Вы находитесь на странице: 1из 29

Immunological Responses to Exogenous Insulin

S. Edwin Fineberg, Thomas T. Kawabata, Deborah Finco-Kent, Robert J. Fountaine, Gregory L. Finch and Alan S.
Krasner

Endocr. Rev. 2007 28:625-652 originally published online Sep 4, 2007; , doi: 10.1210/er.2007-0002

To subscribe to Endocrine Reviews or any of the other journals published by The Endocrine
Society please go to: http://edrv.endojournals.org//subscriptions/

Copyright © The Endocrine Society. All rights reserved. Print ISSN: 0021-972X. Online
0163-769X/07/$20.00/0 Endocrine Reviews 28(6):625– 652
Printed in U.S.A. Copyright © 2007 by The Endocrine Society
doi: 10.1210/er.2007-0002

Immunological Responses to Exogenous Insulin


S. Edwin Fineberg, Thomas T. Kawabata, Deborah Finco-Kent, Robert J. Fountaine, Gregory L. Finch, and
Alan S. Krasner
Indiana University School of Medicine (S.E.F.), Indianapolis, Indiana 46202; University of Alabama Birmingham School of
Medicine (S.E.F.), Birmingham, Alabama 35294; and Pfizer Global Research and Development (T.T.K., D.F.-K., R.J.F.,
G.L.F., A.S.K.), Groton, Connecticut 06320

Regardless of purity and origin, therapeutic insulins continue insulin dose requirements, and incidence of hypoglycemia, or
to be immunogenic in humans. However, severe immunolog- contributes to ␤-cell failure or to long-term complications of
ical complications occur rarely, and less severe events affect diabetes. Studies in which pregnant women with diabetes
a small minority of patients. Insulin autoantibodies (IAAs) were monitored for glycemic control argue against a connec-
may be detectable in insulin-naive individuals who have a tion between IAs and fetal risk. Although studies have shown
high likelihood of developing type 1 diabetes or in patients increased levels of immune complexes in patients with dia-
who have had viral disorders, have been treated with various betic microangiopathic complications, these immune com-
drugs, or have autoimmune disorders or paraneoplastic syn- plexes often do not contain insulin or IAs, and insulin admin-
dromes. This suggests that under certain circumstances, im- istration does not contribute to their formation. The majority
mune tolerance to insulin can be overcome. Factors that can of studies have shown no relationship between IAs and dia-
lead to more or less susceptibility to humoral responses to betic angiopathic complications, including nephropathy, ret-
exogenous insulin include the recipient’s immune response inopathy, and neuropathy. With the advent of novel insulin
genes, age, the presence of sufficient circulating autologous formulations and delivery systems, such as insulin pumps and
insulin, and the site of insulin delivery. Little proof exists, inhaled insulin, examination of these issues is increasingly
however, that the development of insulin antibodies (IAs) to relevant. (Endocrine Reviews 28: 625– 652, 2007)
exogenous insulin therapy affects integrated glucose control,

I. Introduction V. Clinical Significance of Insulin Antibodies


II. Human Experience with Exogenous Insulin Therapy A. Hypersensitivity reactions
A. Animal insulin preparations B. Glycemic effects of insulin antibodies
B. Recombinant human insulins C. Pregnancy
C. Insulin analogs D. Autoimmune diseases
D. Subcutaneous and implantable insulin pumps E. Inhaled insulin antibodies and pulmonary function
E. Inhaled insulin F. Immune complexes
F. Modifying factors in the antibody response to exoge- G. Buffering effect of insulin antibodies
nous insulin H. Insulin antibodies and risk for diabetes
III. Predicting Human Immunogenicity with Animal Models VI. Conclusion
A. Effects of inhalation of insulin
B. Mucosal delivery of insulin and prevention of type 1
diabetes in NOD mice I. Introduction
IV. Characterization of Insulin Antibodies
A. Measurement of insulin antibodies
B. Immunoglobulin classes
C. Antibody affinity
D ESPITE EXPERIENCE FROM more than three quarters
of a century, uncertainties about the significance of
the immune response to therapeutic insulins remain. Such
D. Insulin epitope and insulin antibody idiotype analyses issues are largely the residuum of early experiences with
E. Characterization of insulin antibody immune complexes pancreatic extracts of animal insulins, however, and have
little connection to modern-day preparations of uncontam-
First Published Online September 4, 2007 inated human insulin, which now has a time-tested safety
Abbreviations: APC, antigen-presenting cell(s); AUC, area under the record. Currently, severe immunological sequelae occur only
curve; CPII, continuous peritoneal insulin infusion; CSII, continuous sc rarely, and less severe events affect a small minority of pa-
insulin infusion; dpm, disintegrations per minute; FEV1, forced expira- tients. Still, the following questions persist for physicians and
tory volume in 1 sec; HLA, histocompatibility leukocyte antigen; IA,
insulin antibody; IAA, insulin autoantibody, arising spontaneously in researchers:
patients not exposed to exogenous insulin; IAS, insulin autoimmune
syndrome; IIP, implantable insulin pump; MHC, major histocompati- • Why do IAs occur with the use of recombinant human
bility complex; NOD, nonobese diabetic (mouse); NPH, neutral prota- insulins?
mine Hagedorn; rDNA, recombinant DNA; RLB, radioligand binding
assay. • What are the key factors that determine immunogenicity
Endocrine Reviews is published by The Endocrine Society (http://
of insulin in humans?
www.endo-society.org), the foremost professional society serving the • What are the mechanisms that lead to a break in immu-
endocrine community. nological tolerance for insulin?

625
626 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

• What do IA detection and quantification techniques tell us The use of isoelectric precipitation by Eli Lilly and Com-
about the nature of the antibodies? pany increased the purity of exogenous bovine and porcine
• Do IAs lead to increased insulin dose requirements or insulin by 10- to 100-fold over previous preparations (2).
hypoglycemic events? However, improved analytical methods showed that crys-
• If the presence of autoantibodies to insulin (IAA) predicts tallized insulin included proinsulin and intermediates, non-
the development of type 1 diabetes in patients not previ- convertible insulin dimers, arginyl and ethyl insulin, deami-
ously treated with exogenous insulin, what is the impact dated insulin, monodesamido insulin, and various
on residual ␤-cell function of antibody responses to ex- noninsulin peptides (7–9). RIAs done in 1982 showed that
ogenous insulin therapy in patients with established twice-crystallized Novo monocomponent insulin contained
diabetes? proinsulin and its intermediates as well as trace amounts of
• Because antibodies cross the placenta, do IAs cause dia- other peptide hormones: proinsulin-like immunoreactivity
betes in a developing fetus or contribute to birth defects or up to 20,000 ppm, glucagon 45 ppm, pancreatic polypeptide
postnatal complications? 43 ppm, somatostatin 1.4 ppm, and vasoactive intestinal pep-
tide 0.3 ppm (10). These findings raised the possibility that
This review will evaluate these questions by examining the
noninsulin peptides were responsible for the antigenicity of
development of IAs to exogenous insulin (Table 1) and by
therapeutic insulins.
reviewing the literature on antibody responses to different
Local reactions to nonchromatographed insulins occurred
types of insulin therapies with a focus on the clinical impact
in up to 55% of recipients, although systemic reactions were
of IAs.
rarely seen (11). These dermal and systemic allergic reactions
to insulin were demonstrated to be associated with circulat-
II. Human Experience with Exogenous ing IgE and antibodies to insulin, which were directed to-
Insulin Therapy ward extracted but not native insulin (12). Patients being
treated with insulin were shown to have a broad array of IA
A. Animal insulin preparations binding classes (see Section IV.B) (13).
The first effective injectable insulins developed by Banting The incidence of insulin resistance of an immunological
and Best (1) were crude acid-ethanol extracts of abattoir nature continued to be quite rare—less than 0.1% of recip-
bovine or porcine pancreases. Potencies were highly vari- ients (14, 15). From the late 1970s onward, manufacturers
able, large volumes were required to reduce blood glucose in took further steps in insulin purification, including column
diabetic dogs, and injections often involved toxic reactions chromatography, separation of peptides by molecular siev-
and abscess formation. The subsequent discovery of an in- ing and charge (16), HPLC (17), and reversed phase HPLC
sulin crystallization method by Collip in 1922 led to the first (18). These highly purified monocomponent insulins, which
commercial insulin preparations for diabetes therapy (2). have less than 3 ppm proinsulin-like immunoreactivity, de-
As purification increased, complaints of local abscesses in creased but did not eliminate the development of IAs (19),
trial patients decreased; however, reports emerged of ana- dermal reactions, systemic hypersensitivity, and insulin re-
phylactic, local, and systemic hypersensitivity reactions (2, sistance (20 –22).
3). In one patient, Tuft (4) demonstrated immunological spec-
ificity for insulin protein with regard to urticaria and edema, B. Recombinant human insulins
and he was able to transfer dermal reactivity to nonsensitized
individuals (Prausnitz-Kustner reaction). The first human insulins for clinical use were derived from
Resistance to insulin therapy and the discordant appear- porcine insulin by semisynthetic conversion (23). The next
ance of cutaneous reactions to insulin were reported from the advance in insulin therapy was the development of recom-
1920s onward. The immunological nature of this resistance binant DNA (rDNA) human insulin made by fermentation of
was shown by insulin neutralization in mice that received Escherichia coli or Saccharomyces cerevisiae yeast that contained
sera from insulin-resistant patients (5). Clinical hypersensi- DNA for insulin A and B chains, proinsulin, or modified
tivity and insulin resistance were first linked to circulating proinsulin. Later steps after cell lysis include removal of
antibodies to insulin in the work of Berson et al. (6). bacterial and yeast components, recombination of insulin
chains, or removal of connecting peptides (24, 25).
TABLE 1. Factors affecting IA development Clinical trials revealed that semisynthetic human insulin
was less immunogenic than porcine monocomponent insulin
Insulin-related in patients newly diagnosed with type 1 diabetes. Schern-
Purity
Molecular structure
thaner et al. (26) found IAs of the IgG class in 14% of patients
Storage condition receiving human insulins and in 29% of the patients receiving
Formulation nonhuman insulins. Heding et al. (27) reported that 44% of
Excipients pediatric patients with type 1 diabetes receiving human
Dimer and oxidation products monocomponent insulin were antibody-positive, compared
Patient-related
Age with 59% of similar patients receiving porcine monocompo-
HLA type nent insulins. (These differences may be attributable to an-
Endogenous insulin tibody measurement methodologies, which will be discussed
Delivery route in Section IV.A) In a 1-yr trial, Fineberg et al. (28) compared
HLA, Histocompatibility leukocyte antigen. development of antibodies to rDNA human insulin in 100
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 627

