Вы находитесь на странице: 1из 8

Available online at www.sciencedirect.

com

Minicells: Versatile vectors for targeted drug or si/shRNA cancer


therapy
Jennifer A MacDiarmid and Himanshu Brahmbhatt

Effective cancer therapy continues to be a daunting challenge [2], nanoparticles [3,4], and polymerdrug conjugates
due mainly to considerable tumor cell heterogeneity, drug- [5]. The very large number of different nanocarriers being
resistance, and dose-limiting toxicity of therapeutics. Here we currently developed, several of which are FDA approved,
review a versatile nano-cellular (minicell) delivery vehicle that in clinical trials and marketed is reviewed [6]. However,
can be packaged with therapeutically effective concentrations these technologies are also hampered by shortcomings,
of chemotherapeutic drugs, siRNAs or shRNAs and can be such as drug leakage in vivo, lack of versatility in terms of
targeted to tumors via minicell-surface attached bispecific packaging a diverse range of different drugs without
antibodies. A range of minicell-based therapeutics have shown significant derivatization, thereby reducing drug potency,
highly effective tumor stabilization/regression in the murine and difficulties in production scale-up, particularly for
xenograft model and in case studies in canines with late-stage nanoparticles.
endogenous tumors. Repeat intravenous dosing shows
absence of toxicity or immunogenicity in both species. The Anti-cancer antibodies directed to over-expressed recep-
minicell-based therapeutic has potential applications in tors on cancer cells, such as the epidermal growth factor
personalized cancer medicine. receptor (EGFR; [7,8]) and HER2/neu [9] are less toxic,
but do not possess the potency and wide spectrum anti-
Address tumor activity of chemotherapeutic drugs. Additionally,
EnGeneIC Ltd, Cancer Therapeutics, Building 2, 25 Sirius Road, Lane some for example HER2/neu are only over-expressed in
Cove West, Sydney, NSW 2066, Australia 25% of breast cancer resulting in HER2/neu negative
Corresponding author: Brahmbhatt, Himanshu
breast cancer patients, meaning that the majority of
(hbrahmbhatt@engeneic.com) patients do not benefit from antibody therapy. The high
rate of mutations in the tumors also results in receptor
mutations [10] and consequent development of resist-
Current Opinion in Biotechnology 2011, 22:909916 ance to antibody therapeutics.
This review comes from a themed issue on
Pharmaceutical biotechnology In recent times, RNA interference (RNAi) has emerged
Edited by Luis Angel Fernandez and Serge Muyldermans as a powerful technology for the development of cancer
therapeutics. RNAi is a natural phenomenon resulting in
Available online 6th May 2011
potent post-transcriptional gene silencing produced by
0958-1669/$ see front matter double-strand RNAs that occur in most eukaryotes [11].
# 2011 Elsevier Ltd. All rights reserved. This multi-step process is initiated by double stranded
DOI 10.1016/j.copbio.2011.04.008
2030 nucleotide small interfering RNAs (siRNAs) or
microRNAs (miRNAs) resulting in highly efficient and
sequence-specific knockdown of the targeted genes
expression. siRNAs can also be expressed from plasmid
Introduction DNAs as short hairpin RNAs (shRNA) using an RNA
Cancer remains the major cause of death in most polymerase III promoter. RNAi has considerable poten-
advanced countries and the incidence of cancer increases tial in the treatment of cancer [12] with several ongoing
as populations age. Despite considerable advances in clinical trials. Nevertheless, several challenges need to be
development of cancer therapeutics, the disease con- overcome for exogenous siRNA to be widely used as a
tinues to be plagued with a highly significant mortality cancer therapeutic. These include: firstly, lability of siR-
rate. NAs, resulting in rapid degradation by serum nucleases,
secondly, poor membrane permeability of siRNAs limit-
Chemotherapeutic drugs lack cancer-cell selectivity and ing cellular uptake, thirdly, need for effective design of
indiscriminate drug distribution results in severe toxicity active siRNAs to ensure optimal gene silencing activity
and limits anti-tumor efficacy. Owing to the intrinsic with minimal off-target effects, and fourthly, need to
genetic diversity and the ability to rapidly mutate, tumors achieve efficient intracellular delivery to target cells in
develop multi-drug resistance which contributes to the vivo [13,14]. Several promising strategies have been
high rate of treatment failure. To overcome these draw- developed for systemic siRNA delivery such as nanopar-
backs, efforts are being made to develop targeted drug ticles [15], aptamer-siRNA conjugates [16], nanoimmu-
delivery systems such as stealth liposomes [1], micelles noliposomes [17], cationic polymer and lipid-based

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:909916


910 Pharmaceutical biotechnology

siRNA complexes [18] etc., but several hurdles, including models, as well as in case studies of dogs with endogenous
delivery, still need to be overcome. tumors.

