weekly and IFN-γ was given thrice weekly for 4 weeks until radio-
IL-2 was limited to 10,000 IU intralesionally. The com- therapy, with doses escalated from 5 to 50 micrograms,
bination IL-2 + IFN-α was associated with weakness and after radiotherapy, was resumed at a dose of 50
and fatigue, and two patients exhibited increased peritu- micrograms twice a week for up to 9 weeks. Median
moral edema on radiologic scans. Because of toxicity, survival was 54–55 weeks in both arms.
this combination has not been pursued.
Retinoids
Interferon-beta Several laboratories have shown that retinoic acid
Results with interferon-beta (IFN-β) may be somewhat has growth-inhibitory activity against glioma cells
better than what has been reported with IFN-α. No in vitro in addition to immune modulating and cell
responses were observed in 14 patients with relapsed differentiation effects. A small number of trials have
high grade gliomas who were treated with lymphoblas- examined the activity of 13-cis-retinoic acid (13-CRA)
toid interferon [56]. IFN-β (Betaseron) was given by as a single agent in the treatment of malignant gliomas.
i.v. infusion over 30 min thrice weekly to 29 pediatric In a phase II trial conducted in 50 patients with pro-
patients with a variety of primary malignant brain gressive or recurrent disease after radiation and che-
tumors, including 12 high-grade astrocytomas and nine motherapy, there were three objective responses [91].
brain stem gliomas [3]. Objective responses were noted These same patients were included in a retrospective
in 4/21 evaluable patients. No objective responses were radiographic analysis of 85 patients with recurrent
recorded in ten patients in a pilot study in which IFN-β GBM who were treated with 13-CRA at MD Anderson
(Betaseron) was i.v. thrice weekly at a dose of 90 MIU Cancer Center, and an objective response rate of 4%
to 14 adult patients with recurrent malignant glioma was noted for 82 patients [69]. In a small pilot study,
[90]. In a larger trial using higher i.v. thrice weekly 13 patients with GBM and ten patients with anaplastic
dosing, 15/65 patients with recurrent or non-responsive astrocytoma who had achieved a complete response
malignant glioma had an objective response [92]. after primary surgery, radiation therapy, and chemother-
Histologically, 41 had glioblastoma and 24 had apy, were given 13-CRA at a dose of 60 mg/m2 daily for
anaplastic astrocytoma. A thrice weekly dose of 180 3 weeks of each 4-week cycle with a dose escalation of
MIU was felt to be the optimal dose based on toxicity up to 100 mg/m2 [85]. Treatment was well tolerated,
726 Biological therapy of glioblastoma

but even in this highly selected group of patients with Intravenous adoptive cell therapy approaches
a high representation of non-GBM histology, the
median survival was still only 17 months from the time Although the blood brain barrier is perceived as a limi-
of entry into the study. tation to systemic infusional approaches for adoptive
Other trials have combined 13-CRA with other cellular therapy, some investigators have attempted this
agents. 13-CRA plus the COX-2 inhibitor celecoxib as well. In terms of systemic adoptive cell therapy, one
yielded no responses in 25 patients with recurrent glio- group has tried i.v. approaches with two different cell
blastoma [43]. In a large phase II trial, 88 patients with products. In the first study, following resection of recur-
recurrent or progressive malignant glioma were treated rent anaplastic astrocytoma or GBM, 15 patients were
with a combination of temozolomide plus 13-CRA [38]. immunized with BCG and their own irradiated tumor
Histolologies included 40 glioblastoma multiforme, 28 cells, then 2 weeks later peripheral blood mononuclear
astrocytoma, 14 oligodendroglioma, and six mixed cells were harvested by leukopheresis, cultured in vitro
glioma. There were ten responses among 84 evaluable with irradiated autologous tumor cells and IL-2 then
patients (15%). 13-CRA was added to radiation therapy infused i.v. without additional IL-2 [33]. Feasibility and
and temozolomide in the treatment of 61 adults with safety were confirmed, but clinical benefit was not
newly diagnosed supratentorial glioblastoma [17]. The apparent. In the second study, these same investigators
median survival was 12.2 months and 1-year survival took nine patients with recurrent, but surgically resect-
was estimated to be 57%. 13-CRA was added to radia- able anaplastic astrocytoma or GBM, tapered them off
tion therapy and IFN-α in the treatment of 40 adults steroids after total surgical resection and immunized
with newly diagnosed supratentorial glioblastoma, only them with autologous cancer cells from the resected dis-
four of whom underwent near total resection [23]. The ease that had been admixed with BCG [86]. They then
median survival was 9.3 months, and 1-year survival collected peripheral blood mononuclear cells which
was 42%. were stimulated and expanded in vitro with anti-CD3
All-trans-retinoic acid (ATRA) has also been explored monoclonal antibody and IL-2, then 1010–1011 activated
as a biotherapy for malignant gliomas. In a phase II trial, cells were infused into the patients. The study design
one of 34 evaluable patients with recurrent glioma had makes it hard to interpret claims of objective tumor
an objective response after two cycles of 120–150 mg/m2 response in these patients, but two patients were consid-
given daily for 3 weeks of each 4-week cycle (Kaba 1997). ered disease-free more than 4 years later.
The authors concluded that single agent ATRA has no T cells derived from lymph nodes have also been
significant activity against recurrent cerebral gliomas. infused i.v. Following surgery, 12 patients with newly
In another trial, 30 patients with malignant gliomas, diagnosed gliomas (half lower grade, half GBM) were
including 14 with glioblastoma multiforme and 14 with vaccinated i.d. with GM-CSF + short-term cultured
anaplastic astrocytoma, three (10%) had an objectivew autologous irradiated tumor cells, then lymphocytes
response after treatment with all trans-retinoic acid from resected draining lymph nodes were stimulated
(ATRA) [97]. with staphylococcal enterotoxin A for 48 h and then
cultured in IL-2-containing medium 6–8 days, then
109–1010 cells were administered i.v. to the patients [55].
Adoptive Cell Therapy The cell products were mostly CD4+. Four patients
The use of LAK and other cell therapies are being were interpreted as having partial regression of residual
explored in the management of malignant brain tumors tumor.
[77]. Systemic IL-2 often causes edema around brain
tumors and sometimes hemorrhage. For this reason,
patients with brain tumors have been excluded from
Localized Adoptive Cell Therapy Approaches
most treatment protocols utilizing systemic IL-2. Most efforts with adoptive cellular therapy for malignant
However, many investigators have explored the feasi- gliomas have utilized localized approaches. In a phase
bility of direct instillation of activated cells, with or I trial, nine patients received LAK and/or IL-2, and one
without IL-2, directly into brain tumors or areas in patient received both together [36]. Escalating doses of
which brain tumors have been excised. The validity of LAK cells, 108–1010 or recombinant IL-2, 104–106 units
alleged objective tumor regressions in many of these were suspended in 5 ml of Hanks balanced salt solution
studies is in question, since they LAK were injected into and were directly injected into the tissue surrounding the
a post-surgical tumor bed making interpretation of surgical cavity remaining after tumor extirpation using
response quite difficult. multiple injections via 20 gauge needles. LAK cells were
Robert O. Dillman 727

developed within 2–3 days of surgery. There was no following therapy ranged from 3.5 months to >3 years,
comment regarding whether patients received steroids with a median survival of 4.5 months. Three of nine
before and/or immediately after surgery and immuno- patients with recurrent GBM, who were treated with
therapy. None of the ten patients were felt to have exhib- LAK and IL-2 administered directly into the tumor cav-
ited toxicity beyond that normally seen post-craniotomy. ity via an Ommaya reservoir, were interpreted as having
In another trial, at the time of tumor resection, a catheter an objective response to treatment, although long-term
was implanted in the cavity and used to deliver 6 MIU/ survival was not achieved [9]. One of ten patients with
day IL-2 by continuous infusion for 5 days, with or with- recurrent GBM, who were treated similarly with local
out LAK [11]. In five patients, LAK cells were infused instillations of LAK and IL-2, were felt to have had a
into the cavity on days 1, 3, and 5 after surgery, and eight partial response [67].
patients received IL-2 alone. Fever, confusion, and cere- Hayes et al. gave LAK cells to 19 out of 44 possible
bral edema were observed in all patients and there was patients with recurrent glioma who were considered
no evidence of clinical benefit. candidates for resection [32]. Fifteen tumors were clas-
Several trials utilized instillation of LAK with IL-2. sified as gliomas (grade 4) and four as anaplastic astro-
In one trial, 23 recurrent glioma patients were treated cytomas (grade 3). Patients underwent leukapheresis
with 107–108 LAK cells with 50–400 units of a recombi- within 1–6 weeks of resection after discontinuation or
nant IL-2 via Ommaya reservoir [88]. LAK were gener- reduction of corticosteroids. LAK cells were produced
ated 4–6 days prior to instillation. Many patients had by incubating the mononuclear cells in 6 million inter-
substantial neurological deficits at the time of treat- national units/ml of IL-2 for 4 days. A total of 0.5 – 1 × 106
ment.. Side effects noted were chills and fever. LAK were infused via Ommaya reservoir on day 1, with
Substantial improvement occurred in at least three a maximum-tolerated IL-2 dose of 1.2 MIU/ml IL-2.
patients. In another small study, 13 recurrent glioma The same dose of IL-2 was infused without cells on
patients were treated with LAK generated 3–5 days days 3, 5, 8, 10 and 12, which constituted one treatment
prior to craniotomy and suspended in 5–10 ml saline cycle. The 12-day cycle of therapy was repeated about 2
with 106 units of Cetus IL-2 [47]. Daily injections of 106 weeks later. Patients who had not progressed after two
units of IL-2 were given for 3 additional days. A second treatment cycles could be re-treated at 12-week intervals.
cycle of IL-2/LAK was infused via Ommaya reservoir Since all of these patients had maximum tumor debulk-
1–2 weeks later. All patients had symptoms of aseptic ing at the time of surgery, tumor response could not have
meningitis, including increased intracranial pressure, been a meaningful endpoint of this trial, although the
headache, fever, and malaise. The authors commented authors reported one complete and two partial responses.
on the difficulty of attributing some of these effects However, median survival from the date of operation
to IL-2 as opposed to post-craniotomy. They felt that was 53 weeks for the 15 glioblastoma patients, with a
the approach was sufficiently safe to try on newly diag- 1-year survival of 53% compared to a median survival
nosed patients, although all of these patients were on of only 26 weeks, versus a 1-year survival rate of 5% for
dexamethasone, which is known to inhibit IL-2 and 18 contemporary controls who underwent debulking
LAK activity. Barba et al. treated ten recurrent glioma and then received chemotherapy.
patients with LAK cells and IL-2 given via a catheter The largest phase II trial to date enrolled 40 patients
stereotactically placed in the tumor bed and connected with pathologically confirmed GBM at the time of sur-
to an Ommaya reservoir [5]. Three weeks after surgery, gery, who subsequently had placement of autologous
therapy was begun with 1010 LAK cells in 10 cc of LAK cells into the tumor cavity [25]. LAK cells were
saline. IL-2 was infused for 5 days via the same cath- generated by incubating peripheral blood mononuclear
eter at 10,000–60,000 U/kg/dose, three times per day. cells with interleukin-2 (IL-2) for 3–5 days in vitro.
Neurologic side effects occurred in all patients because Because of the inherent difficulties in interpreting a
of intracerebral edema. Glucocortocoid therapy was tumor response in such patients who have undergone
limited during the immunotherapy, but patients were surgical resection, survival was compared to contempo-
then treated with high-dose dexamethasone after com- rary matched controls. Median survival post-LAK was
pleting IL-2/LAK. One of nine evaluable patients was 9.0 months; 1-year survival was 34%. Gender, age,
considered to have had a beneficial response. Lillehei et al. location of tumor, and number of cells implanted were
treated 11 patients with recurrent high-grade gliomas unrelated to outcome. Inclusion of IL-2 at the time of
[98]. Peripheral blood lymphocytes were stimulated in cell instillation was associated with improved survival
one of two different culture conditions and placed into (p = .097). Median survival from date of original diag-
the tumor cavity with IL-2 in a plasma clot. The survival nosis for 31 patients who had GBM at initial diagnosis
728 Biological therapy of glioblastoma