insulin-naive children and adults with 121 similar individ- reactions and severe dermal reactions have been reported
uals treated with purified porcine insulin. At the end of the (45, 46).
trial period, 44% of patients treated with rDNA human in- Insulin glargine is modified by the addition of two argi-
sulin and 60% of patients treated with porcine insulin had nines to B30 (B31 and B32) and replacement of A21 Asp with
developed significant levels of IAs. Gly. Treatment with glargine in patients previously treated
In a retrospective review, injection site reactions with with insulin for at least 1 yr did not result in increased IA
highly purified mixed bovine (70% bovine, 30% porcine), levels (47).
purified porcine, and rDNA human insulin were reported to
be 12, 3.9, and 2.4%, respectively (29). Sporadic case reports D. Subcutaneous and implantable insulin pumps
have been published regarding individuals who developed
insulin resistance and/or systemic hypersensitivity reactions In a small, 12-month study, 45 patients with type 1 diabetes
while being treated with rDNA human insulins, including were randomized to conventional therapy, multiple dose, or
short-acting and long-acting human insulin analogs (30 –34). continuous sc insulin infusion (CSII) with highly purified
porcine insulins (48). Researchers found an increase in IAs in
the multiple dose and CSII groups but not in the conventional
C. Insulin analogs group, suggesting that differences in methods of sc insulin
administration affect antibody development (the study has
Insulin’s tendency to self-associate in concentrated solu- not been repeated with contemporary human insulins) (49,
tions to form dimers and hexamers is largely the reason that 50).
sc regular rDNA human insulin and animal insulin extracts Implantable insulin pumps (IPPs) have been associated in
have nonphysiological action profiles (35). In response to this various studies with increases in IA levels, leading research-
problem, researchers developed rDNA human insulin ana- ers to suggest causative mechanisms such as the storage of
logs that have decreased tendencies to self-associate. Cur- insulin in the pump reservoir or to the presence of surfactant
rently, three rapid-acting human insulin analogs are in clin- or polyethylene glycol (48 –50). In a 2002 study, two groups
ical use: lispro, aspart, and glulisine. These analogs consist of of patients with type 1 diabetes were challenged for 6 months
molecular modifications that result in few, if any, differences with either continuous peritoneal insulin infusion (CPII) or
from unmodified human insulin pertaining to antigenicity CSII (400 or 100 U/ml, respectively) using Hoe 21 PH, a
and receptor binding of insulin (36, 37). Amino acids essen- semisynthetic human insulin using polyethylene glycol. An-
tial for insulin receptor binding are A1 (Gly), A2 (Ile), A3 tibodies in the CPII group increased significantly vs. the CSII
(Val), A19 (Tyr), B6 (Leu), B12 (Val), B23 (Gly), B24 (Phe), and group. Although these data argue against the insulin prep-
B25 (Asp). In addition, IGF-I binding is affected by B10 (His) aration itself as the culprit, they do not eliminate the possi-
and B26 –30 (Tyr-Thr-Pro-Lys-Ala). Insulin lispro has a re- bility of storage degradation or an effect of silicone oil con-
versal of the sequence of amino acids B28 and B29 (Lis-Pro tributing to the increased antibody development associated
vs. Pro-Lis), lowering the potential for dimer formation. In- with CPII (see Section V.B.3.c) (51).
sulin aspart substitutes Asp B28 for B28 (Pro). Both of these
rapid-acting insulins have IGF-I receptor binding equivalent
E. Inhaled insulin
to unmodified human insulin. Insulin glulisine substitutes
Glu for B29 (Lis) and Lis for B3 (Asp), resulting in decreased Despite well-established, long-term benefits of good gly-
self-association but somewhat longer residence time on the cemic control (52), many patients and physicians are reluc-
IGF-I receptor (38). The rapid-acting insulin analogs, when tant to initiate exogenous insulin therapy (53, 54). In response
given sc, result in development of humoral IAs, with no to this reluctance, researchers have explored pulmonary de-
significant differences shown between the analogs and un- livery of insulin as an alternative to sc injection (55). Inhaled
modified human insulin (39 – 42). In all the clinical trials human insulin [Exubera (insulin human [rDNA origin]) In-
cited, IAs were cross-reactive, i.e., they react equally to un- halation Powder] is the first inhalable insulin approved by
modified and analog insulins with no demonstrable effects the United States and the European Union for the treatment
on insulin dose, hypoglycemic events, or glycemic control. of type 1 and type 2 diabetes. The Exubera inhaler delivers
The rapid-acting human insulin analogs are available pre- dry powder human insulin contained in blister packets (56).
mixed with modifications having intermediate-duration Diabetes trials with inhaled insulins have been reviewed
characteristics (e.g., insulin lispro protamine suspension). in a recent article (57), and IA data have been presented for
Antibodies directed to or cross-reacting antibodies with pro- Exubera (Pfizer Inc., New York, NY) in patients with type 1
tamine have been associated with anaphylaxis during rever- and 2 diabetes (58, 59), and also for the AERx iDMS system
sal of anticoagulation in patients treated with neutral pro- (Novo Nordisk, Bagsvaerd, Denmark) (60). [All inhaled in-
tamine Hagedorn (NPH) insulin (43, 44). sulins currently are used as premeal insulins because they are
Two commercially available long-acting human insulin absorbed in the deep lung tissues as rapidly as short-acting
analogs are detemir and glargine. Insulin detemir is des B30 insulin analogs but do not have enough duration of activity
Thr with a B29 side chain (Lys-myristic acid [tetradecanoyl-]). to meet basal insulin needs (58, 61, 62).]
When concentrated, detemir forms hexamers in sc injection In the Exubera trials, IA development in patients with type
sites and, after absorption of insulin monomers, binds re- 1 diabetes exceeded that in comparator regular sc human
versibly to albumin. IA data from clinical trials of this analog insulin groups; median IA binding increased from 3 to 29%
have not been presented; however, type 3 hypersensitivity in the Exubera group, compared with no change in the sc
628 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

group (58). Among patients with type 2 diabetes who were helper T cells with a corresponding T cell receptor that rec-
previously treated with insulin, median IA binding increased ognizes the peptide. In addition to MHC-peptide-T cell re-
to 6% in the Exubera group and was unchanged in the sc ceptor binding, a second signal from the binding of B7 (co-
group. In patients with type 2 diabetes who had not previ- stimulatory molecule) on APCs to CD28 on T cells will lead
ously used insulin, no change in median levels of IA binding to T cell activation and the production of various cytokines.
was seen in either the Exubera or comparator group (treated Costimulatory molecules are up-regulated by proinflamma-
with an oral antihyperglycemic agent). Differences between tory cytokines produced with tissue damage or infection.
patients with type 1 and type 2 diabetes were maintained Protein antigen binding to the Ig B-cell receptors along with
throughout a 24-month extension study. The IAs were of the cytokines from the activated T cells leads to the proliferation
IgG class (Fig. 1), and no effects on insulin efficacy or adverse and differentiation of B cells to antibody-secreting plasma
effects could be attributed to IA development (58, 59). Fur- cells.
thermore, in another study IA levels were shown to decline Immunological tolerance prevents the development of an-
after cessation of Exubera (63). tibody response to self-proteins. This may involve clonal
The AERx iDMS device delivers a liquid droplet of human deletion of T cells that recognize self-proteins during devel-
insulin onto a dosage strip for inhalation (64). In a 12-wk opment in the thymus. Without costimulation by APC, T cells
intensive treatment trial among patients with type 2 diabetes may become anergic and will not respond to antigen. Tol-
being treated with insulin, there was a significant increase in erance may also occur by means of regulatory T cells that
IAs in the inhaled insulin group, but no change in the com- actively down modulate T-helper cell activity. Down-regu-
parator sc group (60). No clinical effects were attributed to lation of immune responses to self and nonself antigens
IAs. involves a subset of regulatory T lymphocytes that express
Causes of the increased antibody development with in- CD4, CD25, and the transcription factor Foxp3 (66). These
haled insulin compared with sc insulin therapy have not regulatory T cells, called suppressor T lymphocytes in the
been identified. The excipients used in the formulation of past, may be critical in warding off or terminating autoim-
powdered insulin are not known to incite an immune re- munity (67– 69). It is likely that these regulatory cells may
sponse, nor can structural differences or alterations in bio- also play a role in the development and levels of immune
activity be demonstrated (58, 59). However, differences in responses to exogenous insulin. These cells are present
site of delivery per se may be paramount, because both liquid throughout life in humans and have a very rapid turnover
and powdered formulations of pulmonary insulin are more rate. The peripheral T regulatory cells are prone to apoptosis
immunogenic than sc comparators. and may require the presence of continuous concentrations
of self-antigens and/or growth factors such as TGF-␤ (70).
F. Modifying factors in the antibody response to Such conditions may be less prevalent in autoimmune dis-
exogenous insulin eases, such as type 1 diabetes, resulting in deficient T-reg-
ulatory cell numbers or function (71, 72). T-regulatory cell
1. Immunological mechanism of antibody responses. The gener- deficiencies have been demonstrated in a mouse model of
ation of a primary antibody response to foreign proteins autoimmune diabetes (73, 74). Tiittanen et al. (75) recently
involves the uptake by antigen-presenting cells (APC) such reported that insulin treatment may activate insulin-specific
as dendritic cells (65). Dendritic APC are found throughout regulatory T cells in children with newly diagnosed type 1
the body, including all current insulin delivery sites, i.e., the diabetes.
sc space, the lungs, and lymph nodes that drain the perito- The H2b strain of mouse does not form antibodies to
neum. The protein antigen is processed to smaller peptide porcine insulin; however, both helper and suppressor T cells
fragments in endosomal vesicles. These peptides bind to are activated in that strain, allowing the secondary devel-
major histocompatibility complex (MHC) class II molecules opment of antibodies to bovine insulin but suppressing the
and are transported to the surface of the APC. There are response to porcine insulin. When T suppressor cells are
different types of class II molecules, and each has multiple removed, in vitro development of antiporcine IAs can be
alleles. MHC binding of antigen fragments is determined by shown (76). In addition, nonresponder strains of mice im-
the peptide sequence and type of MHC molecule. Thus, not munized with porcine insulin have been shown to have
all peptides formed associate with MHC. primed T helper cells and dominant T suppressor cells that
The MHC-peptide complex on the APC surface binds to cross-react with autologous insulin. Thus, the balance be-
tween the effects of T suppressor and T helper cells deter-
mined the effectiveness of immune tolerance to heterologous
and autologous insulins (77).

2. Sequence differences and impurities. Differences between the


amino acid sequence of a protein and the sequence of a
self-protein will increase its inherent immunogenicity. As
shown in Table 2, the sequences of bovine and porcine in-
sulin differ from human insulin by three amino acids and one
FIG. 1. Findings from the Exubera trials indicate that IAs associated amino acid, respectively. Therefore, the administration of
with inhaled and sc insulin therapy are predominantly of the IgG bovine insulin or bovine-porcine insulin mixtures resulted in
class (58). A wide range of IgG levels was observed. greater IA responses than porcine or human insulin (21,
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 629

TABLE 2. Comparison of amino acid sequences in human and animal insulin

A chain B chain No. of differences


Human GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKT
Chimpanzee GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKT 0
AfrGr GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKT 0
Macaque GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKT 0
Pig GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 1
Dog GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 1
Squirrel GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKS 1
Rabbit GIVEQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKS 1
Hamster GIVDQCCTSICSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKS 2
Elephant GIVEQCCTGVCSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKT 2
Cow GIVEQCCASVCSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 3
Whale GIVEQCCASTCSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 3
Cat GIVEQCCASVCSLYQLEHYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 4
Mouse GIVDQCCTSICSLYQLENYCN FVKQHLCGPHLVEALYLVCGERGFFYTPKS 4
Rat GIVDQCCTSICSLYQLENYCN FVKQHLCGPHLVEALYLVCGERGFFYTPKS 4
Sheep GIVEQCCAGVCSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 4
Goat GIVEQCCAGVCSLYQLENYCN FVNQHLCGSHLVEALYLVCGERGFFYTPKA 4
Fish (bonito) GIHEQCCHKPCDIFQLENYCN AANPHLCGSHLVDALYLVCGDRGFFYQPK– 16
Guinea pig GIVDQCCTGTCTRHQLQSYCN FVSRHLCGSNLVETLYSVCQDDGFFYIPKD 18
The sequences of amino acids for insulin A and B chains are given for each species. Differences from human insulin are highlighted in bold
type, and the number of differences is given in the far right column. Sources: National Center for Biotechnology, GenBank, www.ncbi.nlm.
nih.gov/Genbank; National Biomedical Research Foundation Protein Identification Resource, Protein Sequence Database. Fish (bonito) se-
quence from J. F. Cutfield et al.: Eur J Biochem 158:117–123, 1986 (369). AfrGr, African green monkey.