In this article we review a versatile tumor-targeted, nano- Packaging chemotherapeutic drugs or siRNAs
cellular carrier, a bacterially derived minicell, capable of or shRNAs into minicells and tumor targeting
carrying therapeutically significant concentrations of via attachment of bispecific antibodies
drugs or siRNAs or shRNAs and can be targeted to tumors Minicells are derived from a minCDE-chromosomal
via attachment of bispecific antibodies (BsAbs) to the deletion mutant of Salmonella enterica serovar Typhimur-
minicell-surface O-polysaccharide. ium (S. Typhimurium) and purified as described pre-
viously [23].
Bacterially derived minicells were first observed,
described, and termed minicells by Howard Adler A range of chemotherapeutic and molecularly targeted
and colleagues in 1967 [19]. They are anucleate, non- drugs with differing structure, charge, hydrophobicity,
living nano-sized cells (400 nm in diameter) and are and solubility such as doxorubicin, paclitaxel, irinotecan,
produced as a result of mutations in genes that control 5-fluorouracil, cisplatin, carboplatin, gemcitabine, vin-
normal bacterial cell division [20,21,22] thereby de- blastine, and monastrol, could be readily packaged within
repressing polar sites of cell fission. the minicells ([23,24]; schematic shown in Figure 1).
This was accomplished by co-incubating each drug with
Previously we had reported that minicells can be pack- intact minicells for a period of time that was optimized for
aged with therapeutically significant concentrations of a each drug [23].
range of chemotherapeutics [23] or siRNAs and
shRNAs [24]. These minicells selectively targeted to Drug-packaging in minicells was shown to be dependent
cancer cells via BsAbs, effect highly significant tumor on both the concentration of drug in the loading solution,
stabilization/regression in a variety of tumor xenograft and time of incubation [23]. Drug permeation into

Figure 1

Empty minicells Insertion of anti-cancer drug

OR

siRNAs

Attachment of
bispecific
antibody

Attachment to
cancer cell
surface receptor

Current Opinion in Biotechnology

Schematic showing bispecific antibody-targeted, drug/siRNA-packaged minicells. Schematic showing the packaging of anti-cancer drugs or siRNAs
into empty minicells and targeting them to tumor cell-surface receptor using bispecific antibodies where one arm of the antibody has minicell-surface
O-polysaccharide specificity and the other arm has specificity for the tumor cell-surface receptor for example EGFR.

Current Opinion in Biotechnology 2011, 22:909916 www.sciencedirect.com


Minicells as versatile vectors for targeted delivery MacDiarmid and Brahmbhatt 911

minicells possibly occurs via non-specific porin channels as liposomes have been shown to package 10,000 mol-
[25] in the outer membrane. High resolution structural ecules of drug per liposome [32]. Similarly, armed anti-
analysis of some bacterial porins suggests that permeation bodies can conjugate only less than 10 drug molecules per
of chemicals through the porin channel is driven by antibody. The potency of anti-tumor effects may depend
molecular interactions with the surface rather than by on the concentration of a drug that is delivered intra-
free diffusion [26,27]. Detailed studies of porins have cellularly within cancer cells.
revealed charged residues within the channels resulting
in a transversal electric field that separates polar and non- siRNAs are also packaged in minicells through co-incu-
polar solutes. Polar solutes are thought to be oriented in bation ([24]; schematic shown in Figure 1). shRNA
the field during permeation which therefore becomes a encoding plasmids are initially transformed into the min-
rapid one-dimensional entry process [28]. Nonspecific CDE-S. Typhimurium strain and the plasmid segregates
movement of hydrophobic solutes across the outer mem- into the minicells. When the minicells are purified, they
brane possibly occurs through other channels such as the are therefore pre-packaged with therapeutically signifi-
FadL family of outer membrane proteins [29,30] and cant copies of the plasmid. PCR quantitation studies
OmpW [31]. Thus it is likely that drug entry into mini- showed that 50 copies of plasmid or 14,000 copies
cells is through a facilitated entry process. Leakage of the of siRNA are packaged per minicell.
drug from the minicells was not observed following
incubation in buffer or serum for over 24 hours. Targeting of minicells to tumor cells was achieved using
bispecific antibodies in which one arm recognizes the O-
Quantitation of drug-packaged in minicells showed that polysaccharide component of the minicell surface lipo-
approximately a million molecules of doxorubicin were polysaccharide (LPS) and the other, a cell-surface re-
loaded per minicell. In contrast, other nanoparticles such ceptor specific for the mammalian cell to be targeted,

Figure 2

Transport of antibody-targeted drug minicell attachment to tumor


or siRNA-packaged minicells cell surface (active targeting)

minicells fall out


of leaky vasculature
(passive targeting)

Current Opinion in Biotechnology

Schematic showing passive and active targeting to tumors in vivo. Schematic showing that post-i.v. administration of the minicell-based anti-cancer
therapeutic, the minicells accumulate in the tumor microenvironment due likely to the EPR effect (passive targeting) which is then followed by the
process of active targeting via attachment of the minicell-surface associated bispecific antibody to the tumor cell surface receptor for example EGFR
(depicted in Figure 3).