was 17.5 months versus 13.6 months for a control vivo binding was confirmed. Eight patients with primary
group of 41 contemporary GBM patients (p = .012). or recurrent EGF-R-positive glioblastomas were treated
The median survival rates were higher than reported intratumorally twice weekly through an implantable
in most published series of patients who had undergone catheter with total doses of 4 mg and 120 mg of Mab 425
re-operation for recurrent GBM. This approach is [83]. The treatment induced an intense inflammatory
currently being combined with surgery, radiation reaction and a substantial necrosis. In an early pilot
therapy and temozolomide and preliminary results are study, 25 patients with primary malignant glioma were
encouraging [26]. given i.v. or intra-arterial I-131-labeled anti EGFR (425)
In a slightly different approach, peripheral blood 4–6 weeks following initial surgery and radiation ther-
lymphocytes were stimulated with both phytohemag- apy [12]. The median survival was 16 months for the 15
glutin (PHA) and IL-2 to produce what were called patients with GBM. During 1987–1997, this group
mitogen activated killer (MAK) cells. In a phase I/II treated 180 patients following surgery and radiation
trial involving 55 patients with various glioma histolo- therapy, with and without chemotherapy, with an aver-
gies, MAK were generated over 7–10 days prior to cran- age total dose of 140 mCi given typically as three weekly
iotomy, then suspended in 15 ml of autologous injections [27]. The median survival for patients with
EDTA-plasma (total volume 15–20 ml) for placement in GBM was 13 months.
the tumor bed following surgery [34]. Therapy was gen- The humanized anti-EGFR Mab h-R3 was given to
erally well tolerated, other than fever and nausea, and 29 patients with newly diagnosed malignant gliomas
most patients were discharged within 5–7 days after including 16 glioblastoma, 12 anaplastic astrocytoma
craniotomy. There was no effort made to judge tumor and one anaplastic oligodendroglioma [58]. Following
response, but survival for the patient who underwent debulking surgery, patients received six 200 mg infu-
treatment was considered encouraging. MAK were also sions of h-R3 weekly during radiotherapy. Interestingly,
used to treat 19 patients with high-grade gliomas [39] At no patients developed acneiform rash, and there were no
the time of treatment both PHA and IL-2 were placed in allergic reactions. Median survival was 17 months for
the tumor site as well in 16 patients. Three patients were the glioblastoma patients. A trial of the anti-EGFR mab
felt to have a significant decrease in radiologic enhance- cetuximab with temozolomide and radiation therapy is
ment of the tumor area. being conducted in patients with previously untreated
Because of the paucity of lymphocytes found in glioblastoma [21].
malignant gliomas, few investigators have explored the For many years investigators at Duke University have
use of tumor infiltrating lymphocytes (TIL) as adoptive been testing monoclonal antibodies (Mab) that react
cell therapy for GBM. Following surgical resection and tenascin, which is a large, disulfide-bonded glycopro-
placement of an Ommaya reservoir, six patients were tein of the extracellular matrix that is highly expressed
treated with low doses of TIL every 2 weeks concurrent in the stroma of gliomas, but not normal brain tissue.
with thrice weekly low doses of IL-2 for 1 month [57]. Radiommunotherapy trials have been carried out with
Three patients were interpreted as having a partial murine Mab 81C6, but a chimeric version has also been
response to the treatment. tested. In a phase I trial conducted in 34 previously irra-
diated patients with recurrent or metastatic brain tumors,
the maximum tolerated dose for 131I-labeled 81C6
Antibody Therapy administered through an Ommaya reservoir into the
The epidermal growth factor receptor (EGFR) is surgical resection cavity was 120 mCi based on dose-
expressed on malignant glioma cells; and therefore has limiting delayed neurologic toxicities [7]. Patients with
been a target for antibody-based therapy of glioblas- recurrent GBM had an encouraging survival of about
toma. EMD55900 (Mab 425) is a murine IgG2A Mab 13 months. The same MTD was determined in a phase
directed against EGFR. Mab 425 was given i.v. to 16 I trial involving 42 newly diagnosed patients who had
patients previously treated with surgery, radiotherapy not received prior radiation therapy or chemotherapy
and chemotherapy for high grade supratentorial malig- [18]. Median survival for patients with GBM was 16
nant gliomas (11 GBM) [73]. There were no tumor months. A small number of patients had to undergo sur-
responses including the last six patients who received gical excision of symptomatic radionecrosis. In a subse-
200 mg thrice weekly for 4 weeks. Single doses of 20, quent trial, newly diagnosed patients were treated with
40, 100, 200, or 400 mg of the murine MAb 425 were direct injections of 131-I-labeled anti-tenascin murine
given i.v. before surgery to 30 patients with malignant 81C6 into the surgical cavity followed by conventional
brain tumors [28]. Treatment was well-tolerated and in external-beam radiotherapy and chemotherapy [1].
Robert O. Dillman 729

Biopsies from 15 patients all showed tumor and/or radi- excluded because of concerns that there might be an
onecrosis. Subsequently, a phase II trial was carried out increased risk of intracerebral hemorrhage. However, in
in 33 newly diagnosed patients (27 GBM) using 120 mCi patients with malignant gliomas, there were no instances
of 131-I-labeled murine 81C6 injected directly into the of central nervous system hemorrhage, but four patients
surgical resection cavity prior to receiving conventional (12%) experienced thromboemboic complications.
external-beam radiotherapy, followed by a year of alky-
lator-based chemotherapy [59]. Median survival for the Vaccine Therapy
GBM patients was 18 months. In a phase II trial in 43
patients with recurrent gliomas (33 GBM), 100 mCi of A variety of vaccine approaches are being explored in
131-I-labeled murine 81C6 was injected directly into patients with glioblastomas. These include Newcastle-
the surgical resection cavity. The median overall sur- Disease-Virus (NDV) infected autologous tumor cells
vival for patients with recurrent GBM was 15 months derived from short term cultures [68], irradiated autolo-
[60]. In animal models, a 131-I-labeled IgG2/mouse gous tumor cells from continuously proliferating cell
chimeric antitenascin 81C6 (ch81C6) construct exhib- cultures [24], autologous dendritic cells pulsed with
ited higher tumor accumulation and enhanced stability lysates of autologous tumor [89], autologous dendritic
compared to the murine antibody, but in 43 patients the cells pulsed with of acid-eluted peptides from autolo-
maximum tolerated dose was only 80 mCi because of gous tumor [44], autologous tumor vaccines prepared
dose-limiting hematologic toxicity associated with a from formalin-fixed and/or paraffin-embedded tumor
longer serum half life [61]. tissue obtained at the time of surgery [35], autologous
Another group has conducted trials with murine anti- glioma cells and interleukin (IL)-4 gene transfected
tenascin Mabs called BC-2 and BC-4, which were radi- fibroblasts [53] and intratumor injection of Herpses
olabeled with I-131. In one trial, an early analysis of 17 Simplex retroviral vector-producing cells, followed by
of 23 patients with recurrent malignant gliomas who intravenous ganciclovir [20]. These were mostly small
had received intracavitary doses ranging from 15 mCi to pilot studies to demonstrate safety, involving only 10–15
57 mCi, showed a median survival of about 16 months patients in most instances.
[63]. In an expanded group of 50 patients (24 with pri-
mary and 26 with recurrent disease) the median survival
was 18 months for 26 patients with recurrent disease
and 20 months in 24 patients with newly diagnosed
Summary
lesions [64]. BC-4 was labeled with Y-90 and adminis- Currently there are no biological products that have
tered to another 20 patients with recurrent high grade a regulatory approval for marketing as treatment for
gliomas (18 GBM) using doses ranging from 5–30 mCi malignant gliomas, although several appear to have some
[65]. The maximum tolerated dose to the brain was activity. Whether any of these products become widely
25 mCi delivering 3,200 cGy/mCi. available and paid for by payment providers will depend
In patients who have experienced recurrence of GBM, on the choices made by large biotechnology companies.
encouraging results for the combination of irinotecan Because of current treatment paradigms, a successful
and the anti-vascular endothelial growth factor mono- biological will almost certainly have to be integrated into
clonal antibody bevacizumab have been reported. In a existing treatment algorithms as part of treatment of newly
phase II trial, adult patients with recurrent anaplastic diagnosed glioblastoma. Unintentional patient selection
astrocytoma or glioblastoma (grade III or IV glioma) issues and difficulties in measuring a tumor response,
received bevacizumab at 10 mg/kg and irinotecan i.v. especially as these relate to local instillation of cell therapies
every 2 weeks of a 6-week cycle [79]. The dose of irino- and antibodies, makes it difficult to determine the clinical
tecan was determined based on whether patients were significance of apparent improvements in survival.
taking antiepileptic drugs that induced hepatic enzymes Randomized trials will be needed to determine if any of
that accelerate metabolism of irinotecan. Patients taking these therapies are adding benefit.
enzyme-inducing antiepileptic drugs received irinote-
can at 340 mg/m2, while other patients received irinote-
can at 125 mg/m2. Although the significance of
radiographic changes in treated gliomas is questionable,
References
1. Akabani G, Reardon DA, Coleman RE, et al. Dosimetry and radio-
based on study design, the authors alleged an overall graphic analysis of 131I-labeled anti-tenascin 81C6 murine mono-
response rate of 63% for all patients. In other trials of clonal antibody in newly diagnosed patients with malignant
bevacizumab, patients with brain metastases were gliomas: a phase II study. J Nucl Med 2005;46:1042–51.
730 Biological therapy of glioblastoma