78 – 80). Studies of insulin aspart, which differs from human antibodies that bind to insulin dimers have been demon-
insulin by one amino acid, have shown a robust antibody strated (92). Lymphocyte transformation studies suggest that
response in a subpopulation of patients (81). However, in- insulin aggregates, rather than monomers, are the cause of
sulin lispro is thought to have comparable rates of antibody persistent cutaneous allergy (93). Insulin aggregation and
formation as regular human insulin (40). degradation products in a glycerol-based formulation of in-
Owing to the presence of contaminants (including gluca- sulin for use in IPPs have been shown to be associated with
gon, pancreatic polypeptide, somatostatin, proinsulin, and increased immunogenicity (94).
insulin complexes) in early bovine and mixed bovine-porcine Studies in animal models also indicate that aggregates are
preparations, some of the antibodies generated may have more immunogenic. Insulin samples taken directly from an
been against noninsulin proteins (see Section II.A) (30). More- insulin vial (material that contained no aggregates) and in-
over, other contaminants may also have adjuvant effects and sulin from an ip pump reservoir (material with a high level
thereby enhance the generation of IAs (19). This finding is of aggregates) were administered to rats, and IA levels were
consistent with studies that have shown minimal difference subsequently measured (95). The percentage binding in sera
in immunogenic potential between purified porcine and hu- from rats immunized with insulin from the pump reservoir
man insulin (28, 78, 82). was approximately 2-fold higher than that obtained with
Most commonly, IAs that develop in response to exoge-
nous bovine, porcine, or human insulins are cross-reactive
with all three species (Fig. 2 and Table 3) (83– 86). In rare
cases, however, antibodies bind differentially to insulins
from different species, correlating with clinical responses
observed when switching insulin sources (87, 88).

3. Formulation and aggregates. Insulin formulation can also


affect immunogenic potential. Soluble forms of insulin, such
as regular and semilente, are less allergenic than intermedi-
ate- or long-acting preparations, such as NPH, lente, prota-
mine zinc, and ultralente (21, 89). Acid preparations are more
immunogenic than neutral ones (21). Allergic reactions to
both protamine and zinc have been reported and require
special testing to distinguish from insulin allergy (90, 91).
FIG. 2. In 1988, Fineberg et al. (85) reported antibody-bound insulin
Aggregation can clearly affect insulin immunogenicity. in previously insulin-naive individuals with type 1 or type 2 diabetes
This is also true for other biological therapeutics and has treated with rDNA human insulin, purified porcine insulin, or mixed
resulted in strict limitation on aggregates in final drug prod- bovine-porcine insulin over 2 yr. On the left, patients with type 1
ucts. It is thought that insulin aggregates may remain for diabetes reach a peak response within 6 months, with the greatest
response to mixed bovine-porcine insulin. On the right, antibody re-
longer periods at the injection site or be more readily taken sponses for patients with type 2 diabetes are diminished for all the
up by APC. Insulin aggregates are present in the circulation insulins. The insets compare antibody response to human and porcine
of patients with diabetes being treated with insulin (92), and insulin.
630 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

TABLE 3. Cross-reactivity of IAs

Median Lower quartile Upper quartile


n
(␮U/ml) (␮U/ml) (␮U/ml)
Patients with type 1 diabetes treated with EXU
Human IA 353 29.0 13.0 44.0
Bovine IA 353 23.0 12.0 37.0
Porcine IA 353 25.0 12.0 42.0
Insulin-using patients with type 2 diabetes
treated with EXU
Human IA 134 5.0 1.5 15.0
Bovine IA 134 4.0 1.5 14.0
Porcine IA 134 4.0 1.5 11.0
EXU, Inhaled human insulin. Data on file, Pfizer Inc., New York, NY.

insulin directly from the vial, suggesting that insulin aggre- whereas HLA-B8 and HLA-DR3 are associated with de-
gates formed in the reservoir were slightly more immuno- creased formation rates (26, 107–110). However, Asplin et al.
genic. This may be relevant to elevated IA levels that have (111) assessed the development of IAs in 54 newly treated
been observed in patients with implanted ip pumps (see patients with type 1 diabetes and found no effects of HLA-
Sections II.D and V.B.3.c). DR3, HLA-DR4, or combinations on insulin-binding anti-
body formation. Most of these studies were conducted dur-
4. Route of delivery. Animal and human studies suggest that
ing the late 1970s through the 1980s and evaluated antibody
the site of antigen delivery can affect antibody responses.
responses in patients treated with bovine or porcine insulin
Insulin-specific tolerance induction by iv injection in mice is
preparations; however, Fineberg et al. (83) studied patients
antigen-specific and dose-dependent. The same dose of in-
treated with human insulin and demonstrated no difference
sulin given ip results in an augmented rather than an inhib-
in IA levels based on DR4 status, whereas significant differ-
ited antibody response (96). Intraperitoneal injection of in-
sulin in mice has been found to enhance the immune ences in IA levels were observed in DR4-positive vs. DR4-
responses to multiple antigens, including insulin (76, 97, 98). negative patients treated with porcine and mixed bovine-
Oral, parenteral, and aerosol insulin administration in dia- porcine insulin (Fig. 3). This is not unexpected because the
betes-prone animal models can induce immune tolerance peptide sequences can determine the binding affinity to dif-
(see Section III.B). ferent alleles of MHC class II molecules. The IgG heavy-chain
Intraperitoneal insulin delivery via IPPs has been shown gene complex has also been shown to modulate susceptibility
to increase levels of IAs in patients with type 1 diabetes (50, to IA formation (112).
99), but not in those with type 2 (see Sections II.D and V.B.3.b) The presence of IAA at baseline may predict a greater IA
(100). Intravenous insulin delivery via IPPs (101, 102), as well response to exogenous insulin. In one study, IAA-positive
as oral insulin delivery, does not result in detectable IAs (103, individuals developed a greater IA level than IAA-negative
104). Pulmonary delivery of human insulin, as discussed patients throughout 1 yr of therapy (113). However, the
above, has been shown to be associated with higher levels of increased levels could be attributed to the sum of IAA and
IAs than sc delivery (see Section II.E) (58, 105). IA.
The findings from studies with inhaled insulin suggest
5. Dose-response effects. In humans, evidence that the devel- that patients with type 1 diabetes develop greater levels of
opment of IAs secondary to exogenous insulin is related to IAs than those with type 2 diabetes (58). Studies with insulin
dose has not been directly tested; however, clinical trial data lispro also demonstrated that patients with type 1 diabetes
show no significant relationship between insulin dose and had higher IA levels than those with type 2 diabetes (41). The
the development of antibodies. In a study of 46 patients mechanism for this difference in response is not clear, but it
newly diagnosed with type 1 diabetes, Karjalainen et al. (106) may be due to defects in regulatory T cell function, as re-
found that 35% of them were positive for IAAs before ther- ported in nonobese diabetic (NOD) mice—a model of type 1
apy. After 3 months of therapy, the IAA-positive patients had autoimmune diabetes (73). This defect may lead to enhanced
higher antibody levels than those who were initially IAA- immune responses to human insulin in patients with type 1
negative, but this difference in IA levels did not persist with diabetes (see Sections II.F.1 and V). However, it is not clear
further duration of therapy. At no time was insulin dosage
whether exogenous antibody formation can be attributed to
found to be related to the presence of IAs. Additional pro-
further expansion of autoreactive antiinsulin T cells and B
spective treatment trials of animal and human insulins, in-
cells that produce IAAs. There are conflicting data on
sulin analogs, and inhaled human insulin in insulin-naive
whether the presence of IAAs predicts the IA response to
and insulin-treated patients have shown no significant rela-
exogenous insulin (106, 113, 114).
tionship between insulin dose and level of IAs (see Section
Exogenous insulin administration may also result in an-
V.B.2.b) (22, 41, 58).
tibody formation in individuals who do not have diabetes.
6. Patient-related factors. Known genetic determinants influ- Antibody formation induced by as few as six injections of
ence susceptibility to antibody formation. The presence of human insulin is comparable to that seen in chronically
histocompatibility leukocyte antigen (HLA)-B15, -DR4, and treated patients with type 1 diabetes (115). IAAs are also
-DR7 has been shown to increase the rate of IA formation, found more commonly in individuals who have endocrine
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 631

III. Predicting Human Immunogenicity with


Animal Models
Several animal species have been used for investigations
into the immunogenicity of insulins. Antibody responses can
be generated in rabbits, mice, rats, and guinea pigs with sc
administration of human insulin and Freund’s adjuvant;
however, given the differences in amino acid sequences of
insulin between these species and humans (Table 2), these
findings are not surprising. The immunogenic potential of
insulin lispro, biosynthetic human insulin, and purified por-
cine insulin in Freund’s adjuvant were compared in rhesus
monkeys over a 6-wk period (four per dose group) (123).
Measurement of insulin-specific IgG antibody in serum by
ELISA indicated that none of the test insulin preparations
stimulated antibody formation; thus, these data indicated
that human insulin is nonimmunogenic in monkeys. Like
other species tested, however, the monkey is a poor predictor
of immunogenicity of insulins, including inhaled insulin, in
humans. In summary, it appears that animal models are poor
predictors of the immunogenicity of exogenous insulin in
humans. This is consistent with findings for other protein
therapeutic agents. Many drugs are found to be strongly
immunogenic in monkeys but weakly immunogenic in hu-
mans. Therefore, immunogenicity data from animal studies
FIG. 3. In a 1-yr trial by Fineberg et al. (83), insulin-naive patients are usually not used to predict immunogenicity in humans
received porcine insulin, mixed bovine-porcine, or rDNA human in- but only to interpret findings in the animals studied (e.g.,
sulin. Patients who were DR4-positive had greater antibody re- changes in toxicity, efficacy, and pharmacokinetics due to
sponses, in terms of percent binding, than DR4-negative subjects to antidrug antibodies) (124).
mixed bovine-porcine and porcine insulins but not for human insulin,
suggesting an interplay among HLA type, antigen structure, and However, data exist that suggest animal models may be
possibly other patient-related factors affecting antibody responses. helpful in predicting relative differences in immunogenicity
The researchers concluded that HLA type affects the magnitude of the when comparing modifications of insulin. As discussed in
immune response. Section III.B, a study was conducted in rats to compare im-
munogenic responses with different levels of insulin aggre-
gates. In another study, immunological tolerance for human
autoimmune syndromes, such as Graves’ disease, Hashimo-
insulin was induced by creating a transgenic mouse express-
to’s thyroiditis, and nonimmune thyroid disease (116).
ing the human insulin gene in the ␤-cells of the mouse pan-
Age may play an important role in the IA response with creas. This model has been used to evaluate the immuno-
exogenous insulin administration. Immunological competence genicity of human insulin analogs (125). Twelve analogs
declines as an individual ages, causing decreased ability to form having varying self-association properties were found to
high-affinity antibodies, decreased ability to generate long- have differing immunogenic potential. Substitution of single
lasting memory cells, and delayed hypersensitivity responses amino acids in the A8 –10 loop resulted in antibody responses
(117–119). The age of the individual also affects the presence of in responder mice, whereas substitutions or deletions in B3,
IAAs before the development of type 1 diabetes and whether B9, B27, and B28 did not produce antibody responses. This
an individual is more or less likely to develop a significant model may help identify less immunogenic insulin analogs
antibody response to exogenous insulin (120). in the future. Comparing antibody responses to various in-
In the studies of Fineberg et al. (41, 83, 121), the develop- sulin analogs in these mice to those in humans will be nec-
ment of significant levels of antibodies to exogenous insulin essary to validate this approach.
has been shown to be inversely related to C-peptide levels
and age. The two factors—age and C-peptide level—are in-
dependently correlated, although there is more evidence for A. Effects of inhalation of insulin
a correlation to age than to C-peptide level. A logistic re- IA responses were evaluated in rats and monkeys exposed
gression analysis of antibody development in 744 individ- daily for either 1 or 6 months to the insulin inhalation powder
uals who were insulin naive at the beginning of therapy used in Exubera (126). Exposures were up to maximally
revealed a 3% decrease in the chances of antibody develop- tolerated doses that were limited by hypoglycemia. IAs were
ment for every 1-yr increase in age (odds ratio, 0.97; 95% measured by both ELISA and radioligand binding (RLB)
confidence interval, 0.96 – 0.99; P ⬍ 0.001) (122). Additional assay in serum of rats and monkeys and bronchoalveolar
analysis showed a 46% decrease for every 1-nmol/liter in- lavage fluid in monkeys. A weak antibody response was
crease in C-peptide levels (odds ratio, 0.54; 95% confidence observed in rat serum by RLB assay only, but IAs were not
interval, 0.38 – 0.78; P ⬍ 0.001). detectable in monkey serum or bronchoalveolar lavage fluid.
632 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