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:909916


912 Pharmaceutical biotechnology

for example EGFR [33] or HER2/neu receptor [34] on Mechanism of intracellular delivery of drug/si/
breast and ovarian cancer cells, respectively. Linkage of shRNA via drug/si/shRNA-packaged,
these two antibodies via their Fc regions was achieved BsAb-targeted minicells
using protein A/G. This ensures absence of complement-
mediated in vivo toxicity since the Fc part of each Once in the tumor microenvironment, the BsAb-targeted
monoclonal antibody is blocked by protein A/G. The minicells actively target tumor cells via binding of the
tumor targeting antibody can be varied depending on tumor receptor-specific antibody to the tumor cell surface
the desired tumor target. receptor. In vitro studies demonstrated that the minicells
are endocytosed, degraded in late endosomes/lysosomes
Mechanism of passive targeting of solid and the payload (drug or siRNA or shRNA) is released
tumors post-i.v. administration of drug/si/ ([23,24]; schematic shown in Figure 3). These studies
shRNA-packaged, BsAb-targeted minicells also showed that the payload appears to escape into the
Post-intravenous administration, the BsAb-targeted, tumor-cell cytoplasm in therapeutically significant concen-
drug/siRNA/shRNA-packaged minicells appear to trations although the mechanism of lysosomal membrane
rapidly fall out of the vascular circulation and into escape is not understood. Interestingly, payloads like
the tumor microenvironment possibly due to the leaky shRNA encoding plasmids also escape the lysosomal mem-
vasculature associated with solid tumors, a phenom- brane and are translocated into the tumor cell nucleus
enon recognized as the enhanced permeation and where shRNA expression occurs. In vivo xenograft studies
retention effect (EPR; [35,36,37,38,6]; schematic reveal that therapeutically significant concentrations of
shown in Figure 2). shRNA are expressed in the tumor xenograft to enable
tumor stabilization and even regression. This has been
These investigators demonstrated that most solid observed following the expression of shRNAs or delivered
tumors have blood vessels with defective architecture siRNAs targeting mRNAs encoding multi-drug resistance
and usually produce extensive amounts of various vas- (MDR1, P-glycoprotein; [46]) and cell cycle-associated
cular permeability factors. Most solid tumors therefore proteins implicated in tumor cell proliferation, such as
exhibit enhanced vascular permeability thus ensuring polo-like kinase 1 (PLK1; [47]), kinesin spindle protein
nutrient and oxygen supply to tumor tissues. There is (KSP; [48]) and cyclin-dependent kinase 1 (CDK1; [49]).
general agreement that the fenestrations associated siRNAs or shRNAs delivered via BsAb-targeted minicells
with abnormal tumor vasculature can range from to tumor cells were shown to effect potent G2 arrest and
10 nm to 1000 nm or more depending on the tumor apoptosis both in vitro and in vivo.
type, malignancy, and stage of disease [39,40,41].
Additionally, it is also recognized that proliferating These cell-cycle associated proteins are of interest for
cancer cells in tumor tissue compress lymphatic vessels cancer therapy, since they are critically involved in cell
particularly at the center of the tumor [42] resulting in proliferation. Thus, PLK1 and CDK1 play an essential
inefficient drainage of fluid from the tumor center and role in the control of mitotic progression in proliferating
this coupled with vascular content leakage from tumor cells. KSP provides the propulsive forces required to
vessels causes interstitial hypertension which is thought separate centrosomes during prophase, enabling them
to reduce the delivery of therapeutic agents to solid to migrate to opposite poles and establish a functional
tumors [43,44]. bipolar spindle.

Although the EPR effect has provided an excellent oppor- Interestingly, chemotherapeutic payloads that produced
tunity for the development of a large number of different these potent anti-tumor effects were achieved with the
nanoparticle delivery systems for cancer therapy, in recent delivery of amounts of drug that are markedly smaller
times, new hurdles have been identified. For example, the than those required with systemic delivery of free drug.
larger the tumor, the greater the pathophysiological hetero- For example, highly significant anti-tumor effects were
geneity and the central area of these tumors do not appear observed with 1875-fold and 8000-fold lower amounts
to exhibit the EPR effect [45]. of Dox and Pac respectively delivered to xenografts via
minicells compared with the respective free drugs.
This unique phenomenon in solid tumors the EPR
effect is considered to be a landmark principle in This was evident in dog case studies where rapid tumor
tumor-targeting chemotherapy and is becoming increas- regression was evident in two dogs diagnosed with
ingly important for anticancer drug development. For advanced (stage IV) T-cell non-Hodgkins lymphoma
example, Doxil1, a PEGylated (polyethylene glycol- (NHL) when treated i.v. with anti-canine-CD3minicellsDox
coated) doxorubicin-packaged liposome, DaunoX- [23]. One dog (4 kg) received a total of five doses over
ome1, liposomal daunorubicin, and Abraxane1, a nano- 35 days, and the other (40 kg), seven doses over 48 days
particle albumin-bound paclitaxel have all been providing an average of 4.8 mg and 83.4 mg of Dox per dose
approved for treatment of several different cancers. respectively. Interestingly, conventional chemotherapy in