2. Albright L, Seab JA, Ommaya AK, et al. Intracerebral delayed 21. Combs SE, Heeger S, Haselmann R, et al. Treatment of primary
hypersensitivity reactions in glioblastoma multiforme patients. glioblastoma multiforme with cetuximab, radiotherapy and temo-
Cancer 1977;39:1331–6. zolomide (GERT) – phase I/II trial: study protocol. BMC Cancer
3. Allen J, Packer R, Bleyer A, et al. Recombinant interferon beta: a 2006;6:133.
phase I–II trial in children with recurrent brain tumors. J Clin 22. Dillman RO, Wiemann M, Oldham RK, et al. Interferon alpha-2a
Oncol 1991;9:783–8. and external beam radiotherapy in the initial management of
4. Aoki T, Takahashi JA, Ueba T, et al. Phase II study of nimustine, patients with glioma: a pilot study of the National Biotherapy
carboplatin, vincristine, and interferon-beta with radiotherapy for Study Group. Cancer Biother 1995;10:265–71.
glioblastoma multiforme: experience of the Kyoto Neuro- 23. Dillman RO, Shea WM, Tai DF, et al. Interferon-alpha2a and
Oncology Group. J Neurosurg 2006;105:385–91. 13-cis-retinoic acid with radiation treatment for high-grade glioma.
5. Barba D, Saris S, Holder C, et al. Intramural LAK cell and interleukin-2 Neuro Oncol 2001;3:35–41.
therapy of human gliomas. J Neurosurg 1989;70:175–82. 24. Dillman RO, Beutel LD, Barth NM, et al. Irradiated cells from
6. Bigner DD. Biology of gliomas: potential clinical implications of autologous tumor cell lines as patient-specific vaccine therapy in
glioma cellular heterogeneity. Neuro Surg 1981;9:320–26. 125 patients with metastatic cancer: induction of delayed-type
7. Bigner DD, Brown MT, Friedman AH, et al. Iodine-131-labeled hypersensitivity to autologous tumor is associated with improved
antitenascin monoclonal antibody 81C6 treatment of patients with survival. Cancer Biother Radiopharm 2002;17:51–66.
recurrent malignant gliomas: phase I trial results. J Clin Oncol 25. Dillman RO, Duma CM, Schiltz PM, et al. Intracavitary placement
1998;16:2202–12. of autologous lymphokine-activated killer (LAK) cells after resec-
8. Bioardi A, Silvani A, Milanesi I, et al. Local immunotherapy (beta- tion of recurrent glioblastoma. J Immunother 2004;27:398–404.
IFN) and systemic chemotherapy in primary glial tumors. Ital J 26. Dillman RO, Duma C, Schiltz PM, et al. Intracavitary placement
Neurol Sci 1991;12:163–8. of lymphokine-activated killer (LAK) cells after resection of pri-
9. Bioardi A, Silvani A, Ruffini PA, et al. Loco-regional immuno- mary glioblastoma: a planned interim analysis. International
therapy with recombinant interleukin-2 and adherent lymphokine- Society for Biological Therapy of Cancer 20th Annual Meeting,
activated killer cells (A-LAK) in recurrent glioblastoma patients. Arlington, VA. 10–13 November 2005. J Immunother 2005;
Cancer Immunol Immunother 1994;39:193–7. 28:612.
10. Black PM. Brain tumors. N Engl J Med 1991;324:1471–76, 27. Emrich JG, Brady LW, Quang TS, et al. Radioiodinated (I-125)
1555–14. monoclonal antibody 425 in the treatment of high grade glioma
11. Blancher A, Roubinet F, Grancher AS, et al. Local immunotherapy patients: ten-year synopsis of a novel treatment. Am J Clin Oncol
of recurrent glioblastoma multiforme by intracerebral perfusion of 2002;25:541–6.
interleukin-2 and LAK cells. Eur Cytokine Netw 1993;4:331–41. 28. Faillot T, Magdelenat H, Mady E, et al. A phase I study of an anti-
12. Brady LW, Miyzmoto C, Woo DV, et al. Malignant astrocytomas epidermal growth factor receptor monoclonal antibody for the
treated with iodine-125 labeled monoclonal antibody 425 against treatment of malignant gliomas. Neurosurgery 1996;39:478–83.
epidermal growth factor receptor: a phase II trial. Int J Radiat 29. Fine HA, Dear KBG, Loeffler JS, et al. Meta-analysis of radiation
Oncol Biol Phys 1992;22:225–30. therapy with and without adjuvant chemotherapy for malignant
13. Brandes AA, Scelzi E, Zampieri P, et al. Phase II trial with BCNU gliomas in adults. Cancer 1993;71:2582–97.
plus alpha-interferon in patients with recurrent high-grade gliomas. 30. Fiveash JB, Spencer SA. Role of radiation therapy and radiosur-
Am J Clin Oncol 1997;20:364–7. gery in glioblastoma multiforme. Cancer J 2003; 9:222–9.
14. Buckner JC, Brown LD, Kugler JW, et al. Phase II evaluation of 31. Farkkila M, Jaaskelainen J, Kallio M, et al. Randomised, con-
recombinant interferon alpha and BCNU in recurrent glioma. J trolled study of intratumoral recombinant gamma-interferon treat-
Neurosurg 1995;82:430–5. ment in newly diagnosed glioblastoma. Br J Cancer 1994;70:
15. Buckner JC, Burch PA, Cascino TL, et al. Phase II trial of recom- 138–41.
binant interferon-alpha-2a and eflornithine in patients with recur- 32. Hayes RL, Koslow M, Hiesiger EM, et al. Improved long term
rent glioma. J Neurooncol 1998;36:65–70. survival after intracavitary interleukin-2 and lymphokine-activated
16. Buckner JC, Schomberg PJ, McGinnis WL, et al. A phase III study killer cells for adults with recurrent malignant glioma. Cancer
of radiation therapy plus carmustine with or without recombinant 1995;76:840–52.
interferon-alpha in the treatment of patients with newly diagnosed 33. Holladay FP, Heitz-Turner T, Bayer WL, Wood GW. Autologous
high-grade glioma. Cancer 2001;92:420–33. tumor cell vaccination combined with adoptive cellular immuno-
17. Butowski N, Prados MD, Lamborn KR, et al. A phase II study of therapy in patients with grade III/IV astrocytoma. J Neurooncol
concurrent temozolomide and cis-retinoic acid with radiation for 1996;27:179–89.
adult patients with newly diagnosed supratentorial glioblastoma. 34. Ingram M, Jacques S, Freshwater DB, et al. Salvage immunother-
Int J Radiat Oncol Biol Phys 2005;61:1454–9. apy of malignant glioma. Arch Surg 1987;122:183–6.
18. Cokgor I, Akabani G, Kuan CT, et al. Phase I trial results of iodine- 35. Ishikawa E, Tsuboi K, Yamamoto T, et al. Clinical trial of autolo-
131-labeled antitenascin monoclonal antibody 81C6 treatment of gous formalin-fixed tumor vaccine for glioblastoma multiforme
patients with newly diagnosed malignant gliomas. J Clin Oncol patients. Cancer Sci 2007;98:1226–33.
2000;18:3862–72. 36. Jacobs SK, Wilson DJ, Kornblith PL, Grimm EA. Interleukin-2 or
19. Colman H, Berkey BA, Maor MH, et al. Phase II Radiation autologous lymphokine-activated killer cell treatment of malig-
Therapy Oncology Group trial of conventional radiation therapy nant glioma: phase I trial. Cancer Res 1986;46:2101–04.
followed by treatment with recombinant interferon-beta for 37. Jaeckle KA. Immunotherapy of malignant gliomas. Semin Oncol
supratentorial glioblastoma: results of RTOG 9710. Int J Radiat 1994;21:249–59.
Oncol Biol Phys 2006;66:818–24. 38. Jaeckle KA, Hess KR, Yung WK, et al. Phase II evaluation of
20. Colombo F, Barzon L, Franchin E, et al. Combined HSV-TK/IL-2 temozolomide and 13-cis-retinoic acid for the treatment of recur-
gene therapy in patients with recurrent glioblastoma multiforme: rent and progressive malignant glioma: a North American Brain
biological and clinical results. Cancer Gene Ther 2005;12:835–48. Tumor Consortium study. J Clin Oncol 2003;21:2305–11.
Robert O. Dillman 731