A comparable weak antibody response in rats was observed RLB assay was used, and it remains the most common format
after sc injection. There were no histopathological effects of in the measurement of IAs and IAAs. Recently, filter plate
exposure in the respiratory tract or bronchial lymph nodes separation of insulin/IA complexes from unbound insulin
of either species. Specifically, no evidence was found for an has been incorporated into RLB assay protocols allowing for
exposure-related immunogenic response in the respiratory smaller sample volume requirements and higher throughput
tract. (145). In general, RLB assays for total insulin-binding capac-
The toxicity of a different dry powder formulation of in- ity involve coincubating aliquots of serum, buffer, and trace
haled insulin was evaluated in beagle dogs exposed for 13 wk amounts of monoiodinated 125I-insulin. Unbound and bound
(127). Although the researchers did not describe any IA mea- insulin in the serum is often removed before incubation by
surements, they found no effects of exposure on pulmonary using an acidification process followed by treatment with
pathological examination. dextran-activated charcoal. Nonspecific binding can be as-
The IA response in rats, monkeys, and dogs to insulin sessed by incubating a duplicate aliquot of the test serum
inhalation is consistent with findings that inhalation of harm- with a high concentration of unlabeled insulin and trace
less antigens leads to immunological tolerance. Inhaled an- levels of 125I-labeled insulin.
tigens are likely captured by intrapulmonary dendritic cells. Immune complexes are precipitated by the addition of
Under noninflammatory conditions, dendritic cells then mi- antihuman globulin, protein A/G (conjugated to sepharose
grate to draining lymph nodes and stimulate generation of beads), polyethylene glycol, or other precipitants. The pellets
T regulatory cells that down-modulate the immune response of immune complexes are separated by filtration or centrif-
to the inhaled antigen. In this manner, immune-mediated ugation, washed to remove unbound radiolabeled insulin,
inflammatory responses to reexposure to harmless antigens and measured on a gamma counter. The mean nonspecific
are avoided (128). binding disintegrations per minute (dpm) are subtracted
The effects of a nebulized solution of rDNA human insulin from total binding dpm to determine specific binding. The
were evaluated in the NOD mouse model by Harrison et al. specific binding dpm are converted to microunits or nano-
(129). Mice were exposed to either nebulized insulin or grams of IA-bound insulin, based on the specific activity of
ovalbumin, and IAs were measured using an immunopre- the labeled insulin. Data may be expressed as insulin-binding
cipitation assay. Serum IA levels from aerosol–insulin- capacity (nanograms or microunits of insulin bound per mil-
treated mice were significantly higher than in ovalbumin- liliter of serum). Other laboratories express the data as per-
treated mice. Aerosol–insulin-treated mice also experienced centage binding (counts in the pellet relative to the total
a significant delay in the onset of spontaneous diabetes. This counts of 125I- insulin added).
antibody response to an inhaled protein may be attributed in The ELISA for IAs involves adsorbing insulin to plastic
part to inadequate function or numbers of regulatory T cells wells of 96-well plates with subsequent addition of test sera.
in NOD mice (see Section II.F.6) (73). Insulin-specific antibodies in the test sera bind to the insulin
on the plate, and the remaining antibody is washed away.
B. Mucosal delivery of insulin and prevention of type 1 Anti-Ig labeled with horseradish peroxidase or alkaline
diabetes in NOD mice phosphatase is added and binds to the IA. Substrate for
horseradish peroxidase or alkaline phosphatase is added to
Most studies that have evaluated the effects of exogenous each well, and a color product is formed and monitored by
insulin on insulin-specific T cell responses have been con- measuring light absorbance. The amount of color product
ducted to investigate prevention of diabetes in NOD mice. formed increases with the amount of antibody bound to the
Exogenous insulin administration by parenteral (130 –134), plate. Data for ELISAs are often expressed as end point titers
oral (135–140), or inhalation (129) routes has been shown to (minimally detectable) or titers that produce 50% maximal
result in delay of diabetes onset and/or decreased lympho- signal.
cytic infiltration of islets. The mechanism for this protective In a few studies of serum from patients with diabetes being
effect in NOD mice appears to be immunological rather than treated with insulin, the ELISA was found to correlate with
metabolic. An inactive insulin analog (141), insulin B chain the RLB assay (99, 146 –148). However, other studies have
(142), and insulin peptide B9 –23 (143) have all been shown demonstrated a poor correlation between the two assays
to be effective in preventing diabetes with sc treatment in (149 –151). Serum samples were found to have very high
NOD mice. Regulatory T cells induced by insulin adminis- antibody levels/titers by the ELISA but very low titers when
tration are believed to attenuate the Th1-mediated insulitis using the RLB assay and vice versa. The RLB has been shown
and diabetes through local production of Th2 cytokines (e.g., to be more sensitive than the ELISA for detection of IAA.
IL-4, TGF-␤, IL-10) in the islet (129, 137, 144). Furthermore, a positive RLB assay for IAA has been found
to be a better predictor for the development of diabetes than
a similar ELISA result (152).
IV. Characterization of Insulin Antibodies The differences between the RLB and ELISA findings have
A. Measurement of insulin antibodies been attributed to inherent differences between assay for-
mats. The RLB is a solution-based assay in which low levels
Initial studies conducted to characterize IAs showed that of 125I-labeled insulin are used. Thus, high affinity IAs are
insulin/IA immune complexes do not precipitate and there- primarily measured in this assay (150, 151). In contrast, as
fore could not be measured using standard immunoprecipi- serum is progressively diluted, the ELISA reaches the stage
tation/agglutination analytical methods (6). Therefore, the of excess antigen and thus has the capacity for measuring IAs
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 633

of varying affinities (150, 151, 153). In addition, it is believed specific IgE (169), and patients with detectable antiinsulin
that the binding of insulin to solid phase (plastic wells) in the IgE have not always exhibited allergic reactions (166, 169 –
ELISA results in antibody-binding epitopes that are different 171). IgG antibodies have been associated with cases of se-
from those available with 125I-labeled insulin in solution vere insulin resistance (164, 167); however, most studies
phase. show no correlation between IAs and increasing insulin dose
Although ELISA assays can be processed rapidly and in requirements (see Section V.B.2.b). Antibodies to insulin—
larger batch sizes and do not require radiolabeled insulin, both sc and inhaled—are predominantly of the IgG class (58).
most laboratories that measure IAs use the RLB assay for the In a study of patients receiving either inhaled or sc insulin,
reasons described above. Insulin autoantibody assays are distributions of IgG subclasses were similar for both thera-
designed with high sensitivity in mind because autoanti- pies (172). In general, IgG1 levels were greater than those of
bodies are found at significantly lower concentrations than IgG4, which were slightly greater than IgG2 and IgG3 levels
are IAs that develop in response to exogenous insulin ther- in both study groups. Similarly, Füchtenbusch et al. (173)
apy; therefore, autoantibody assays may need further mod- demonstrated that after 12 months of sc insulin treatment,
ification and validation to accurately measure higher IA patients with type 1 diabetes had predominantly IgG1 and
concentrations. IgG4 antibodies to insulin and that IgG2 and IgG3 were lower
Progress has been made toward standardizing assays for in concentration. These authors also showed that IgG1 levels
the measurement of IAAs (154, 155), but there is no stan- tend to decline, whereas IgG4 levels rise with increased du-
dardization for assays designed to quantify insulin-therapy- ration of sc insulin treatment.
induced IAs, particularly when IAs are present at high con- Ig antibody class distributions resulting from inhaled in-
centrations. Because of the large volume of samples sulin therapy have been reported in two articles describing
generated in the Diabetes Prevention Trial Type 1 study, immunogenicity data for Exubera and AERx iDMS (58, 60).
micro methods were developed to analyze combined IAA Although a wide range of antibody levels were found in the
glutamic acid decarboxylase 65 and ICA512 (splice variant of Exubera studies, only IgG antibodies could be identified;
islet antigen-2 autoantigen, also referred to as IA-2A) auto- IgA, IgE, and IgM antibodies were undetectable (58). In 54
antibodies in less than 3 h (156). Even slight differences in the patients treated with AERx-iDMS, percentage binding levels
concentration of labeled insulin used, washing conditions, rose substantially from a baseline of 6 to 39% after 12 wk (60).
precipitating agent (polyethylene glycol or protein A/G This rise in antibodies was accounted for primarily by an
beads), or reagents can yield significantly different results, so increase in IgG antibodies, with small increases in IgE anti-
the absolute concentration of antibodies determined by one bodies in four patients. These data are consistent with anti-
assay cannot be assumed to have a quantitative correspon- body development reported in studies of sc insulins and
dence with the results of another. Comparison of assays used suggest that pulmonary antibody responses result from sim-
by different laboratories would require bridging studies be- ilar immunological mechanisms.
tween the two methods using a set of laboratory standards
of varying antibody levels. This has been accomplished with
C. Antibody affinity
regard to IAA, using multiple laboratory quality controls,
and expressing insulin binding as Juvenile Diabetes Foun- The RLB assay format has been used to assess relative
dation units (154). However, the lack of standardization for affinity of IAs. In the classic equilibrium-binding assay, mul-
the IA assay in patients treated with insulin hampers the tiple radioligand binding incubations are set up for a single
ability to correlate titers with clinical findings when IAs are test serum. In this way, a fixed concentration of antibody in
measured by different laboratories using different assay con- the test serum is incubated with increasing concentrations of
ditions and sometimes different assay formats. unlabeled insulin and a fixed amount of 125I-insulin. Using
higher concentrations of unlabeled insulin allows IAs with
B. Immunoglobulin classes lower binding affinities to bind the labeled insulin. The
amount of 125I-insulin bound in each tube with different
IA responses consisting of virtually all Ig classes and IgG concentrations of insulin is determined, and a Scatchard
subclasses have been reported. Insulin-specific antibodies analysis of the data has been performed in several studies by
are primarily composed of IgG1– 4 antibodies (157), but IgM, plotting the bound/free vs. bound insulin concentration.
IgA, and IgE have been reported. Antiinsulin IgM has been Using this method, Goldman et al. (84) and Brooks-Worrel et
detected during early insulin treatment (158 –160), and both al. (174) found curvilinear Scatchard plots when serum samples
Andersen (157) and Reisman et al. (160) reported the presence from patients with diabetes treated with insulin were analyzed.
of that class in patients with immunological insulin resis- Two populations of IAs were identified: high-affinity antibod-
tance. However, Patterson et al. (161) failed to detect IgM in ies with low binding capacity and low-affinity antibodies with
patients with diabetes who were treated with insulin. Faulk high binding capacity. These two subpopulations were typi-
et al. (159) reported detectable IgA in patients, and Kniker et cally described by reporting the binding affinity and binding
al. (162) associated IgA with allergic reactions in patients capacity determined from the apparent linear regions of cur-
with diabetes. vilinear Scatchard plots. The binding capacities, association
Most reports regarding allergic reactions implicate IgE constant (Ka), and calculated dissociation constant (Kd) of each
alone or in combination with IgG (see Section V.A) (163–168). of these two antibody subpopulations were determined. A
However, correlations have not always been found between mean Kd for the high-affinity and low-affinity antibody pop-
patients exhibiting allergic reactions and assays for insulin- ulations were 1.9 ⫻ 10⫺10 m and 1.6 ⫻ 10⫺6 m, respectively.
634 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