Current Opinion in Biotechnology 2011, 22:909916 www.sciencedirect.com


Minicells as versatile vectors for targeted delivery MacDiarmid and Brahmbhatt 913

Figure 3

(a) Targeted minicell carrying therapeutic (b) minicells are swallowed


drug or siRNA binds to cancer cell by the cancer cell
surface receptor

(c) minicells broken down


B in the cancer cell

(d) Drug or siRNA escapes


E
from the minicell and
enters the cancer cell
(e) Tumor cell death
Current Opinion in Biotechnology

Schematic showing mechanism of intracellular delivery of drugs/siRNAs. Schematic showing the mechanism of minicell-mediated intracellular delivery
of the payload. The key steps are shown sequentially from A to E where (a) active binding of the minicell to the tumor cell-surface receptor via the
bispecific antibody, (b) endocytosis of the minicells by the tumor cells, (c) breakdown of the minicells in late endosomes/lysosomes, (d) release of drug
or siRNA into the vacuole and subsequent escape from the lysosomal membrane and into the cell cytoplasm, (e) tumor cell death following binding of
the therapeutic moiety to the respective intracellular target.

these dogs would require the administration of 8470 mg resulted in a dramatic reversal of drug resistance in vivo in
and 39,300 mg of Dox per dose respectively (30 mg/m2) as various xenograft murine models and even highly aggres-
part of multi-drug combination chemotherapy. Thus the sive multi-drug resistant uterine cancer xenografts were
treatment with CD3minicellsDox required 1764-fold and eliminated with 100% survival in mice [24] treated with
471-fold less Dox per dose respectively, to achieve highly the dual-minicell protocol. These results show that drug
significant tumor regression. resistance can be effectively reversed in previously highly
resistant tumor xenografts using minicell-mediated si/
Treatment of drug resistant tumor via shRNA delivery, and that such tumors are then exqui-
sequential minicell-mediated delivery of sitely sensitive to the second wave of minicells packaged
siRNA followed by drugs with the cytotoxic drug. This dual treatment strategy may
Drug-resistance is a major limitation to effective long- assist in the development of personalized treatment of
term cancer treatment [50]. Proof-of-concept studies in cancer particularly in the treatment of late-stage cancers
vivo to treat drug-resistant cancers via minicell-delivered where current treatment options are seriously limited.
therapies was demonstrated [24] using a dual sequential
treatment protocol in which the first treatment targets a Biodistribution of minicells in mouse
known drug resistance mechanism (e.g. overexpression of xenograft studies
the multi-drug resistance protein MDR1) via BsAb-tar- The biodistribution of i.v. administered 125I-labeled-mini-
geted, si/shRNA-packaged minicells. After allowing for cells in nude mice with EGFR overexpressing breast
sufficient time to achieve highly significant knockdown of cancer (MDA-MB-468) xenografts revealed that at two
the drug-resistance mediating protein, a second i.v. hours post-treatment, 30% of the EGFRminicells were
administration is carried out with BsAb-targeted, cyto- localized in the tumor [23]. This was in contrast to 3% of
toxic drug-packaged minicells where the tumor was the anti-EGFR/O-polysaccharide BsAb that accumulated
known to be highly resistant to the cytotoxic drug. This in the tumor at two hours post-i.v. administration. By 6 and