39. Jeffes EW 3rd, Beamer YB, Jacques S, et al. Therapy of recurrent receptor (EGFR) antibody h-R3: report from a phase I/II trial.
high grade gliomas with surgery, and autologous mitogen acti- Cancer Biol Ther 2006;5:375–9.
vated IL-2 stimulated killer (MAK) lymphocytes: I. Enhancement 59. Reardon DA, Akabani G, Coleman RE, et al. Phase II trial of
of MAK lytic activity and cytokine production by PHA and clini- murine (131)I-labeled antitenascin monoclonal antibody 81C6
cal use of PHA. J Neurooncol 1993;15:141–55. administered into surgically created resection cavities of patients
40. Jereb B, Petric J, Lamovec J, et al. Intratumor application of with newly diagnosed malignant gliomas. J Clin Oncol 2002;20:
human leukocyte interferon-alpha in patients with malignant brain 1389–97.
tumors. Am J Clin Oncol 1989;12:1–7. 60. Reardon DA, Akabani G, Coleman RE, et al. Salvage radioimmu-
41. Kelley KA, Kirkwood JM, Kapp DS. Glioblastoma multiforme: notherapy with murine iodine-131-labeled antitenascin monoclo-
pathology, natural history, and treatment. Cancer Treat Rep nal antibody 81C6 for patients with recurrent primary and
1984;11:1–26. metastatic malignant brain tumors: phase II study results. J Clin
42. Laws DR Jr, Thapar K. Brain tumors. CA Cancer J Clin Oncol 2006a;24:115–22.
1993;43:263–71. 61. Reardon DA, Quinn JA, Akabani G, et al. Novel human IgG2b/
43. Levin VA, Giglio P, Puduvalli VK, et al. Combination chemother- murine chimeric antitenascin monoclonal antibody construct radi-
apy with 13-cis-retinoic acid and celecoxib in the treatment of olabeled with 131I and administered into the surgically created
glioblastoma multiforme. J Neurooncol 2006;78:85–90. resection cavity of patients with malignant glioma: phase I trial
44. Liau LM, Prins RM, Kiertscher SM, et al. Dendritic cell vaccina- results. J Nucl Med 2006b;47:912–8.
tion in glioblastoma patients induces systemic and intracranial 62. Reardon DA, Rich JN, Friedman HS, Bigner DD. Recent advances
T-cell responses modulated by the local central nervous system in the treatment of malignant astrocytoma. J Clin Oncol 2006;24:
tumor microenvironment. Clin Cancer Res 2005;11:5515–25. 1253–65.
45. Malone C, Schiltz PM, Nayak SK, et al. Combination interferon- 63. Riva P, Arista A, Tison V, et al. Intralesional radioimmunotherapy
α2a and 13 cis-retinoic acid enhances radiosensitization of human of malignant gliomas. An effective treatment in recurrent tumors.
malignant glioma cells in vitro. Clin Cancer Res 1999;5:417–23. Cancer 1994;73(3 Suppl):1076–82.
46. McDermott MW, Berger MS, Kunwar S, et al. Stereotactic radio- 64. Riva P, Arista A, Franceschi G, et al. Local treatment of malignant
surgery and interstitial brachytherapy for glial neoplasms. J gliomas by direct infusion of specific monoclonal antibodies
Neurooncol 2004;69:83–100. labeled with 131I: comparison of the results obtained in recurrent
47. Merchant RE, Grant AJ, Merchant LH, Young HF. Adoptive and newly diagnosed tumors. Cancer Res 1995;55(23 Suppl):
immunotherapy for recurrent glioblastoma multiformi using lym- 5952s–6s.
phokine activated killer cells and recombinant interleukin-2. 65. Riva P, Franceschi G, Frattarelli M, et al. Loco-regional radioim-
Cancer 1988;62:665–71. munotherapy of high-grade malignant gliomas using specific
48. Merchant RE, McVicar DW, Merchant LH, Young HF. Treatment monoclonal antibodies labeled with 90Y: a phase I study. Clin
of recurrent malignant glioma by repeated intracerebral injections Cancer Res 1999;5(10 Suppl):3275s–80s.
of human recombinant interleukin-2 alone or in combination with 66. Robins HI, Chang S, Butowski N, Mehta M. Therapeutic advances
systemic interferon-alpha. Results of a phase I clinical trial. J for glioblastoma multiforme: current status and future prospects.
Neurooncol 1992;12:75–83. Curr Oncol Rep 2007;9:66–70.
49. McVie JG. The therapeutic challenge of gliomas. Eur J Cancer 67. Sankhla SK, Nadkarni JS, Bhagwati SN. Adoptive immunotherapy
1993;29A:936–9. using lymphokine-activated killer (LAK) cells and interleukin-2
50. Miller CR, Perry A. Glioblastoma. Arch Pathol Lab Med for recurrent malignant primary brain tumors. J Neurooncol
2007;131:397–406. 1996;27:133–40.
51. Nwokedi EC, DiBiase SJ, Jabbour S, et al. Gamma knife stereot- 68. Schneider T, Gerhards R, Kirches E, Firsching R. Preliminary
actic radiosurgery for patients with glioblastoma multiforme. results of active specific immunization with modified tumor cell
Neurosurgery 2002;50:41–47. vaccine in glioblastoma multiforme. J Neurooncol 2001;53:39–46.
52. Ohgaki H, Kleihues P. Genetic pathways to primary and secondary 69. See SJ, Levin VA, Yung WK, et al. 13-cis-retinoic acid in the treatment
glioblastoma. Am J Pathol 2007;170:1445–53. of recurrent glioblastoma multiforme. Neuro Oncol 2004;6:253–8.
53. Okada H, Lieberman FS, Walter KA, et al. Autologous glioma cell 70. Silverstein MD, Cascino TL, Harmsen WS. High-grade astrocy-
vaccine admixed with interleukin-4 gene transfected fibroblasts in tomas: resource use, clinical outcomes, and cost of care. Mayo
the treatment of patients with malignant gliomas. J Transl Med Clin Proc 1996;71:936–44.
2007;5:67. 71. Spiegel BM, Esrailian E, Laine L, Chamberlain MC. Clinical
54. Olson JJ, James CD, Lawson D, et al. Correlation of the response impact of adjuvant chemotherapy in glioblastoma multiforme: a
of recurrent malignant gliomas treated with interferon alpha with meta-analysis. CNS Drugs 2007;21(9):775–87.
tumor interferon alpha gene content. Int J Oncol 2004;25:419–27. 72. Souhami L, Seiferheld W, Brachman D, et al. Randomized com-
55. Plautz GE, Miller DW, Barnett GH, et al. T cell adoptive immuno- parison of stereotactic radiosurgery followed by conventional
therapy of newly diagnosed gliomas. Clin Cancer Res 2000;6: radiotherapy with carmustine to conventional radiotherapy with
2209–18. carmustine for patients with glioblastoma multiforme: report of
56. Priestman TJ, Bleehen NM, Rampling R, et al. A phase II evaluation Radiation Therapy Oncology Group 93-05 protocol. Int J Radiat
of human lymphoblastoid interferon (Wellferon) in relapsed high Oncol Biol Phys 2004;60:853–60.
grade malignant glioma. Medical Research Council Brain Tumour 73. Stragliotto G, Vega F, Stasiecki P, et al. Multiple infusions of anti-
Working Party. Clin Oncol (R Coll Radiol) 1993;5:165–8. epidermal growth factor receptor (EGFR) monoclonal antibody
57. Quattrocchi KB, Miller CH, Cush S, et al. Pilot study of local (EMD 55,900) in patients with recurrent malignant gliomas. Eur J
autologous tumor infiltrating lymphocytes for the treatment of Cancer 1996;32A:636–40.
recurrent malignant gliomas. J Neurooncol 1999;45:141–57. 74. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus
58. Ramos TC, Figueredo J, Catala M, et al. Treatment of high-grade concomitant and adjuvant temozolomide for glioblastoma. N Engl
glioma patients with the humanized anti-epidermal growth factor J Med 2005;352:987–96.
732 Biological therapy of glioblastoma

75. Stupp R, Hegi ME, Gilbert MR, Chakravarti A. Chemoradiotherapy anti-CD3 stimulated lymphocytes in patients with recurrent grade
in malignant glioma: standard of care and future directions. J Clin III/IV astrocytoma. J Neurooncol 2000;48:113–20.
Oncol 2007;25:4127–36. 87. Yoshida J, Kajita Y, Wakabayashi T, Sugita K. Long-term follow-
76. Tardridge WM, Oldendorf WH, Cancilla P, Frank HJL. Blood- up results of 175 patients with malignant glioma: importance of
brain barrier: interface between internal medicine and the brain. radical tumour resection and postoperative adjuvant therapy with
Ann Intern Med 1986;105:82–95. interferon, ACNU and radiation. Acta Neurochir 1994;127:55–9.
77. Terzis AJ, Niclou SP, Rajcevic U, et al. Cell therapies for glioblas- 88. Yoshida S, Tanaka R, Takai N, Ono K. Local administration of
toma. Expert Opin Biol Ther 2006;6:739–49. autologous lymphokine-activated killer cells and recombinant
78. Von Wild KR, Knocke TH. The effects of local and systemic inter- interleukin-2 in patients with malignant brain tumors. Cancer Res
feron beta (Fiblaferon) on supratentorial malignant cerebral glioma 1988;48:5011–6.
– a phase II study. Neurosurg Rev 1991;14:203–13. 89. Yu JS, Liu G, Ying H et al. Vaccination with tumor lysate-pulsed
79. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Phase II trial dendritic cells elicits antigen-specific, cytotoxic T-cells in patients
of bevacizumab and irinotecan in recurrent malignant glioma. Clin with malignant glioma. Cancer Res 2004;64:4973–9.
Cancer Res 2007;13:1253–9. 90. Yung WK, Castellanos AM, Van Tassel P, et al. A pilot study of
80. Watanabe T, Katayama Y, Yoshino A, et al. Human interferon beta, recombinant interferon beta (IFN-beta ser) in patients with recur-
nimustine hydrochloride, and radiation therapy in the treatment of rent glioma. J Neurooncol 1990;9:29–34.
newly diagnosed malignant astrocytomas. J Neurooncol 2005;72: 91. Yung WK, Kyritsis AP, Gleason MJ, Levin VA. Treatment of
57–62. recurrent malignant gliomas with high-dose 13-cis-retinoic acid.
81. Weller M, Fontana A. The failure of current immunotherapy for Clin Cancer Res 1996;2:1931–5.
malignant glioma. Tumor derived TGF-beta, T-cell apoptosis, and 92. Yung WK, Prados M, LevinVA et al. Intravenous recombinant
the immune privilege of the brain. Brain Res Rev 1995;21: interferon beta in patients with recurrent malignant gliomas: a
128–51. phase I/II study. J Clin Oncol 1991;9:1945–9.
82. Werner-Wasik M, Scott CB, Nelson DF, et al. Final report of a 93. Knerich R, Robustelli della Cuna G, Butti G, et al. Chemotherapy
phase I/II trial of hyperfractionated and accelerated hyperfraction- plus immunotherapy for patients with primary and metastatic brain
ated radiation therapy with carmustine for adults with supratento- tumors. J Neurosurg Sci 1985;29:19–24.
rial malignant gliomas. Radiation Therapy Oncology Group Study 94. Fischer SP, Lindermuth J, Hash C, et al. Levamisole in the treat-
83-02. Cancer 1996;77:1535–43. ment of glioblastoma multiforme. J Surg Oncol 1985;28:214–6.
83. Wersall P, Ohlsson I, Biberfeld P et al. Intratumoral infusion of the 95. Chang SM, Barker FG 2nd, Huhn SL, et al. High dose oral tamox-
monoclonal antibody, mAb 425, against the epidermal-growth- ifen and subcutaneous interferon alpha-2a for recurrent glioma. J
factor receptor in patients with advanced malignant glioma. Cancer Neurooncol 1998;37:169–76.
Immunol Immunother 1997;44:157–64. 96. Kaba SE, Kyritsis AP, Conrad C, et al. The treatment of recurrent
84. Wirtz CR, Albert FK, Schwaderer M, et al. The benefit of neuro- cerebral gliomas with all-trans-retinoic acid (tretinoin). J
navigation for neurosurgery analyzed by its impact on glioblas- Neurooncol. 1997;34:145–51.
toma surgery. Neurol Res 2000;22:354–60. 97. Phuphanich S, Scott C, Fischbach AJ, et al. All-trans-retinoic acid:
85. Wismeth C, Hau P, Fabel K, et al. Maintenance therapy with 13-cis a phase II Radiation Therapy Oncology Group study (RTOG
retinoid acid in high-grade glioma at complete response after first- 91–13) in patients with recurrent malignant astrocytoma. J
line multimodal therapy – a phase-II study. J Neurooncol Neurooncol1997;34:193–200
2004;68:79–86. 98. Lillehei KO, Mitchell DH, Johnson SD, et al. Long-term follow-up
86. Wood GW, Holladay FP, Turner T, et al. A pilot study of autolo- of patients with recurrent malignant gliomas treated with adjuvant
gous cancer cell vaccination and cellular immunotherapy using adoptive immunotherapy. Neurosurg 1991; 28:16–23.
22 Speculations for 2009 and beyond
ROBERT K. OLDHAM