Similar findings were also observed in an earlier study in which ring polyclonal IAs. This approach has been used to evaluate
a gel-filtration method was used to measure bound and un- epitopes recognized by IAAs (185), but not by IAs.
bound insulin (175). Scatchard analysis has also been exten- Phage display libraries have also been used to investigate
sively used to evaluate the relationship between antibody af- insulin epitopes. The displayed random hexapeptide
finity and hypoglycemia (see Section V.B.3) (49, 176 –180). phagotopes recognized by IAA and IA from different sources
Concerns have been raised with regard to the use of Scat- have been identified and sequenced (186 –188), and consen-
chard analysis to determine the binding affinities and bind- sus sequences have been determined and compared against
ing capacities of polyclonal antibody responses (181, 182). the amino acid sequence of human insulin. These compari-
Antibody responses to most antigens, including insulin, are sons may lead to the identification of new immune markers
polyclonal and comprise antibodies with a wide range of of diabetes disease states based on insulin epitopes recog-
affinities that recognize different epitopes on the insulin mol- nized by IAA and may be used to identify patients for type
ecule (86). Determining antibody affinity and binding ca- 1 diabetes prevention therapy. Phagotype analysis may al-
pacity of two selected subpopulations in Scatchard analyses low IAAs to be distinguished from IAs (186).
excludes information about many other potentially relevant
subpopulations of antibodies. Scatchard analyses are best E. Characterization of insulin antibody immune complexes
suited for investigations of monoclonal antibodies to deter-
It was discovered early on that IAs do not form precipi-
mine binding affinity characteristics. When evaluating poly-
table immune complexes as observed with immune com-
clonal antibody responses, Scatchard analyses are best suited
plexes against larger proteins (6). Using ultracentrifugation
to determine the variance of affinities (181).
methods to analyze the nature of the IA immune complexes,
A simpler way to describe data from the equilibrium bind-
it was determined that insulin may bind IAs and form mono-
ing assay was reported by Heise et al. (59). Insulin-binding
mers and dimers. The dimers consisted of two IgG molecules
capacities determined from the RLB assay and expressed as
bound to one or two molecules of insulin (189). Two different
microunits of insulin bound per milliliter of serum at specific
epitopes on insulin are recognized (bivalent) and act as a
insulin concentrations were reported. Binding capacities at
bridge between two IgG molecules. Larger immune com-
corresponding insulin concentrations can be compared and
plexes are not formed. It is known that small immune com-
analyzed directly. Reporting binding data in this way is not
plexes do not activate complement (C1q binding) and are not
dependent upon applying a two-site binding model to de-
rapidly cleared (190). This is consistent with the finding that
termine the binding/dissociation constants of arbitrarily se-
IA immune complexes remain in circulation and are not
lected antibody subpopulations.
readily cleared. As discussed in Section V.B.1, IA immune
Heise et al. (59) used this reporting method to evaluate the
complexes circulate and thereby act as a “sink” for insulin in
clinical significance of low-affinity and high-affinity anti-
the circulation. It was also determined that C1q binding to
bodies in patients with type 1 diabetes treated with inhaled
immune complexes of serum samples taken from patients
insulin (Exubera). The researchers employed a parallel group
with type 1 diabetes with IAs was not greater than normal
design in which 23 patients were treated with sc insulin and
controls (191). Immune complex size may also explain the
24 patients were treated with inhaled insulin for 24 wk. Mean
rarity of immune complex hypersensitivity reactions or im-
(⫾sd) IA levels for the sc and inhaled insulin groups were
mune complex diseases in patients with high levels of IAs
4.3 ⫾ 9.4 and 101.4 ⫾ 140.4 ␮U/ml, respectively. The binding
who receive daily exogenous insulin (see Sections V.A and
capacities of samples measured with 10⫺10 m (high-affinity)
V.F).
and 10⫺8 m (low-affinity) insulin were determined. For the
Exubera group, the binding capacities of lower affinity an-
tibodies ranged from 667 to 3360 ␮U/ml and from 41 to 387 V. Clinical Significance of Insulin Antibodies
␮U/ml for the higher-affinity antibodies. The greater bind-
The incidence and severity of immunological complica-
ing capacities of the low-affinity antibodies are similar to
tions of insulin therapy have dramatically decreased with the
those reported with Scatchard analysis in previous studies
use of highly purified porcine or rDNA human insulin. Al-
with sc insulin. The binding capacities of the low-affinity and
though the new preparations still produce IAs, the titers are
high-affinity antibodies were compared with postprandial
lower, and they are rarely associated with clinical events.
glucose area under the curve (AUC), duration of insulin
This section will review previous investigations into the re-
action, hypoglycemic events, and fasting plasma glucose. No
lationships of IA and the following clinical issues: hyper-
correlation was observed between these pharmacodynamic
sensitivity reactions, hyper- or hypoglycemia, pregnancy,
markers and binding capacities of high or low affinities.
autoimmune markers, pulmonary function, immune com-
plexes, glycemic variability, and diabetes risk. It should be
D. Insulin epitope and insulin antibody idiotype analyses noted that interpretation of these investigations is often lim-
ited by uncontrolled observations, small sample sizes usually
Numerous mouse monoclonal antibodies against human
consisting of individual case reports, and nonstandardized
insulin have been generated to identify potential binding
antibody measurement methods.
sites within the insulin molecule (51, 56, 59, 183, 184). These
studies have shown that insulin administration could result A. Hypersensitivity reactions
in antibodies capable of recognizing many different epitopes.
Monoclonal antibodies have been used in competition assays Before the 1980s, local hypersensitivity reactions were
to determine the major sites recognized by naturally occur- common complications of insulin therapy. Since the advent
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 635

of highly purified insulin in clinical practice, the incidence of begin 8 to 12 h after an insulin injection and peak at 24 to 48 h.
allergic complications has markedly decreased (78). Local Most local delayed reactions are mild and self-limited. Al-
and systemic reactions with insulin administration may be though histological analyses suggest that some of these re-
mediated by insulin-specific IgE (type 1, immediate hyper- actions may be mediated by sensitized T lymphocytes (192,
sensitivity reactions) or IgG (type 3, intermediate immune– 194), it is not known how many of these delayed reactions are
complex-mediated reactions) antibodies. In addition, non- truly type 4 reactions. Occasionally, local reactions follow a
antibody-mediated type 4, delayed hypersensitivity biphasic course, with temporary improvement occurring be-
reactions have been reported. Surveys of these published tween an immediate and a delayed reaction. Type 4 hyper-
cases and reviews reveal that the exact type of local reaction sensitivity reactions may also be attributed to contaminants
is difficult to identify, and in-depth evaluation of such pa- in the older insulin preparations.
tients using skin tests and skin biopsies have not been com-
4. Allergic response to inhaled insulin. In a pooled analysis of
monly reported; thus, the frequency of insulin reactions as-
phase 2 and phase 3 trials of Exubera, no evidence was found
cribed in the literature may not be entirely accurate.
for excess adverse events of an allergic nature despite higher
Furthermore, local allergic reactions to insulin may be of
IA responses in patients who switched to Exubera-based
mixed types.
regimens (Table 4) (58).
1. Type 1 hypersensitivity. Local, immediate reactions are the
least common cutaneous insulin reaction, occurring in less B. Glycemic effects of insulin antibodies
than 1% of patients (192). These reactions begin within min-
A number of cross-sectional studies involving small num-
utes of an insulin injection, peak from 12 to 24 h later, and
bers of patients have suggested that IAs can be associated
can be followed by generalized anaphylaxis (170). Type 1
with alterations in a variety of insulin pharmacokinetic pa-
reactions can occur at any time with respect to the initiation
rameters. Some of these observed alterations might predict
of insulin, particularly when there is a history of interrupted
a clinical tendency to hyperglycemia or insulin resistance by
insulin therapy (78).
neutralizing the biological effect of circulating bioactive in-
Anaphylactic reactions are often preceded by a series of
sulin, whereas other observed alterations might predict a
local, immediate reactions (193). They are initiated by the
predisposition to clinical hypoglycemia by prolonging du-
binding of antiinsulin IgE to insulin. These complexes bind
ration of insulin action. Most clinical trial data do not show
to mast cells, releasing into the circulation a variety of va-
pharmacodynamic consequences on glucose control corre-
soactive substances that mediate the syndrome.
sponding to the pharmacokinetic observations, but case re-
Patients with generalized reactions have higher insulin-
ports of immunological insulin resistance or hypoglycemia
specific IgE levels than do patients with only local immediate
syndromes attributed to IAs continue to be published. This
reactions (194). Antiinsulin IgE levels can be 10-fold to 20-
section will first review the published pharmacokinetic stud-
fold higher in patients with allergic disease than in patients
ies, then clinical data on relationships between IAs and the
without clinical allergies (167, 195). However, the demon-
following glycemic parameters: insulin resistance, metabolic
stration of circulating antiinsulin IgE does not establish the
control, insulin dose requirements, and hypoglycemia.
diagnosis of insulin allergy because IgE can be found in
patients with no apparent allergy (see Section V.B) (171, 196). 1. Antibodies and insulin pharmacokinetics and pharmacodynam-
Patients with antiinsulin IgEs can have concomitant insulin- ics. Insulin-IA interactions like insulin-insulin receptor in-
specific IgG antibodies (88, 167). teractions are reversible and follow the principles of equi-
librium binding. Thus, the amount of insulin bound is
2. Type 3 hypersensitivity. Local, intermediate reactions appear dependent on antibody affinity, insulin concentration, and
4 to 8 h after insulin injections, peak at 12 h, and generally antibody concentration (insulin-binding capacity). Hypo-
subside within a day (197). These phenomena are believed to thetically, the amount and time course of bioavailable insulin
be a vasculitic response to soluble antigen-antibody com- in plasma (insulin pharmacokinetics) could be influenced by
plexes (Arthus reactions) (192, 194, 198). Lipoatrophy— or IAs and may be mediated by antibody affinity and binding
loss of fat at insulin injection sites—may represent a persis-
tent, localized Arthus reaction. In biopsies from affected TABLE 4. All-causality adverse events of an allergic nature in
sites, IgM, IgE, and C3 have been demonstrated in dermal controlled phase 2 and phase 3 Exubera studies
vessel walls (199). Lipoatrophy was found to occur in up to
Inhaled Subcutaneous
20% of patients before the wide availability of highly purified insulin insulin
forms of insulin (197), but it has become virtually nonexistent (n ⫽ 920) (n ⫽ 602)
in patients treated with purified porcine or human insulin. Subjects’ months of exposure 3791 2939
It is possible that lipoatrophy is mediated by contaminant- Body as a whole (includes potential 40 (4.3) 33 (5.5)
specific immune responses rather than by IAs. In recent injection site reactions and
allergic reactions)
years, the more commonly observed lipohypertrophy at in- Lipodystrophy 4 (0.4) 4 (0.7)
sulin injection sites has been linked to immune responses to Respiratory (includes asthma) 107 (11.6) 81 (13.5)
insulin as well (200). Skin and appendages (includes 64 (7.0) 28 (4.7)
nonspecific rashes and
3. Type 4 hypersensitivity. Local, delayed reactions are the dermatitis)
most common hypersensitivity reaction and usually occur at Data represent number of adverse events (%). Reprinted with per-
the start of insulin therapy (192, 198, 201, 202). They generally mission of the Journal of Clinical Endocrinology & Metabolism (58).
636 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