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:909916


914 Pharmaceutical biotechnology

24 hours, 4.6% and 0.5%, respectively, of specifically (controls). The treatments were administered four times,
targeted EGFRminicells remained in the tumors. This data 24 hours apart. Groups of mice were sacrificed at 4 (early
suggested that the EGFRminicells rapidly extravasate from response) and 24 (late response) hours post-treatment and
the tumor-associated leaky vasculature and are trapped in serum was collected and assayed for mouse and human
the tumor microenvironment likely due to the EPR effect. Type I and II interferons, and for inflammatory cytokines
produced by cells of the immune system. These cyto-
Biodistribution of minicell-packaged drug following i.v. kines are known to be elicited following siRNA delivery
administration of EGFRminicellsDox or free Dox to nude in mice [52].
mice with breast cancer xenografts (tumor volume be-
tween 140 and 170 mm3) showed that at six hours, 30% The results showed that at both time points, human
of the Dox dose administered by the EGFRminicellsDox interferon and cytokine levels were low (range of 10
was found in the tumors, as compared to only 1% of free to 30 pg/ml) and the levels elicited by EGFRminicells-
Dox. siPlk1 were indistinguishable from those observed with
saline or EGFRminicellssiNonsense. In contrast, mouse inter-
Similar results were observed in mice with much larger feron and cytokine responses were higher, being in the
tumors (400600 mm3), in which 28.1% of the Dox dose range of 50 to 150 pg/ml for IFN-a, IFN-b and IFN-g,
in EGFRminicellsDox was found in the tumors at six hours, 300 to 1200 pg/ml for TNF-a, and 10 to 100 pg/ml
as compared to only 0.21% of that in non-targeted, for IL-6. Both IFN-g and IL-6 showed a spike at four
minicellsDox, and 1.8% for free Dox. Plasma concen- hours but returned to baseline (saline control level) by 24
tration of Dox at both time-points was undetectable. hours. Moreover, the TNF-a response was greater at 24
hours than at four hours. Importantly, however, there
At six hours, biodistribution to the liver, spleen, and lungs were no significant differences in any of the interferon
was higher with the EGFRminicellsDox and minicellsDox or cytokine levels in mice treated with EGFRminicells-
EGFR
compared to free Dox and showed a rapid decrease by 24 siPlk1, compared to minicellssiNonsense. The significant
hours. rise in mouse TNF-a and IL-6 at 24 hours may be
attributed to the minicell vector itself since it is well
Thus targeted minicell delivery provides at least a 30-fold known that systemic administration of bacterial cells does
enrichment in tumor drug delivery. activate the Toll-like receptors resulting in TNF-a and
IL-6 responses. However, with minicell administrations
Immune and cytokine/interferon response to in mice, these responses have been self-limiting and have
drug/si/shRNA-packaged, bispecific antibody not resulted in toxicity to the mice.
targeted minicells
In mouse xenograft studies there was no evidence of any The above results indicate that the potent anti-tumor
toxicity [23,24] despite repeat dosing of BsAb-tar- effects observed in mouse xenografts are unlikely to be
geted, drug/si/shRNA-packaged minicells (1520 doses due to interferon or inflammatory cytokine responses,
have been administered in several mouse xenograft since treatment with EGFR-targeted siNonsense-pack-
experiments). This was evident by the lack of a febrile aged minicells did not produce any anti-tumor effects
response, weight loss, or skin/fur changes etc. in the despite similar interferon and cytokine responses as those
murine xenograft model. Importantly, minicells were well in mice treated with EGFRminicellssiPlk1.
tolerated with no adverse side-effects or deaths in any of
the treated animals. Conclusions
The minicell vector has quite remarkable properties in
Since minicells are of bacterial origin, it is necessary to be being able to package, in therapeutically significant con-
cautious with systemic administration as bacterial pro- centrations, a range of different drugs, siRNAs or shRNAs.
ducts are known to elicit potent inflammatory responses Each of these loadings is carried out following a simple co-
activated by Toll-like receptors [51]. A minicell purifi- incubation of the therapeutic moiety with the intact mini-
cation procedure to eliminate free endotoxin and free cells in an appropriate buffer. The size of the minicell
bacterial components was developed to minimize the (400 nm) ensures retention within the reticuloendothe-
potential for toxic side effects [23,24]. lial system and prevents it from penetration into normal
tissues which is a hurdle faced by drug-conjugated mono-
Recent studies have questioned whether the anti-tumor clonal antibody therapeutics. For example, extravasation of
effects of siRNA are due to specific knockdown of target Cetuximab1 (anti-EGFR monoclonal antibody) into the
mRNA or, rather, are non-specific and merely due to skin causes severe skin toxicity. The minicell vector is too
siRNA-mediated activation of the innate immune large to enter into normal skin, gastrointestinal or liver
response [52]. To address this issue, HCT116 xenograft tissue although these organs also exhibit leaky vasculature.
experiment was carried out [24] with mice being treated However, the fenestrations in these normal tissues range
with EGFRminicellssiPlk1, EGFRminicellssiNonsense or saline from 50 nm to 100 nm, thus preventing minicell-based