It is now apparent that the genomic capacity of mam- claim a high success rate in terms of new substances that
malian cells to produce biological substances that have have come through the complete testing program with
medicinal value is enormous. Now that a substantial approval for clinical use.
number of lymphokines/cytokines, growth and matura- The process of drug development was historically
tion factors, cellular therapies, and antibody-based based on the idea that sufficient random testing would
approaches have been explored, there is clear evidence identify those active substances that could selectively
of clinical activity with respect to colony-stimulating kill cancer cells. Ancillary was the hope that specific
factors (CSF), blocking factors for epidermal growth processes, unique to cancer cells, would be identified
receptors (EGF) and vascular endothelial growth factor that might allow chemicals to be selectively active on
(VEGF), interferon (IFN), interleukin 2 (IL-2), acti- those cells. A secondary hypothesis that rational drug
vated cells, vaccines, monoclonal antibodies, and their development could be based on structure–function rela-
immunoconjugates. If one analyzes the current level of tionships, receptor information, or analog development
clinical activity for biotherapy in the historical context has proven equally difficult. These concepts have been
of chemotherapy, it is obvious that much more rapid difficult to validate, and the level of selectivity for che-
progress is being made in biotherapy, leading to devel- motherapy is very low. In fact, for every chemothera-
opment of improved, less toxic and more selective forms peutic agent, there are normal tissues that are probably
of treatment. From the earliest days of chemotherapy, more affected by the drug than the cancer in which the
massive searches were done among tens of thousands of drug is active. While there is still hope for the discovery
chemical compounds, searching for the one right drug of some unique chemotherapeutic drug that is selec-
that might have antitumor activity without excessive tively active against cancer, this goal seems much less
toxicity for the patient. The successes were few. From likely to be achieved than it was a decade ago.
analyzing more than a million chemical structures, In contrast, biotherapy and targeted therapy work
less than 80 anticancer drugs have come to the clinic, through physiologic molecules for which the body has
and no more than 30 of these can be considered even receptors and known mechanisms of action. These sub-
moderately active. stances are often used in pharmacological amounts, but
This is not to say that the drug development paradigm they are message molecules known to our cellular sys-
has not had some success. However, the search has been tems. Emerging evidence in laboratory animal model
arduous, complex, expensive, and almost wholly based systems raises the hope that cancer cells are not com-
on the process of random screening of large numbers of pletely without the ability to be controlled by appropri-
compounds to find the rare active chemical. By contrast, ate growth, differentiation, and antiproliferative
the first genetically engineered biological to be approved influences. Most of the data for interferon assumed an
as an anticancer agent, alpha-interferon, has been an antiproliferative activity on cancer cells, in that higher
unqualified success in the clinic. It has been the drug of doses have generally been more effective, and tumors
choice for the treatment of hairy-cell leukemia and resistant to lower doses sometimes respond to an
chronic myelogenous leukemia (until even better tar- increased dose. In addition, the doses active in many
geted therapies were developed) and is moderately cancers are well above the level where interferon is most
active in other forms of leukemia and lymphoma. active as an immunomodulator.
Substantial activity has been reported in renal cancer, As the process of developmental therapeutics for bio-
melanoma, and selected other tumors. therapy is expanded, there is a great need for all those
IL-2 is now approved for renal cancer and melanoma. involved to remain open-minded with respect to new
Monoclonal antibodies and their immunoconjugates paradigms for drug development. Simply to presume that
have proven broadly effective, and the first inhibitors of biologicals must fit into the mold used for chemotherapy
EGF receptor and VEGF have been approved. Thus, drug development has been a major error, since the
biotherapy and small molecule targeted therapy can process of developmental therapeutics for biologics is

R.K. Oldham and R.O. Dillman (eds.), Principles of Cancer Biotherapy, 733
© Springer Science + Business Media B.V. 2009
734 Speculations for 2009 and beyond

very different from that for drugs. Even in the preclini- the control of cellular proliferation. These molecules
cal screening programs now in use for biologicals, the include IL-1-32 and beyond. The future may allow the
process is more one of rational selection than of random use of these components in combination or sequence
screening – the need is to exploit the activities of known to strengthen what are natural but ineffective bodily
molecules rather than to search among the thousands for responses to the problem of cancer growth and metasta-
the singularly active substance. As the information base sis. Although induction of such a cascade by one par-
in cancer biology grows, it will become increasingly ticular substance may not be feasible, the artificial
important to exploit what we know of the power of bio- programming and the medicinal use of these lymphokines
logical molecules rather than to search among unknown are now both feasible and practical. As the process of
chemicals for active substances. administration of biotherapy becomes better defined, so
will the programmability of therapeutic regimens using
these substances. One can envision pre-programmed
infusions of combinations of lymphokines designed for
Beyond Interferon maximal exploitation of their biological activities.
Alpha-interferon is an approved agent for hairy-cell
leukemia, chronic myelogenous leukemia, Kaposi’s
sarcoma, renal cancer and melanoma; it is also active
against a variety of other cancers. Beta- and gamma- Antigen-specific Lymphokines
interferon are both now approved for clinical use. Over and above the use of lymphokines in a more general
Beyond these three interferons, the process of genetic sense, as just described, there is evidence that certain
engineering is bringing forth a huge number of interferon lymphokines act in antigen-specific ways. As the proteins
molecules for testing. Given any small protein of 140– constituting tumor-associated antigens become more
160 amino acids, the possibility now exists for making thoroughly characterized, as a result of monoclonal
an infinite number of molecules simply for altering antibody technology and genetic engineering, one can
portions of the amino acid chain. This process is envision the use of specific antigens with specific
underway for the interferons, and will demonstrate lymphokines, thus allowing oncologists to mount antigen-
whether more active interferons can be developed to specific, and perhaps cancer-specific, immunologic
enhance the immunomodularity, antiviral, or antipro- responses in individual patients. It is likely that these
liferative activity of the natural molecules. antigen-specific lymphokines will have to be used in
ways that are selected specifically for individual patients.
If, as will be reasoned below, the array of tumor-associated
Lymphokines/cytokines antigens present on the tumor cells of each individual is
slightly different from the array present on cancers of a
Interleukin 2 came into the clinic in the mid-1980s by like type in other individuals, the use of antigen-specific
way of a process that used the growth factor to activate lymphokines with specific antigenic preparations will of
lymphocytes that could subsequently be used therapeu- necessity be unique to each individual. Parallels for the
tically as effector cells in patients. The cancer-killing clinic are already being explored in animal models for
capacity of these activated cells was apparent from their transplantation and other specific immune responses.
in-vitro activity, and the major questions related to The application of this technology to human therapeutics
their in-vivo use, distribution, and anticancer activity. will occur in the not-too-distant future.
Interleukin 2 is being used in amounts far in excess of
its normal physiologic role, and toxicities are the natural
result. This particular approach was very expensive; it
required hospitalization and truly enormous doses of Growth and Differentiation
both IL-2 and the activated cells. The future may bring
a better strategy for using this approach in patients,
Factors
whereby repeated treatment may become less toxic and Cancer might be likened to a juvenile delinquent; a
more active than the current intensive regimens. derivative of one’s own self but without the controls
Outpatient regimens that are much less toxic are now necessary for appropriate socialization. If cancer cells
used more frequently. were to have only a limited spread and growth and if
Interleukin 2 is part of a broad cascade of biological subsequent therapeutic approaches could simply limit
molecules that have activities in cellular activation and further growth, there would be no need to eradicate the
Robert K. Oldham 735