capacity. However, evaluating the pharmacokinetic effects of effect of IAs on postprandial insulin and glucose was largely
IAs presents several challenges, not the least of which is accounted for by the association constant of the high-affinity
validation of the accurate and reproducible measurement of IA binding sites (K1). Peak postprandial glucose was 237 ⫾
insulin concentrations in the presence of IAs (203, 204). Free 10 mg/dl in patients in the upper quartile, compared with
insulin results may not always be true reflections of bioactive 166 ⫾ 12 mg/dl in patients in the lowest association constant
insulin in the presence of IAs (205). quartile.
With an increase in IA binding, there is an apparent in- In contrast, a prospective, open-label, parallel-group trial
crease in the volume of distribution of insulin (206, 207). By of 47 patients with type 1 diabetes randomized to receive
acting as a “sink” for exogenously administered insulin, an inhaled insulin (Exubera) or sc regular human insulin, was
increase in the apparent volume of distribution of free insulin designed to evaluate whether IA development with inhaled
related to IAs could in theory result in alterations in the insulin is associated with the loss of postprandial glucose
disposition kinetics of free insulin. control (59). Mean IA levels increased from baseline after
It has been reported that patients with elevated levels of inhaled insulin treatment but not after sc regular insulin
IAs or antibodies with higher binding capacity experience treatment. The researchers found no significant differences in
reduced initial rates of increase and delayed time to peak (120 postprandial plasma glucose profiles between treatment
to 180 min vs. 90 min), and prolonged return to baseline of arms, and no correlation between postprandial blood glucose
plasma-free insulin levels after sc insulin injection in com- exposure and antibody-binding affinity was apparent.
parison with antibody-negative patients or patients with A body of literature suggests antibody-mediated prolon-
lower antibody-binding capacity (207–210). Although time to gation of elevated insulin levels after sc injection (206 –208,
peak insulin level after administration of insulin appears to 211–213); however, pharmacodynamic consequences of this
be delayed in most reports, the absolute magnitude of the finding have not been consistently demonstrated. Impor-
peak does not appear to be markedly different between high tantly, increased rates of hypoglycemia have not been con-
and low levels of IAs (178, 208, 210). The presence of IAs has firmed in clinical trials (see Section V.B.3.d). Although IAs
also been associated with lower maximal (Cmax) free insulin may not cause hypoglycemia, Bolli et al. (212, 213) published
concentrations and AUCs after sc injection of NPH insulin two studies that demonstrated prolonged recovery times
and iv infusion of porcine insulin (178, 209). Prolonged half- from experimentally induced hypoglycemia in patients with
life (up to 8-fold greater), increased distribution space (up to IAs. The researchers concluded that patients with insulin-
10-fold greater), and faster metabolic clearance rates have dependent diabetes can have impaired glucose counterregu-
been documented after iv infusion of recombinant human latory hormone reserve, which can be compounded by a
and/or bovine insulin in patients with type 1 diabetes (206, prolongation of the half-life of insulin by IAs. To explain such
207, 211). Alterations in pharmacokinetic parameters ap- an IA-mediated phenomenon mechanistically, one must pos-
peared to be correlated with various measures of antibody tulate dissociation of free insulin from antibody bound in-
characteristics, such as insulin-binding affinity, insulin-bind- sulin complexes (see Section V.B.3.d).
ing capacity, or percentage of insulin binding. IAs have not consistently been shown to have the property
Waldhausl et al. (211) found heterogeneous total insulin of prolonging the duration of insulin action. Euglycemic
responses to iv insulin administration in subjects with more clamp studies have been conducted to explore the effect of
than 25 ␮g/liter insulin-binding capacity. Their data suggest IAs on the pharmacodynamic response to exogenously ad-
a shift in the time course of the plasma free insulin profile but ministered insulin. In one study, glucose infusion rates did
not in the overall extent of the plasma free insulin exposure. not vary between subjects with high and low insulin-binding
Measurements of the pharmacodynamic responses to in- capacity but were lower than in subjects without diabetes
sulin are more clinically relevant than pharmacokinetic re- (lower glucose infusion rates indicate decreased insulin ac-
sponses and not subject to questions regarding the validity tion) (211).
of insulin measurements in the presence of IAs. Higher post- Peters et al. (209) noted that exposure (AUC) to free insulin
prandial plasma glucose concentrations in subjects with IAs and glucose infusion rates were lower in patients with
have been described in two pharmacodynamic studies (208, greater than 10% antibody binding compared with subjects
210). In the first report (208), the effects of IAs appeared to with less than 1.5% antibody binding. Gardner et al. (214)
vary with the insulin species under study. Postprandial found no correlation between antibody status and the onset
plasma glucose concentrations after injection with rDNA of action of insulin administered sc. Furthermore, the peak
human insulin did not differ among patients with low (⬍10 effects of insulin action as well as the duration of insulin
U/liter) or moderate (⬎10 U/liter) insulin-binding capacity, action were similar both in patients who were antibody pos-
but postprandial plasma glucose concentrations were sig- itive and in those who were antibody negative. Similarly,
nificantly greater in the moderate binding group when bo- Heise et al. (59) were not able to detect a significant difference
vine insulin was administered. in duration of inhaled insulin action in subjects with or with-
In the second report, Van Haeften et al. (210) found no out IAs. They reported no correlation between IA-binding
differences in postprandial insulin or glucose when compar- affinity and the duration of insulin action.
ing porcine to human insulin injections, although postpran- It seems possible that pharmacokinetic differences attrib-
dial glucose excursions in response to a standard meal did utable to IAs may be observed, but the effects are small in the
appear correlated with the IA binding. Correspondingly, context of the myriad of other factors that influence glycemic
increases in plasma free insulin levels after injections of both response. The inconsistency between pharmacokinetic and
insulins were negatively correlated with IA binding. The pharmacodynamic observations may lie in the incomplete
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 637

understanding of the in vivo conditions that control insulin creasing concentrations of IgG can inhibit IgE-mediated clin-
bioactivity in the presence of IAs. This is further complicated ical events (20, 167, 219). Although the dominant feature of
by the imprecise relationship between insulin pharmacoki- this syndrome is decreased insulin action, episodes of hy-
netics and pharmacodynamics that exists in the absence of poglycemia can occur. Furthermore, the frequency of hypo-
IAs (215, 216). glycemia may diminish after resolution of the insulin-resis-
tant state (218).
2. IAs and hyperglycemia/interference with insulin action
Immunological insulin resistance is thought to occur less
a. Immunological insulin resistance. IAs are determined to be commonly now than it did before the 1980s, but cases of at
present when binding of labeled insulin is demonstrated in least partial resistance attributed to IAs continue to be re-
vitro. Whether in vitro binding quantitatively reflects the ported— even in patients treated only with human insulin or
binding of circulating insulin in vivo is difficult to directly insulin analogs (30, 164, 220 –227). Severe insulin resistance
determine (see Section V.B.1). If antibody binding did occur has also rarely been described in the setting of high levels of
in vivo to significant levels, one might expect to observe serum insulin-binding activity associated with underlying
increasing insulin dose requirements and, possibly, worsen- chronic lymphocytic leukemia, lymphoma, multiple my-
ing glycemic control in patients with diabetes. Although a eloma, and macroglobulinemia (228). Possibly, the severe
rare syndrome of severe insulin resistance has been de- resistance in these cases was due at least in part to the pro-
scribed in patients with high IA levels, a mechanism for a duction of monoclonal paraproteins with significant insulin-
causal relationship between the antibodies and the syndrome binding activity, although monoclonal insulin binding was
has not been clearly established. Furthermore, IAs do not demonstrated in only two cases. Insulin resistance occurring
correlate with measures of glycemic control or insulin dose in patients with hematological malignancy is not always
requirements in most large population studies. associated with an insulin-binding monoclonal protein (87).
Insulin resistance can be defined as a daily insulin dose
requirement that exceeds the normal daily pancreatic output b. Treatment of immunological insulin resistance. Because im-
in the nondiabetic state, i.e., approximately 40 ⫾ 20 U/d mune insulin resistance tends to be self-limited (50% of pa-
(217). Severe insulin resistance is usually defined as insulin tients are insulin resistant for ⬍ 6 months and 75% for ⬍ 1
requirement of more than 200 U/d for at least 2 d (14, 217, yr), therapy is aimed at speeding a remission while avoiding
218). The incidence of insulin resistance in patients with metabolic complications (14). The initial treatment for severe
diabetes was estimated to be 0.1% in a study at the Joslin insulin resistance has generally been to switch therapy to
Clinic between 1940 and 1960 (14). However, this incidence alternative insulin formulations. Before the advent of human
was acknowledged as likely to represent an overestimate insulin in clinical practice, such patients were successfully
because of referral bias, and IA measurements were not treated by switching to purified porcine insulin (20, 229), fish
reported. insulin (219, 230), and modified insulins, including sulfated
In a subsequent case series, Davidson and DeBra (218) insulin (218) and desalanine-porcine insulin (231). The ben-
characterized 35 patients with severe insulin resistance as- eficial effect on immunological insulin resistance of sulfated
sociated with the presence of high levels of circulating IAs. insulin may have been mediated by CD8⫹ T cells (232).
Patients studied in this series had mean daily insulin re- In patients with type 2 diabetes, insulin cessation may be
quirement of 550 U/d (range, 200 to 2000 U/d) and maxi- a successful treatment for immunological insulin resistance
mum insulin-binding capacity greater than 10,000 ␮U/ml. In (221, 233). Switching insulin-resistant patients to human in-
addition, the following underlying explanations for large sulin (87) or insulin lispro (220, 224) or from U-100 to U-500
insulin requirements were ruled out: diabetic ketoacidosis, insulin has also been successful (234). When switching in-
significant infection, significant dietary indiscretion, li- sulin formulations in patients with immune-mediated insu-
poatrophic diabetes, significant complicating endocrine dis- lin resistance, substantially lower doses may be necessary to
ease, insulin receptor defects, antibodies to insulin receptors, avoid hypoglycemia (217). In a case report, endogenous in-
or factitious claim of insulin dose greater than 200 U/d. sulin stimulated by tolbutamide was found to be effective in
At least 60% of patients diagnosed with immunological a patient with type 2 diabetes who was felt to be unrespon-
insulin resistance had a history of diabetes with onset after sive to exogenous insulin (235).
age 30 yr, suggesting that the syndrome is more commonly Glucocorticoids are a second-line therapy for patients with
observed in patients with type 2 diabetes (20, 218). The onset nonremitting immunological insulin resistance who do not
and subsequent course of insulin resistance is often associ- respond adequately to switching insulin formulations (14, 15,
ated with symptomatic hyperglycemia, including episodes 20, 236), or in whom ketoacidosis ensues (79). Prednisone at
of ketoacidosis and hyperosmolar coma (217, 218). The du- doses of 60 to 80 mg/d can result in lower insulin require-
ration of insulin therapy before the onset of severe insulin ments in up to 75% of patients, often within a few days and
resistance has been reported to range from 1 month to 15 yr even before declines in antibody levels (20). Responses in
(217, 218), although 50 – 85% of patients with insulin resis- some cases can be dramatic, and severe hypoglycemia can
tance received insulin for less than 1 yr, and 10 –25% received ensue (14, 217, 236), leading some to recommend that glu-
insulin for less than 1 month before the onset of severe insulin cocorticoid therapy be administered in a hospital or other
resistance (217, 218). Insulin allergy may coexist with im- monitored setting (217). Prednisone doses should be tapered
mune insulin resistance in 10 –35% of cases (20, 217). Some when there is a clinical response. Often, 2 to 3 wk of glu-
cases of insulin resistance that occurred as insulin allergy cocorticoid therapy is adequate to achieve a sustained re-
spontaneously subsided, prompting speculation that in- sponse (20, 217). In rare cases, plasmapheresis (223, 237) or
638 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