Current Opinion in Biotechnology 2011, 22:909916 www.sciencedirect.com


Minicells as versatile vectors for targeted delivery MacDiarmid and Brahmbhatt 915

therapeutics from entering into these tissues. This was 12. Aagaard L, Rossi JJ: RNAi therapeutics: principles, prospects
 and challenges. Adv Drug Del Rev 2007, 59:75-86.
demonstrated by MacDiarmid and colleagues in a rhesus Discusses mechanistic aspects of RNAi, the potential problem areas and
monkey trial (n = 36) where anti-monkey EGFR mono- solutions and therapeutic applications.
clonal antibody-targeted, doxorubicin-packaged minicells 13. Kawakami S, Hashida M: Targeted delivery systems of small
interfering RNA by systemic administration. Drug Metab
were administered i.v. in monkeys in a dose-escalation Pharmacokinet 2007, 22:142-151.
toxicology trial. Five repeat doses (one per week) were
14. Shim MS, Kwon YJ: Efficient and targeted delivery of siRNA in
administered. The results showed complete absence of vivo. FEBS J 2010, 277:4814-4827.
toxicity in the monkeys and inflammatory cytokine and
15. Bartlett DW, Davis ME: Impact of tumor-specific targeting and
immune responses were also normal (unpublished data). dosing schedule on tumor growth inhibition after intravenous
administration of siRNA-containing nanoparticles. Biotechnol
Bioeng 2008, 99:975-985.
These results paved the way for the first-in-man, Phase I/
IIa multi-center clinical trial in cancer patients and this 16. Cerchia L, Giangrande PH, McNamara JO, de Franciscis V: Cell-
 specific aptamers for targeted therapies. Methods Mol Biol
trial is currently in progress. 2009, 535:59-78.
Recent advances in the field of aptamers is reviewed together with
detailed descriptions of the relevant experimental approaches.
Acknowledgement
We thank Martin Hale, Animated Biomedical Productions, Sydney, for the 17. Pirollo KF, Chang EH: Targeted delivery of small interfering
artwork in Figures 13.  RNA: approaching effective cancer therapies. Cancer Res
2008, 68:1247-1250.
Discusses the current hurdles in the development of development of
References and recommended reading tumor-targeted delivery systems for in vivo use and suggests the devel-
Papers of particular interest, published within the period of review, opment of immunoliposomes nanocomplex to overcome the problems.
have been highlighted as:
18. Sorensen DR, Leirdal M, Sioud M: Gene silencing by systemic
delivery of synthetic siRNAs in adult mice. J Mol Biol 2003,
 of special interest
327:761-766.
 of outstanding interest
19. Adler HI, Fisher WD, Cohen A, Hardigree AA: Miniature
 Escherichia coli cells deficient in DNA. Proc Natl Acad Sci USA
1. Torchilin VP: Recent advances with liposomes as 1967, 57:321-326.
 pharmaceutical carriers. Nat Rev Drug Discov 2005, 4:145-160. Seminal paper describing the discovery and early characterization of
Comprehensive insightful review describing recent innovations in devel- bacterially derived minicells.
opment of liposomal drug carriers.
20. De Boer PA, Crossley RE, Rothfield LI: A division inhibitor and a
2. Matsumura Y: Preclinical and clinical studies of NK012, an SN- topological specificity factor coded for by the minicell locus
38-incorporating polymeric micelles, which is designed based determine proper placement of the division septum in E. coli.
on EPR effect. Adv Drug Deliv Rev 2010. [Epub ahead of print]. Cell 1989, 56:641-649.
3. Ferrari M: Cancer nanotechnology: opportunities and 21. Ma L, King GF, Rothfield L: Positioning of the MinE binding site
challenges. Nat Rev Cancer 2005, 5:161-171.  on the MinD surface suggests a plausible mechanism for