cancer cells completely. Such an approach has an obvi- the clinical application of this biotherapeutic approach,
ous parallel in the use of physiologic mediators for it is already apparent that monoclonal antibodies offer
endocrinopathies. For example, diabetes did not disap- the probability of selective cancer treatment that has
pear by virtue of the discovery of insulin. The genetic never before been available [34].
anomaly continues to exist, and diabetes has become a The search for the perfect ‘magic bullet’ is probably
more frequent clinical problem in our society. However, futile. With the exception of anti-idiotypic antibodies,
insulin has been used to control the pathologic manifes- which can be quite specific for the B cell clone that has
tations of this disorder. Is it possible there are growth deviated and become a malignant lymphoma or leuke-
and differentiation factors, coded for by the mammalian mia, it seems unlikely that such singular and specific
genome, that could be used similarly for treating can- antigens are represented on all cancers. Rather, the evi-
cer? There is early evidence of such mediators in vitro dence seems to support a bewildering array of cancer-
and in animal models. The control of growth factors, as associated antigens present in different quantities on
well as agonists and antibodies to those factors, provide cancers from different individuals.
substance to the argument that molecular manipulation Data are accumulating concerning the use of typing
of growth and differentiation is a major area for devel- panels of monoclonal antibodies. They indicate that
opmental therapeutics for cancer. one can assess the antigenic array on each individual’s
If one takes the growth and differentiation of cells in tumor cells and decide which antibodies should com-
vitro as a model, the use of epidermal growth factor, pose a cocktail to cover the available specificities of that
nerve growth factor, insulin, and other hormones has a cancer. This testing mechanism reveals cancer to be
profound effect on both the rate of replication and the individualistic. It is different for every patient and per-
degree of deviance of replicating cells from their ‘nor- haps even among the clones of cancer present within an
mal’ counterparts. Similarly, cancer growth, prolifera- individual patient. This should not be surprising, since
tion, and spread may be controllable by manipulation of the genetic apparatus that governs each of us is indi-
growth factors made by normal tissues and/or by the vidualistic. With the exception of identical twins, no
cancer cells themselves. two humans are exactly alike, and one would be amazed
Similar approaches are now in the clinic as targeted if cancers were exactly alike. Unless a specific genetic
therapy using both monoclonal antibodies and small lesion is identified as the cause of each histological type
molecules interacting with EGF, VEGF and their recep- of cancer, each malignancy in each patient may be an
tors. Rudimentary attempts using certain chemicals that individual problem in biology. It is unlikely that a singu-
cause in-vitro differentiation, have had interesting effects lar change leads to all forms of lung cancer, breast can-
on cell lines and experimental modes; however, the cer, or colon cancer. It is possible that genetic changes
use of retinoids, butyrates, and other factors that might will result in specific markers for each of those cancers,
regulate growth and differentiation have had minimal which can be exploited in treatment. However, it is per-
impact in the clinic. With almost daily discovery of sub- fectly obvious that the cells in which those changes
stances that support growth of cancer cells in vitro, one occur are genetically distinct from one person to another.
might imagine that the antithesis of such factors might Many genetic alterations, caused by the same chemical
also be developed, exploited, and utilized for clinical or physical carcinogen, result in the outgrowth of cells
therapeutics. In biology, as in physics, there seems to that markedly differ from each other. If such is the case,
be a reaction for every action. For many of the mole- one might postulate that cancer in each patient is the
cules that support growth, experimental data also indi- individualistic expression of a response to that multi-
cates that other molecules exist to offset that increased step, complex carcinogenic impulse, thereby embody-
growth. As these molecules are isolated and charac- ing the genetic characteristics of both the host and the
terized, and subsequently produced by genetic engineer- carcinogenic influence. If such is the case, then the
ing, they will broaden our potential arsenal for cancer actual problem in developmental therapeutics will be
biotherapy. individualistic and each patient will have to be analyzed
as an individual problem in cancer biology.
This concept of individuality of cancers has been diffi-
cult for many cancer biologists and clinicians to accept.
Monoclonal Antibodies This concept was first present in the 1st Edition of this text
The current evidence to support the therapeutic use of book in 1987 and is still being debated. Recently, it has
monoclonal antibodies and their immunoconjugates has been “rediscovered” as “Personalized Medicine”. We have
been presented in earlier chapters. While it is early in learned and use the histological classification systems
736 Speculations for 2009 and beyond

embedded in the minds of pathologists and transmitted the best minds of science rather than proceeding in an
through textbooks of medicine. These concepts classify overly structured and rigidified way, new paradigms for
cancers categorically according to tissue of origin and his- developmental therapeutics may allow for greater strides
tological features. In spite of the clinical and laboratory to be made using biotherapy.
observation that phenotypic analysis and even ‘genotype’ End-point reductions in cancer death rates are difficult
in cancer biology confer great diversity within cancers of to achieve, or even to detect, because of time factors. To
the same histological type, we continue to develop thera- know that the death rate from breast cancer has been
peutics as if all breast cancer, all lung cancer, and all colon reduced by 50%, one would have to have at least a 5-year
cancer are replicates. This is the simplest to teach, the sim- follow up of the patients at risk since the recurrence rate,
plest to learn, and the simplest to practice. However, it is although highest in the first 2 years, continues to be sig-
fundamentally and biologically incorrect. nificant for several years after diagnosis. For lung cancer,
Heretofore, there has never been a technology that the problem is somewhat more straightforward, in that
allows cancer biologists to understand cancer on an end point of an effective change in therapeutics can be
individualistic basis. Monoclonal antibody technology, discerned within 3–5 years. Breast, colon, and lung can-
proteonomics, and genomics represent what may be the cers being the most common malignancies, the reduction
solutions to the problem. If cancer is indeed diverse of death rate by 50% in these three tumors by the year
and individualistic, might not the immune system, 2000 would have required that the actual change would
itself individualistic and diverse, be able to solve the have to have occurred between 1990 and 1995. That sim-
problem? Each person’s immune system has the ability ply did not occur. Although biotherapy is likely to play
to produce several hundred thousand to several million an increasingly important role within the next decade, it
different antibody molecules. Taken together, the diver- is doubtful that changes of such magnitude can be pro-
sity of the immune system from the broader perspective duced in the near term. Rather, it seems more likely that
of a population of individuals is gigantic. By the use of the first 2 decades of the 2000s will be those in which the
in-vitro techniques and the cellular diversity inherent in early phases of such changes may occur, and the actual
the immune system, it is probable that the response end point for significant death-rate reduction will then
capabilities in antibody technology exceed the diversity fall well after the year 2020.
implicit to cancer. It may well be postulated that each It is 2009. Interferon, interleukin 1–32, growth and
cancer in each patient presents with an individual array maturation factors, tumor necrosis factor, colony stimulat-
of antigens. If such is the case, it might be possible to ing factors, activated cells, monoclonal antibodies, immu-
generate antibodies and/or type tumors with a multipha- noconjugates, vaccines, and gene therapy are clinical
sic approach, leading to the generation of cocktails of realities – but they are all in an early phase of developmen-
antibody to respond to the diversity inherent in cancer tal therapeutics. If developed as drugs have been tested in
biology. Clearly, T cells could be generated and manipu- the past, the average time to take these from the laboratory
lated in a similar manner for specific cellular therapy. to an approved therapeutic is 10 years, at a cost of over
$800 million each. If we are to move forward in develop-
mental therapeutics for biologicals, we cannot take 10
years to develop each approach individually. Certainly, it
Cancer Treatment: The Future is impossible to imagine a series of events that will allow
It has been categorically stated by various authorities an end-point reduction of 50% by the year 2020 or even
that cancer is a problem that should be largely solved or 2050 if each biopharmaceutical takes 10 years to be
under control within 10–20 years. The National Cancer brought from concept to widespread clinical use.
Institute set a national priority of reducing the cancer
death rate by at least 50% by the year 2000. These
projections were certainly optimistic, and they were not
achieved. On the horizon for surgery, radiotherapy, and Biotherapy is not Chemotherapy
chemotherapy, there seems little to support the goal of
reducing the cancer death rate by 50% by any future
and it not just Immunotherapy
date. Although it is doubtful that the cancer death rate The fourth modality of cancer treatment is constituted
can easily be reduced by this magnitude, it is likely that more broadly to include all of the factors described in
the approaches available through biotherapy will play a this book [3]. To take maximum advantage of the oppor-
major role in developmental therapeutics in the next tunities available through biotherapy, major structural
decade. If the field is allowed to progress by using changes are necessary in our system of translation of
Robert K. Oldham 737