cytotoxic immunosuppressive agents, such as 6-mercapto- higher average glucose in populations of patients (246 –248),
purine (238), have been used for immune insulin resistance but most showed no correlation between IA and glucose
not related to an underlying paraproteinemia. control— usually measured as glycated hemoglobin (19, 58,
Although high levels of IAs have been demonstrated in 99, 114, 241, 249 –265). Most studies published since 1980
patients with syndromes of severe insulin resistance, it is have shown no evidence for a significant relationship be-
difficult to arrive at a causal relationship between the anti- tween IA and average glycemia (58, 99, 114, 241, 250 –256,
bodies and the syndrome with available data. Insulin dose 259 –263, 265).
requirements are not predicted by insulin-binding capacity Researchers have postulated that IAs can be associated
measurements, even in patients with severe insulin resis- with mild degrees of insulin resistance, which would be
tance (20, 218). Furthermore, there is substantial overlap in detected clinically as mild to moderate increases in insulin
antibody levels between resistant and nonresistant patients dose requirements. Additionally, some studies have sug-
(218). An approximate maximum antibody-binding thresh- gested that increasing insulin dose requirements correlate
old of 30,000 ␮U/ml, above which patients may be at risk for with IA levels (249, 264, 266 –268); however, most studies
this syndrome, has been suggested; however, 15% of patients have showed no relationship or correlations between IAs and
with the syndrome described in the literature have values decreasing dose requirements (58, 99, 114, 241, 247, 250, 252,
below this range (217, 218). 255, 259, 260, 262, 265, 266, 269 –273). Evidence suggests that
If an antibody threshold exists, one still needs to postulate IA-positive patients who switch insulins to a less immuno-
that there is an undiscovered attribute of the antibodies or genic preparation can experience reduced dose requirements
related cofactors that determines why some patients develop in concert with declining IA levels (27, 264, 274, 275), or that
insulin resistance, whereas other patients with comparable switching insulin preparations can result in declining IA
antibody levels do not. Subpopulations of antibodies iden- levels without decreased insulin dose requirements (19, 251,
tified by insulin affinity have been correlated with clinical 263, 276 –280).
resistance and response to therapy in case reports (223, 239, 3. IAs and hypoglycemia. Rare syndromes in which recurrent
240). Both high-affinity (221) and low-affinity (240) antibod- or prolonged hypoglycemia is the dominant feature have
ies have been identified as correlating best with clinical been attributed causally to IAs. Most frequently, this situa-
findings. tion is encountered in insulin autoimmune syndrome (IAS;
IA levels clearly do not by themselves explain why some also called Hirata’s disease), in which nondiabetic patients
patients with this syndrome require 200 U/d and others with no history of insulin exposure spontaneously develop
require 2000 U/d (241). Furthermore, the mechanism by IAAs and hypoglycemia. Evidence that IAs induced by ex-
which IAs would cause insulin resistance is not clear. IAs ogenous insulin therapy can also cause hypoglycemia is lim-
could function as a circulating binding reservoir for insulin; ited to case reports. Although hypoglycemia is the most
however, in theory, once this reservoir is saturated, normal notable feature of these syndromes, some affected patients
free insulin levels should be achievable with typical insulin have also been reported to have clinical evidence of atten-
doses (20, 217, 241, 242). uated insulin action (including severe insulin resistance). It
Cyclical expression of important idiotypic determinants— remains to be determined whether antibodies associated
despite unchanged levels of total insulin binding— could with pathological hypoglycemia can be distinguished in vitro
explain clinical syndromes that occur and remit spontane- from the far more commonly occurring IAs that are not
ously. Although such cyclic idiotypic expression has been linked to clinical hypoglycemia.
described in one report (243), it is not clear that this phe-
nomenon involves the insulin-receptor-binding epitope, a. Hirata’s disease. IAS consists of high levels of IAs with or
which would be necessary to explain the occurrence and without concomitant Graves’ disease associated with fasting
remission of immunological insulin resistance. Rarely, insu- hypoglycemia in insulin-naive patients. The disease was first
lin resistance is associated with antiinsulin receptor antibod- described in 1970 by Hirata et al. (281). This disorder is
ies (244). Most patients with insulin resistance due to insulin HLA-linked (282) and is the third most common cause of
receptor antibodies have evidence for systemic autoimmu- hypoglycemia in Japan, but has been sporadically reported
nity to noninsulin autoantigens (244), unlike the immune outside Japan (283, 284).
response to exogenous insulin in patients with diabetes (245). In most patients, remission occurs within 6 months; how-
Modern definitions of immunological insulin resistance re- ever, life-threatening hypoglycemia may necessitate mea-
quire excluding the presence of insulin receptor antibodies sures such as plasmapheresis (285, 286). Impairment in glu-
(218). cose tolerance is reported for some of these patients (283).
Patients with IAS often have late postprandial hypoglyce-
c. Overall metabolic control and insulin dose requirements. mia, possibly due to late release of endogenous insulin from
Some pharmacodynamic studies that examined the relation- the autoantibody pool (286). It is also possible that some
ship between IAs and postprandial glycemia have suggested instances of hypoglycemia associated with IAS can be at-
that IAs can be associated with relative hyperglycemia after tributed to the development of antiidiotypic antibodies that
meals (49, 208, 210) (see Section V.B.1). A prospective study have insulin agonistic properties. Among white patients in
evaluating postprandial glucose tolerance during the devel- the United States, IAS manifests primarily as postprandial
opment of IAs with inhaled insulin therapy showed no loss hypoglycemia; as in Japanese patients, the syndrome can be
of postprandial glucose control (59). associated with polyclonal or monoclonal IgG insulin-bind-
Some investigators have reported IA levels correlated with ing antibodies (287).
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 639

b. Hypoglycemia and antibodies to exogenous insulin. Pub- although one patient remained symptomatic despite a de-
lished case reports have attributed unusually prolonged ep- cline in antibody level to 84 ␮U/ml while still receiving ip
isodes of hypoglycemia to high levels of antibodies to ex- insulin therapy.
ogenous animal insulin (176, 178, 288, 289). The first case,
d. Clinical trials and limitations of literature. Although infor-
reported in 1960 by Harwood (288), described a 44-yr-old
mation from case reports suggests that high levels of IAs can
woman with type 1 diabetes who for 9 yr experienced pe-
be associated with prolonged periods of hypoglycemia, stud-
riods in which she would need to discontinue insulin therapy
ies of large populations have failed to establish a relationship
for up to 23 d because of prolonged hypoglycemia. She was
between IAs and hypoglycemia event rates (58, 59, 251, 259,
found to have 106,000 ␮U/ml insulin-binding capacity with
263, 265, 296, 297). In a pooled analysis of phase 2 and phase
an unusually slow rate of dissociation of the antibody-anti-
3 trials involving more than 350 patients with type 1 diabetes
gen complex.
and more than 400 patients with type 2 diabetes treated with
Seven case reports have been published of hypoglycemia
inhaled insulin (Exubera), no relationships between hypo-
occurring in patients with antibodies found in the setting of
glycemic event rates and IA levels were observed (58).
human insulin therapy (223, 239, 290 –293). Interestingly,
Published case reports describe patients with unusual hy-
these cases were all reported from Japan, where IAS is
poglycemia syndromes who are found to have very high
thought to be more prevalent than in the rest of the world.
levels of IAs. Because of an inherent reporting bias, however,
These cases also occurred predominantly in patients over 70
it is difficult to determine from the literature how many
yr of age, as is the case with IAS.
patients with high antibody levels do not have hypoglyce-
Hypoglycemia has also been reported in up to 54% of
mia. Likewise, it is difficult to know the frequency of similar
patients receiving a combined pancreas and kidney trans-
syndromes in patients who have low IA levels.
plant (294). Tran et al. (295) compared patients who had
Although both hypoglycemia and IAs are common in pa-
repeated episodes of hypoglycemia or hypoglycemic symp-
tients receiving insulin, a causal relationship between the
toms after a pancreatic transplant matched with patients who
two has been difficult to demonstrate. For the presence of
did not have hypoglycemia after pancreatic transplant. They
antibodies to explain hypoglycemia in such patients, it must
found a decrease in the ratio of fasting free insulin to total
be presumed that antibodies first bind the insulin in circu-
insulin in the patients who had a hyperglycemic response to
lation and then later dissociate from the insulin, allowing
a liquid meal challenge. This subgroup had a substantial
activation of cellular insulin receptors. Despite multiple at-
increase in total, but not free, insulin concentrations. Al-
tempts to characterize antibodies associated with hypogly-
though IAs were not directly measured, these data are con-
cemia syndromes, it is not clear that consistent affinity/
sistent with the hypothesis that hypoglycemia was associ-
binding capacity profiles can distinguish antibodies in
ated with high levels of circulating IAs (295).
patients with and without hypoglycemia syndromes (49,
Although anecdotal information from these case reports
179).
suggests that very high levels of IAs can be associated with
However, recent studies have reported potential progress
unusual hypoglycemia syndromes, studies of large popula-
toward identifying in vitro characteristics of IAs associated
tions have failed to establish a relationship between IAs and
with clinical hypoglycemia syndromes in small numbers of
hypoglycemia event rates (58, 59, 251, 259, 263, 265, 296, 297).
patients (177, 180, 222, 223, 298). One of those studies has
c. Hypoglycemia in IIP patients. Intraperitoneal insulin de- suggested that insulin-receiving individuals with high anti-
livery via IIPs has been associated with high levels of IAs in body levels and recurrent hypoglycemia have a higher dis-
40% of patients with type 1 diabetes, and these antibody sociation constant for insulin measured by surface plasmon
levels persist for at least 3 yr (see Sections II.D, II.F.3, and resonance than monoclonal antibodies to human insulin
II.F.4) (50). Lassmann-Vague et al. (99) found no correlations (298). It remains to be seen whether these recently proposed
between IAs and hypoglycemic event rates experienced by assays correlate better with symptoms than with standard IA
24 patients who had been treated for 2 yr with ip insulin measurements.
therapy. IA levels also did not correlate with glycated he- It is also not clear what factors may precipitate the hy-
moglobin, insulin requirements, or free insulin levels in these pothesized unregulated dissociation of antibody-insulin
patients. In a contrasting study, Jeandidier et al. (49) tracked complexes. Because some patients are reported to have noc-
IA response in 62 patients with type 1 diabetes on ip pump turnal hypoglycemia, researchers often presume that bind-
therapy for 2 yr. Although an association between IA status ing equilibrium is shifted toward antibody-insulin dissoci-
and the number of low blood glucose values per month was ation when free insulin concentrations decline as time
reported, severe hypoglycemic event rates dropped dramat- elapses overnight from evening insulin administration.
ically in patients implanted with ip pumps regardless of IA However, were this the case, a correlation between IA and
responses. lower fasting glucose measured in the morning would be
The possibility that nocturnal hypoglycemia could be re- expected, and no such relationships have been demonstrated
lated to the antibody response— despite downward titra- (58, 59, 251, 256).
tions of overnight basal insulin infusion—was first raised by Clinical trials comparing inhalation with sc delivery of
Olsen et al. (50), who described four of 25 patients using IIPs insulin in patients with type 1 or type 2 diabetes have dem-
with unexplained significant decreases in overnight basal onstrated reduced fasting plasma glucose levels by as much
insulin dose requirements. All four of these patients had peak as 40 mg/dl associated with inhaled insulin, the mechanism
IA levels above 200 ␮U/ml (range, 205 to 1021 ␮U/ml), for which is unknown (299). However, IAs were found not
640 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