activation of the Escherichia coli MinD ATPase during division
4. Byrne JD, Betancourt T, Brannon-Peppas L: Active targeting
site selection. Mol Microbiol 2004, 54:99-108.
 schemes for nanoparticle systems in cancer therapeutics. Adv
Study shows the interaction between the Min proteins in the accurate
Drug Deliv Rev 2008, 60:1615-1626.
placement of cell division septation site in Gram-negative bacterial
Outlines current major cancer targets for nanoparticle systems and gives
cells.
insight into the direction of the field.
22. Lutkenhaus J: Assembly dynamics of the bacterial MinCDE
5. Greco F, Vicent MJ: Combination therapy: opportunities and
 system and spatial regulation of the Z ring. Ann Rev Biochem
challenges for polymerdrug conjugates as anticancer
2007, 76:539-562.
nanomedicines. Adv Drug Deliv Rev 2009, 61:1203-1213.
Reviews the understanding of how prokaryotic bacterial cytokinesis
6. Jain KK: Advances in the field of nanooncology. BMC Med occurs through the interaction of proteins involved in the Z ring formation
 2010, 8:83-94. and consequent bacterial cell division.
Current review on emerging developments in nanooncology.
23. MacDiarmid JA, Mugridge NB, Weiss JC, Phillips L, Burn AL,
7. Nicholson RI, Gee JM, Harper ME: EGFR and cancer prognosis.  Paulin RP, Haasdyk JE, Dickson KA, Brahmbhatt VN, Pattison ST
Eur J Cancer 2001, 37:S9-S15. et al.: Bacterially derived 400 nm particles for encapsulation
and cancer cell targeting of chemotherapeutics. Cancer Cell
8. Rae JM, Lippman ME: Evaluation of novel epidermal growth 2007, 11:431-445.
factor receptor tyrosine kinase inhibitors. Breast Cancer Res First publication describing the use of bacterially derived minicells for
Treat 2004, 83:99-107. targeted drug delivery to tumor cells in vitro and in vivo and the mechan-
isms involved in intracellular drug delivery within tumor cells.
9. Chang JC: HER2 inhibition: from discovery to clinical practice.
 Clin Cancer Res 2007, 13:1-3. 24. MacDiarmid JA, Amaro-Mugridge NB, Madrid-Weiss J,
Briefly summarises clinical trial results achieved with anti-HER2 mono-  Sedilarou I, Wetzel S, Kochar K, Brahmbhatt VN, Phillips L,
clonal antibody, Trastuzumab, and describes predictive markers and Pattison ST, Petti C et al.: Sequential treatment of drug-
mechanism of action and resistance. resistant tumors with targeted minicells containing siRNA
or a cytotoxic drug. Nat Biotechnol 2009,
10. Bardelli A, Siena S: Molecular mechanisms of resistance to 27:643-651.
 cetuximab and panitumumab in colorectal cancer. J Clin Oncol Second publication following reference [23] which describes the packa-
2010, 28:1254-1261. ging of therapeutically significant concentrations of siRNA or shRNA in
Discusses the evidence that metastatic colorectal cancers respond minicells and targeted delivery to tumor cells in vitro and in vivo.
differently to EGFR-targeted agents and that the tumor-specific response
has a genetic basis The role of oncogenic activation of EGFR downstream 25. Page`s JM, James CE, Winterhalter M: The porin and the
effectors on response to therapy is also discussed. permeating antibiotic: a selective diffusion barrier in
Gram-negative bacteria. Nat Rev Microbiol 2008,
11. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC: 6:893-903.
 Potent and specific genetic interference by double-stranded
RNA in Caenorhabditis elegans. Nature 1998, 391:806-811. 26. Nestorovich EM, Danelon C, Winterhalter M, Bezrukov SM:
This is a seminal paper describing the first discovery of siRNA.  Designed to penetrate: time-resolved interaction of single