developmental therapeutics from concept to the labora- funding for clinical research is becoming less available.
tory and then to the clinic [2]. We cannot afford to Much of the funding was derived from insurance
develop biologicals in the protracted, expensive unidi- reimbursement for clinical trials, and with the current
mensional manner of drug development. We have a pressure on insurers to reduce costs, at the behest of
huge number of new biological substances, and the cur- employers and patients who pay for their insurance
rent system of access and opportunity for patients, the premiums, it is becoming increasingly difficult to fund
system of funding of research, our method of govern- clinical research through third-party reimbursement.
ment regulation, and our reimbursement system for Thus, while the regulation and the intrinsic cost were
developmental therapeutics must undergo major change the major impediments to rapid drug development prior
[4–11, 15, 18, 19, 21–27, 29–33]. We are now faced to 2000, reimbursement for clinical trials and even
with the reality of many more opportunities for effective reimbursement for the ‘off-label’ use of new forms of
cancer therapy than mechanisms by which these oppor- therapy will become the major limiting factor in the
tunities can be brought to clinical reality. future. We are now faced with the probability that cancer
For more than 2 decades, cancer research and cures are being developed that will not be reimbursed
treatment have operated on the ‘kill and cure’ hypothesis. by government or private insurance programs [13, 16,
Developmental therapeutic programs have functioned 17, 20].
under a format where a new drug is brought to the clinic With our current reimbursement system, autologous
test in phase I for toxicity and phase II for activity with bone marrow transplantation for certain forms of
the presumption that short-term effects on cancer cancer has reached a level of cure of 20% of patients
(response rates) will ultimately lead, if positive, to with selected neoplasms at a cost of $100,000–150,000
survival benefit. While this paradigm has been useful in per procedure. Thus, the upper limit of the price for a
developing cytotoxic drugs, there is much to suggest human life is becoming more clear. New technology
that we should now broaden our concept of developmental tends to be expensive early and becomes less expensive
therapeutics, as cancer biotherapy comes to the fore, to later. However, we now face the probability that
include the idea of cancer control. Much as one treats certain new technologies will be available and highly
diabetes with insulin, it may soon be possible, through effective while still very expensive [20]. Society must
the use of growth and maturation factors and other forms decide how much life is worth and what percentage of
of cancer biotherapy, to arrest the growth and spread of the gross national product should be allocated to
cancer and thus make these diseases more amenable to healthcare. This is an area that needs public debate
long-term control even in the absence of tumor and should not be simply left to ‘social planners and
eradication. The implications in terms of cost and thought leaders’.
reimbursement for clinical services are obvious. We Therefore, it is apparent that cancer biotherapy brings
cannot afford to pay for developmental therapeutics as cancer research and developmental therapeutics to a
we have in the past. How is society going to pay for the crossroads. In this millennium, we will not be limited by
very long-term clinical trials, perhaps constituted over a our ability to conceptualize and develop highly effective
3–5 year period, to test the various hypotheses for cancer treatment. We will be limited by broader social consid-
control? Witness the difficulty in establishing clinical erations of who shall pay for such approaches and how
trials for cancer prevention and the high cost intrinsic much each life is worth [20, 28].
to these programs as examples of the difficulty in Thus, it is apparently that the many opportunities bio-
envisioning clinical research for long-term cancer therapy brings to scientists and clinicians will have to be
control projects. Obviously, this is going to require a moved at a much faster rate from laboratory to clinic if
major restructuring of developmental therapeutics if our patients are to derive benefit from these new
society is to take advantage of these opportunities [8–10, approaches. Perhaps the use of the laboratory in con-
12, 13, 34]. junction with the clinical practice of medicine, as sug-
For more than 3 decades, the major factors slowing gested by the Nobel Laureate, Dr. Peter Medawar, is the
developmental therapeutics have been the regulations true ‘breakthrough’ in the developmental therapeutics
promulgated by Congress and the Food and Drug of cancer [1]:
Administration (FDA) for the development of new “The cure of cancer is never going to be found. It is
drugs [14]. These regulations have resulted in a system far more likely that each tumor in each patient is going
of drug development costing more than $800 million to present a unique research problem for which labora-
per new drug taken through to commercial availability. tory workers and clinicians between them will have to
In the last 10 years, it has become apparent that the work out a unique solution.”
738 Speculations for 2009 and beyond

References 18. Oldham RK, Avent RA. Clinical research: who pays the bill?
Oncol Issues 1989;4:13–14.
1. Medawar P, ed. The Life Sciences. New York: Harper & Row, 1977. 19. Oldham RK. Clinical research in cancer: a time for consensus.
2. Oldham, RK. Biologicals: new horizons in pharmaceutical Mol Biother 1989;1:242–243.
development. J Biol Response Modif 1983; 2:199–206. 20. Oldham RK. Cancer cures. By the people, for the people, at what
3. Oldham RK. Biologicals and biological response modifiers: the cost? (editorial) Mol Biother 1990;2:2–3.
fourth modality of cancer treatment. Cancer Treat Rep 1984;68: 21. Oldham RK. Cancer and diabetes: are there similarities? Mol
221–232. Biother 1990;2:130–131.
4. Oldham RK. The cure for cancer. J Biol Response Modif 22. Oldham RK. Whose rights come first? Mol Biother 1991;3:
1985;4:111–116. 58–59.
5. Oldham RK. Whose rights come first? J Biol Response Modif 23. Oldham RK. Cancer research: a pubic trust. BioPharm 1991;4:
1985;4:211–212. 8–9.
6. Oldham R`K. The government-academic ‘industrial’ complex. J Biol 24. Oldham RK. Is informed consent a function of who pays? Mol
Response Modif 1986;5:109–111. Biother 1991:3:2–5.
7. Oldham RK. Profits: who benefits? J Biol Response Modif 25. Oldham RK The rights and wrongs of national heath care. Pharm
1986;5:203–205. Exec 1991;11:92–93.
8. Oldham RK. Patient-funded cancer research. N Engl J Med 26. Oldham RK. Cancer research for whom? BioTechnology 1991;
1987;316:46–47. 9:772.
9. Oldham RK. Drug development: who foots the bill? Biotechnology 27. Oldham RK. Peer review. Mol Biother 1992;4:2–3.
1987;5:648. 28. Oldham RK. BioEthics: Opportunities, Risks and Ethics: The Privati-
10. Oldham RK. False hope vs. opportunity. Cope 1987; April:66. zation of Cancer Research. Franklin, TN: Media American, 1992.
11. Oldham RK. Whose life is it anyway? Wall Street J 1987;April 24. 29. Oldham RK. Fundamentally flawed. Cancer Biother 1993;8:
12. Oldham RK. Letter to the Editor: patient-funded cancer research. 111–114.
N Engl J Med 1987;316:172. 30. Oldham RK. What’s the score? Cancer Biother 1993;8:187–188.
13. Oldham RK. Who pays for new drugs? Nature 1988;332:795. 31. Oldham RK. Cancer research: does it deliver for the patient?
14. Oldham RK. Regularity hierarchies (editorial). Mol Biother Cancer Biother 1994;9:99–102.
1988;1:3–6. 32. Oldham RK. The war on cancer: new battle plan needed. Cancer
15. Oldham RK. Fundamentally flawed? Mol Biother 1988;1:58–60. Biother 1994;9:289–290.
16. Oldham RK. Why deny health care? The Freeman 1989;March: 33. Oldham RK, Dillman RO. Gold standard or wrong standard (edi-
94–95. torial). Cancer Biother Radiopharm 2004;19(3):271–272.
17. Oldham RK. Clinical research in cancer: a time for consensus. 34. Oldham RK, Dillman RO. Monoclonal antibodies in cancer therapy:
Pharm Exec 1989;July:23–34. twenty-five years of progress. JCO 26(11):1774–1777.
Index

1,25-(OH)2D3 544 autologous bone-marrow transplantation (ABMT) 498


4-1BB ligand 199 autologous tumor cells 507, 511, 585
azocytidine 548
activation-induced cell death (AICD) 169, 204, 276
active specific immunotherapy 5 B cells 279, 333
acute leukemia 584 cytokines 200, 208, 211, 212
acute lymphocytic leukemia (ALL) 366 interleukins 194
acute myelocytic leukemia (ALM) 366 lymphocytes 85, 88
acute myeloid leukemia 548 Bacillus Calmete Guerin (BCG) 669, 679, 713
adeno-associated viruses (AAV) 589–591 bacterial toxins 409
adenoviral vectors 591 bestatin 126, 128
adjuvant chemotherapy 128 beta particles 464
adoptive cellular therapy 652, 684 bifunctional antibodies 307
AIDS/HIV, 118, 576 biological heterogeneity 25, 23, 33
alemtuzumab 340 biological response modifiers (BRM) 1, 41, 101, 364, 484
alkylating agents 112 biological 2, 5, 41, 117, 121, 123, 125, 338, 342, 349, 613,
all-trans retinoic acid (ATRA) 527, 528 616, 660, 671, 681, 700, 715, 724
allergic reactions 321 bispecific antibodies 307
allogeneic bone-marrow transplantation (ALBMT) 497, 500 blood transfusions 109
allogeneic stem cells 652 bone cells 207
allogeneic tumor cells 512 bone marrow 497, 575, 576, 583
alpha interferon 465 breast cancer 344, 347, 348, 473, 669
alpha particles 647, 660, 681, 699, 715, 724
amifostine 685 camptothecins 596
anaphylactoid reactions 320 cancer vaccines see vaccines
angiogenesis 18, 28, 29, 179, 191, 203, 216, 599 carbohydrates 148, 150
animal models 305 CD20, 325, 366
anthracyclines 596 CD27 ligand 200
anthrax 408 CD30 ligand 200
anti-angiogenesis 292 CD40 ligand 201, 369
antibiotics 112 CD52, 340
antibodies 12, 67, 68, 71, 303, 305–312, 315, 338, 363, 377, CEA gene expression 472
640, 661, 672, 686, 696, 701, 702, 711, 735 cell cycle gene therapy 594
see also monoclonal antibodies cell functions 95
anti-idiotype 363 chemoattractants 203
current trends 640, 711, 728, 735 chemokines 203
heterogeneity 451 chemosensitization 603
radiolabelled 375 chemotactic cytokines 179
antibody-dependent cell-mediated cytotoxicity (ADCC) 312 chemotherapy 29, 113, 126
anti-CD20 antibodies 469, 497 differentiation 547
antigen-specific lymphokines 734 ex-vivo purging 498
antigenic modulation 322 immunoconjugates 455
antigens 306, 363, 364, 372, 373, 380, 381, 509, 669 immunosuppression 111, 127, 128, 130
anti-idiotype antibodies 363 interferions 285
anti-idiotype vaccines 314 monoclonal antibodies 332, 333
antimetabolites 111, 596 renal cancer 645
antiproliferative action 290 chemotherapy-induced immunosuppression 111
antisera 303, 305 chemotherapy-induced neutropenia 574, 583
antitumor cocktails 453 chimeric monoclonal antibodies 325
antiviral activity 290 chimeric proteins 71
apoptosis 200, 204, 207, 213, 216, 371 chronic lymphocytic leukemia (CLL) 122, 329, 333, 702

739
740 Index

chronic myeloid leukemia (VML) 284 gene-directed enzyme-prodrug therapy (GDEPT) 595
cimetidine 128 genetic immunopotentiation 595
clinical remission 114 genetically modified tumor cells 654
clinical trials 41, 326, 340, 344, 351, 357, 361, 457, 623 genitourinary cancer 645
antisera 305 genomics 53, 65
immunoconjugates 455, 457 glioma 474
immunotoxins 426, 427 graft-versus-host disease (GVHD) 497
monoclonal antibodies 326, 340, 344, 351, 357, 361 graft-versus-tumor (GVT) effects 500
cloning 53 granulocyte-colony stimulating factor (G-CSF) 569
colon cancer 122, 659 granulocyte-macrophage colony-stimulating factor
colony-stimulating factors (CSF) 62, 551, 569, 581 (GM-CSF) 581
colorectal cancer 351, 357, 359 growth factors 10, 527, 549, 555, 734
combination chemotherapy 113
combined-modality studies 126 hair-cell leukemia (HCL) 279, 329, 693
complement-mediated cytotoxicity (CMC) 312 head cancer 123, 358, 359, 713
conjugates 67 helper T cells see T-helper cells
copper-64, 465 hematopoiesis 165, 174, 183, 191, 544
Corynebacterium parvum 679, 714 interleukins 165, 174, 183, 191
cytokines 8, 57, 155, 549, 734 malignancies 107, 109, 114
see also individual cytkines HER-2, 68, 343, 373, 430, 453, 646, 573
current trends 647, 651, 734 herpes simplkex virus (HSV) 591
cytolytic functions 106 histamine receptor antagonists 116
cytosine arabinoside 548 historical perspective 1
cytotoxicity 95, 312 HIV/AIDS, 118, 576
Hodgkin’s disease 120, 330
daclizumab 367 hormones 113, 680
delayed allergic reactions 321 host-tumor interactions 68
delayed-type hypersensitivity (DTH) 93, 105 human anti-immunoglobulin response (HAMA/HACA/
delivery see drug delivery systems HAHA) 321
dendritic cells 165, 204, 513 human monoclonal antibodies 309
differentiation 174, 292, 527, 734 humanized monoclonal antibodies 310
dimethyl sulfoxide (DMSO) 527 humoral immunity 94
dose response 324 hybridoma technology 307
drug delivery systems 74
drug development 42, 625 idiotype network 313
immune system 1, 85, 429, 505, 633, 645
effector cells 102, 595 colony-stimulating factors 551
embryonic development 206 complexes 321
empirical clinical testing 46 cytokines 180
endothelia 213 immunoselection 323
enzyme-prodrug systems 596, 605 immunotoxins 69, 407, 426, 427, 698, 701
eosinophils 174 interferons 291
epidermal growth factor receptor 314, 356, 372, 373 potentiation 595
escalating-dose trials 46 stimulation 183, 669
ex-vivo purging 498 suppression 101
expression of genes 25, 27, 53 immunity 85, 94, 183, 194, 195, 515
immunoconjugates 451, 452, 455
Fas ligand 204 immunoglobulin 95, 321
febrile neutropenia 332, 347, 571 immunosuppression 101
FISP, 196, 276 chemotherapy-induced 111
Flt-3 ligand 171, 177, 178, 205, 206 radiation therapy-induced 110
free antigen 323 in vitro assays 94, 96
fungal toxins 415 in vivo assays 93
in vivo binding 315
gamma interferon 684, 715, 725 inflammation
gastrointestinal cancer 472 cytokines 201, 208, 213, 216
gene expression 25, 27 interleukins 165, 174, 183, 191
gene therapy 589, 594, 604, 639 infusion reactions 318
Index 741

interferons 9, 45, 57, 197, 277, 649, 660, 715 LIGHT, 207, 161
alpha 647, 660, 681 lipopolysaccharide (LPS) 163
current trends 671 lung cancer 122, 123, 679, 684
gamma 161, 191, 684, 715, 725 lutetium-177, 464, 465
interleukin-1 (IL-1) 60, 156, 165, 552 lym-1 antibody 466, 467
interleukin-2 (IL-2) 61, 156, 169, 637, 638, 649, 661, 671, lymph nodes 209
684, 695, 701, 715 lymphocytes 88, 91, 498, 652
interleukin-3 (IL-3) 61, 171 lymphoid organogenesis 207, 208
interleukin-4 (IL-4) 61, 172, 156, 685 lymphokine-activated (LAK) cells 93, 505, 652
interleukin-5 (IL-5) 157, 174 lymphokines 8, 95, 734
interleukin-6 (IL-6) 157, 174, 552 lymphoma 180, 281, 326
interleukin-7 (IL-7) 157, 177 lymphoproliferative responses (LPR) 94, 105
interleukin-8 (IL-8) 157, 179 lymphotoxins 207, 208, 554
interleukin-9 (IL-9) 157, 180
interleukin-10 (IL-10) 157, 180 macrophages 92, 177
interleukin-11 (IL-11) 158, 182 malignant melanoma 121, 123, 633
interleukin-12 (IL-12) 158,183 mast cells 174, 180, 212
interleukin-13 (IL-13) 158, 185 maturation factors 5, 10
interleukin-14 (IL-14) 158, 186 melanoma 121, 123, 380, 381, 385, 633
interleukin-15 (IL-15) 158, 186 metastasis 17, 20–22, 24, 32
interleukin-16 (IL-16) 158, 187 migration 203
interleukin-17A (IL-17A) 158, 189 ML-1 191, 555, 556
interleukin-17B (IL-17B) 159, 190 molecular chemotherapy 589, 590, 595, 599
interleukin-17C (IL-17C) 159, 191 monoclonal antibodies 12, 71, 303, 305, 307, 309–311, 315,
interleukin-17E (IL-17E) 159, 191 338, 378, 661, 686, 696, 701, 702, 735
interleukin-17F (IL-17F) 159, 191 monocytes 92
interleukin-18 (IL-18) 159, 191 multidrug resistance 32
interleukin-19 (IL-19) 159, 193 multiple myeloma 280, 329, 711
interleukin-20 (IL-20) 159, 194 mutation compensation 593
interleukin-21 (IL-21) 159, 194 myelodysplastic syndrome (MDS) 548
interleukin-22 (IL-22) 159, 195
interleukin-23 (IL-23) 159, 195 natural killer (NK) cells 92, 169, 194, 208
interleukin-24 (IL-24) 159, 196 neck cancer 123, 358
interleukin-24 (IL-25) 160, 196 neutropenia 574, 576, 583
interleukin-26(IL-26) 160, 197 neutrophils 189
interleukin-27 (IL-27) 160, 197 nitric oxide synthetase-inhibiting cytokine 210
interleukin-28A (IL-28A) 160, 197 non-hematological tumors 472
interleukin-28B (IL-28B) 160, 197 non-Hodgkin’s lymphoma (NHL) 281
interleukin-29 (IL-29) 160, 197 non-small cell lung cancer (NSCLC) 684
interleukin-31(IL-31) 160, 198 nonspecific immunomodulators 1, 5
interleukin-32 (IL-32) 160, 198 nonsteriodal anti-inflammatory and antipyretic drugs
interleukin-33 (IL-33) 160, 198 (NSAIDs) 117
interleukin-35 (IL-35) 160, 199 non-viral vectors 590, 592
interleukins 8, 60, 62, 119, 715 novel neurotophin 209
iodine-131, 464, 483 NPT 15392, 117
isolating genes 53
OKT-432, 126, 129, 680, 714
Isoprinosine 124 oncogenes 594
oncostatin M, 209
Kaposi’s sarcoma 286 osteoblasts 213
kidney see renal cancer osteoclasts 212
osteopontin 210
lentinan 128 ovarian cancer 355
leukemia 284, 329, 333, 470, 471, 487, 532, 540, 548, 551, OX40 ligand 211
584, 693, 702
leukemia inhibitory factor (LIF) 206, 553 pancreatic cancer 355
levamisole 115, 123, 126, 129, 659, 670, 680, 713 pathogenesis 17
ligand conjugation 416 patient selection 48
742 Index

PEG interferon 649 stomach cancer 310


peptide cytotoxins 407 surgical adjuvant studies 122
peptides 476, 477, 479
perioperative immunosuppression 109 T cells 638
phagocytes 95 cytokines 200, 206, 209, 211
plant toxins 407 interleukins 169, 174, 177, 180, 186, 194, 195
podophyllotoxins 113 lymphocytes 88, 147
polar-planar compounds 528, 546 lymphoma 699
preclinical models 4, 426 reconstituting factor 118, 125
pro-apoptotic gene therapy 589 T-cytotoxic factor 91
proallergic cytokines 185 T-helper cells 90, 172, 209
prostaglandin antagonists 124 T-suppressor cells 90
prostate cancer 32, 355, 380, 472 technetium-99m 478
proteomics 65 thrombocytosis 182
thrombopoiesis 174
radiation therapy-induced immunosuppression 110 thymic factors 7, 117, 125, 128, 129
radioimmunotherapy (RIT) 71, 463, 466, 469 thymic hormones 680, 714
radiolabelled antibodies 463 toll-like receptors 163
radiosensitization 603 TRAIL, 216
RANKL, 212 TRANCE, 212
recombinant DNA, 7, 53, 571 transductal targeting 592
recruitment 203 transfer factor 119, 125, 129
regulatory approaches 313 transforming growth factor β 555
remission 114 transcriptional targeting 592
renal cancer 288, 353, 380, 645 trastuzumab 343, 344–346, 348, 349, 672
replicative vector oncolysis 601 tumor cell burden 105
reticuloendothelial system 94 tumor infiltrating lymphocytes (TIL) 505
retinoids 119, 125, 672, 682, 701, 716, 725 tumor lysis syndrome 319
retroviral vectors 590 tumor necrosis factors (TNF) 63, 165, 207, 213,
rhenium-186/188, 464 371, 554
rituximab 325–333, 337, 38, 616 type I toxins 415,
type II toxins 415
sarcomas 356 type III toxins 415
screening 4 unconjugated monoclonal antibodies 303
serum sickness 321 urologic cancer 122
severe chronic neutropenia 576 vaccines. 71, 147, 374, 385, 510, 513, 634, 653, 664, 675,
sickle-cell anemia 577 687, 698, 701, 718
skin development 194 current trends 634, 653, 664, 675
small cell lung cancer (SCLC) 684 vertebrate toxins 414
solid tumors 105, 108, 114, 121 vinca alkaloids 113
soluble cytokine receptors 64 viral vectors 589
stem cell factor (SCF) 162, 212 vitamin A analogs 532
stem cells vitamin D analogs 540, 543, 544
bone-marrow transplantation 497, 500, 575
colony-stimulating factors 569 wound repair 165, 209, 468
cytokines 204
radioimmunotherapy 469 Y2B8 antibody 468
renal cancer 645 yttrium90, 464–477, 481–485, 487, 488

Вам также может понравиться