www.sciencedirect.com Current Opinion in Biotechnology 2011, 22:909916


916 Pharmaceutical biotechnology

antibiotic molecules with bacterial pores. Proc Natl Acad Sci tumor vessels: role of tumor type and microenvironment. Proc
USA 2002, 99:9789-9794. Natl Acad Sci USA 1998, 95:4607-4612.
Study shows the mechanism by which an antibiotic like ampicillin pene- Early studies analyzing the functional limits of transvascular transport of
trates a bacterial membrane protein channel and how the process of entry macromolecules and its modulation by the tumor microenvironment.
is facilitated by complementary charges between the antibiotic and the
channel architecture. 40. Siwak DR, Tari AM, Lopez-Berestein G: The potential of drug-
carrying immunoliposomes as anticancer agents. Clin Cancer
27. Danelon C, Nestrovich EM, Winterhalter M, Ceccarelli M, Res 2002, 8:955-956.
Bezrukov SM: Interaction of zwitterionic penicillins with the
OmpF channel facilitates their translocation. Biophys J 2006, 41. Hashizume H, Baluk P, Morikawa S, McLean JW, Thurston G,
90:1617-1627.  Roberge S, Jain RK, McDonald MD: Openings between
defective endothelial cells explain tumor vessel leakiness. Am
28. Schulz GE: Bacterial porins: structure and function. Curr Opin J Pathol 2000, 156:1363-1380.
 Cell Biol 1993, 5:701-707. Using a range of microscopy techniques, tumor architecture and the
Discusses three-dimensional crystal structures of porins and suggests morphology of defective blood vessels along with the size of fenestrae are
possible mechanisms by which porins may be able to discriminate clearly shown in different tumors.
between polar and non-polar solutes.
42. Padera TP, Stoll BR, Tooredman JB, Capen D, di Tomaso E,
29. Van den Berg B, Black PN, Clemons WM Jr, Rapoport TA:  Jain RK: Pathology: cancer cells compress intratumour
Crystal structure of the long-chain fatty acid transporter FadL. vessels. Nature 2004, 427:695.
Science 2004, 304:1506-1509. Provides evidence that proliferating cancer cells cause intratumour ves-
sels to compress and collapse.
30. Van den Berg B: The FadL family: unusual transporters for
 unusual substrates. Curr Opin Struct Biol 2005, 15:401-407. 43. Jain RK: Normalization of tumor vasculature: an emerging
Describes the possible mechanisms by which the FadL family of mem-  concept in antiangiogenic therapy. Science 2005, 307:58-62.
brane associated proteins may transport hydrophobic compounds into Reviews evidence supporting a hypothesis that certain antiangiogenic
bacterial cells. agents can transiently normalize the abnormal structure and function of
tumor vasculature to make it more efficient for oxygen and drug delivery.
31. Hong H, Patel DR, Tamm LK, van den Berg B: The outer
membrane protein OmpW forms an eight-stranded beta- 44. Danquah MK, Zhang XA, Mahato RI: Extravasation of polymeric
barrel with a hydrophobic channel. J Biol Chem 2006,  nanomedicines across tumor vasculature. Adv Drug Deliv Rev
281:7568-7577. 2010. [Epub ahead of print].
Reviews current knowledge on the tumor microenvironment generated
32. Park JW: Liposome-based drug delivery in breast cancer barriers which limit extravasation of drugs and focuses on modalities for
 treatment. Breast Cancer Res 2002, 4:95-99. overcoming these barriers using multi-functional polymeric carriers.
Discusses progress made with liposomal carriers for breast cancer
treatment and suggests future improvements with immunoliposomes. 45. Nagamitsu A, Greish K, Maeda H: Elevating blood pressure as a
 strategy to increase tumor-targeted delivery of
33. El-Rayes BF, LoRusso PM: Targeting the epidermal growth macromolecular drug SMANCS: cases of advanced solid
factor receptor. Br J Cancer 2004, 91:418-424. tumors. Jpn J Oncol 2009, 39:756-766.
Provides the first clinical proof that modulations of vascular pathophy-
34. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL:
siology can uniquely accomplish enhanced tumor selective delivery of
 Human breast cancer: correlation of relapse and survival with
polymeric drugs and thus yielded better clinical outcome.
amplification of the HER-2/neu oncogene. Science 1987,
235:177-182. 46. Goda K, Bacso Z, Szabo G: Multidrug resistance through the
Shows that HER-2/neu was found to be amplified from 2- to greater than spectacle of P-glycoprotein. Curr Cancer Drug Targets 2009,
20-fold in 30% of the 189 primary human breast cancers investigated. 9:281-297.
Amplification of the HER-2/neu gene was a significant predictor of both
overall survival and time to relapse in patients with breast cancer. 47. Strebhardt K: Multifaceted polo-like kinases: drug targets and
 antitargets for cancer therapy. Nat Rev Drug Discov 2010,
35. Matsumura Y, Maeda H: A new concept for macromolecular 9:643-660.
 therapeutics in cancer chemotherapy: mechanism of Reviews the biology of polo-like kinases and its potential as an anti-
tumoritropic accumulation of proteins and the antitumor cancer target.
agent smancs. Cancer Res 1986, 46:6387-6392.
Early studies showing that accumulation of macromolecules in tumor 48. Huszar D, Theoclitou ME, Skolnik J, Herbst R: Kinesin motor
tissue was likely due to enhanced permeability and retention effect. proteins as targets for cancer therapy. Cancer Metastasis Rev
2009, 28:197-208.
36. Maeda H, Matsumura Y: Tumoritropic and lymphotropic
principles of macromolecular drugs. Crit Rev Ther Drug Carrier 49. Nigg EA: Mitotic kinases as regulators of cell division and its
Syst 1989, 6:193-210. checkpoints. Nat Rev Mol Cell Biol 2001, 2:21-32.
37. Maeda H, Seymour LW, Miyamoto Y: Conjugates of anticancer 50. Baguley BC: Multiple drug resistance mechanisms in cancer.
agents and polymers: advantages of macromolecular Mol Biotechnol 2010, 46:308-316.
therapeutics in vivo. Bioconjug Chem 1992, 3:351-362.
51. Shizuo A, Takeda K: Toll-like receptor signaling. Nature Rev
38. Fang J, Nakamura H, Maeda H: The EPR effect: unique features Immunol 2004, 4:499-511.
 of tumor blood vessels for drug delivery, factors involved, and
limitations and augmentation of the effect. Adv Drug Del Rev 52. Robbins M, Judge A, Ambegia E, Choi C, Yaworski E, Palmer L,
2011, 63:136-151.  McClintock K, MacLachlan I: Misinterpreting the therapeutic
Discusses molecular mechanisms of factors related to the EPR effect, the effects of small interfering RNA caused by immune
unique anatomy of tumor vessels, limitations and techniques to avoid stimulation. Hum Gene Ther 2008, 19:991-999.
such limitations. This study cautions regarding interpretations of therapeutic benefits
following siRNA delivered in vivo in mice since unmodified siRNAs can
39. Hobbs SK, Monsky WL, Yuan F, Roberts WG, Griffith L, activate the innate immune response and elicit nonspecific therapeutic
 Torchilin VP, Jain RK et al.: Regulation of transport pathways in benefits.

Current Opinion in Biotechnology 2011, 22:909916 www.sciencedirect.com

Вам также может понравиться