to correlate with these fasting glucose levels (58) (Fig. 4, from control in the patients studied. Maternal glycemic control is
Pfizer data on file). This is consistent with the observation important because it is known to influence many fetal and
that treatment group differences in fasting plasma glucose neonatal risks, including neonatal hypoglycemia and mac-
are fairly consistent in patients with type 1 or type 2 diabetes, rosomia (307). Most notably, despite the strong association
whereas IA responses to inhaled insulin are significantly between neonatal hypoglycemia and increased birth weight
higher in patients with type 1 compared with patients with in the infant of a diabetic mother (307), no consistent linkage
type 2 diabetes (58). was found between IAs and birth weight, even in the early
To be clinically significant, prolonged dissociation of IA studies that linked IAs to neonatal hypoglycemia. Further-
complexes should result in a measurable prolongation of the more, hypoglycemia remains a common complication in ne-
duration of insulin action. This hypothesis was examined in onates born to mothers with diabetes, despite the fact that
a prospective 6-month pharmacodynamic study in which insulin preparations of low immunogenicity are now in rou-
duration of insulin action was measured by 12-h euglycemic tine use (22, 308).
clamps (59). This study showed that in patients with IA The potential for increased incidences of neonatal hypo-
responses to inhaled insulin (Exubera), duration of insulin glycemia, respiratory distress syndrome, and hypocalcemia
action was unchanged, despite a rise in IA levels, and no has also been suggested in small studies published from 1980
differences were found between these patients and those to 1990 (304, 305, 309, 310); however, these reports also lacked
treated with sc insulin. Furthermore, there were no correla- adequate documentation of maternal glycemic control. Sub-
tions between high-affinity or low-affinity insulin binding on sequent studies, performed in the modern era of prenatal
duration of insulin action, fasting glucose, or clinical hypo- care for women with diabetes, argue against a connection
glycemic events (see Section V.B.1). between IAs and fetal morbidities (254, 311). Additionally,
Clinical trial data suggest that hypoglycemia does not multiple studies have failed to show a relationship between
correlate with IAs in large populations of patients. Never- maternal IAs and birth weight. A randomized trial compar-
theless, rare patients with high antibody levels may develop ing human insulin with animal insulin during pregnancy
hypoglycemia syndromes owing in part to prolonged or found that improved glycemic control, not IAs, influenced
unregulated appearance of free insulin in the circulation infant birth weight (312). Wellik et al. (313) showed no cor-
(178). Because the levels of IAs do not in themselves account
relation between IAs and neonatal glucose level or birth
for recurrent hypoglycemia, it seems likely that clinical hy-
weight. Three recent studies with substantially larger sample
poglycemia in this setting would have a multifactorial eti-
sizes reported no relationships between IAs and birth weight
ology, perhaps involving deficient glucose counterregula-
(314 –316). The most recent report found similar birth
tion coupled with a prolonged half-life of free insulin (213,
weights in offspring of 138 mothers with type 1 diabetes
300).
across a range of maternal IA levels and cord blood insulin
levels (314). Islet autoantibody concentrations also were
C. Pregnancy found to have no influence on birth weight.
Maternofetal IgG transfer begins early in the second tri- Two hypotheses have been put forward to explain a pos-
mester, with most antibodies transferring to the fetus during sible relationship between maternal IAs and neonatal hypo-
the third trimester (301). Because organogenesis occurs dur- glycemia (79), both of which would predict clear correlations
ing the first trimester, a lack of correlation between congen- between IA levels, birth weight, and neonatal hypoglycemia
ital malformations and maternal IAs is not surprising (302). risk. In the first, maternally derived antibody interference
Recently, it has been shown that transmission of maternal with insulin action in the fetal circulation could result in
antibodies to exogenous insulin does not affect diabetes risk compensatory fetal hyperinsulinemia. In this scenario, fetal
in offspring (303). hyperinsulinemia results in increased birth weight and neo-
Pregnancy-related risks from IAs have not been clearly natal hypoglycemia. Although neonatal cord blood C-pep-
demonstrated. Some early papers reported associations be- tide levels (reflecting endogenous insulin secretion) were
tween IAs and neonatal hypoglycemia (304 –306). However, found to correlate with IAs in one study (305), the finding
these papers did not adequately describe maternal glycemic was not reproducible (302, 315). Recently, no relationship

FIG. 4. In a long-term phase 3 study (data on file, Pfizer


Inc.), no correlation was found between fasting plasma
glucose and IA levels in patients with type 1 diabetes
(n ⫽ 252) treated with inhaled human insulin (Exubera)
at 24 months (Spearman correlation coefficient,
⫺0.073).
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 641

was found between cord blood insulin levels and birth F. Immune complexes
weight (314). Furthermore, multiple studies, including the
A series of reports published from the 1960s through the
only randomized prospective study, have found no relation-
1980s yielded conflicting results regarding insulin-antiinsu-
ship between IAs and fetal birth weight (312).
lin immune complexes in the development of long-term di-
A second hypothesis suggests that insulin is transferred to
abetic complications in animal models (319). Studies in hu-
the fetus via IA complexes. These complexes could then
mans have not shown consistent links between IAs and
dissociate in the fetal circulation, releasing bioactive insulin.
diabetic complications. Although some studies have shown
Were this to happen during fetal life, increased birth weight
increased levels of immune complex formation in patients
would be expected. As discussed, multiple studies have
with diabetic microangiopathic complications compared
failed to show relationships between maternal IAs and mac-
with patients without complications (320 –322), these im-
rosomia. If IA complex dissociation occurred after birth, un-
mune complexes often do not contain insulin or IA (321–324).
usually prolonged neonatal hypoglycemia syndromes might
Furthermore, insulin administration does not contribute to
be expected, given that the biological half-life of circulating
this immune complex formation (325). The nonspecific im-
IgG is approximately 23 d (317). No distinguishing clinical
mune complexes observed in patients with diabetes may
characteristics of neonatal hypoglycemia associated with
reflect general inflammatory reactions associated with an-
IAs, such as unusual prolongation, have been described in
giopathies (321).
babies born to mothers with IAs.
Overall, no direct evidence has shown that immune com-
plexes, insulin immune complexes, or IAs are capable of
D. Autoimmune diseases mediating vascular or glomerular damage. Associations
have been reported, but they have been largely based on the
Antibody responses to exogenous insulin have not been prevalence of immune complex detectability rather than on
shown to cause generalized immune activation resulting in quantitative measurements of the amount of immune com-
autoimmune disease states. Lassmann-Vague et al. (245) plex present (326). Andersen (327) showed that high porcine
measured a panel of autoantibodies before implantation with insulin-binding levels were more frequent in patients with
pumps for ip insulin delivery and then subsequently every complications from diabetes than in patients without com-
year in 28 patients with type 1 diabetes. At baseline, 19 of 28 plications but found no significant differences in mean an-
patients were negative for all tested autoantibodies (antithy- tibody levels between groups. Virella et al. (328) found cor-
roglobulin, antithyroperoxidase, gastric parietal cell, smooth relations between the presence or absence of detectable
muscle, mitochondrial, liver-kidney microsome, antinuclear, insulin-antiinsulin immune complexes and the presence or
antiendomysium, and antigliadin antibodies). During 2 yr of absence of some diabetic complications.
ip insulin treatment, the sera of the patients negative at Although immune complexes in insulin-treated patients
baseline remained negative throughout the study, despite have been associated with procoagulant markers in some
the expected IA response (see Sections II.D and V.B.2.c). Nine studies (323, 329, 330), the majority of studies have shown no
patients with preexisting autoantibodies had no change in relationship between IA and diabetic angiopathic complica-
most autoantibody titers. Two of these patients had increases tions. Specifically, no relation has been found between IAs
in antithyroperoxidase titers, whereas three patients had de- and histological findings of nephropathy (331), muscle base-
creases in these titers. No difference was seen in IA responses ment membrane thickness (332), clinical nephropathy (114,
to ip insulin delivery in the nine patients with preexisting 333, 334), clinical retinopathy (114, 273, 321–323, 332–336),
autoantibodies relative to those who did not have preexisting clinical autonomic neuropathy (337), clinical peripheral neu-
antibodies. ropathy (338), or complement activation in patients with
diabetes (339).
The absence of a pathogenic effect may be related to the
E. Inhaled insulin antibodies and pulmonary function properties of the immune complex. It has been observed that
insulin and IA immune complexes do not precipitate (at
Inhaled human insulin is associated with higher levels of
antigen or antibody excess) and cannot be detected by stan-
IAs compared with sc human insulin (see Section II.E). In-
dard immunodiffusion methods (340). In addition, insulin-
haled insulin (Exubera) is also associated with small treat-
insulin–antibody immune complexes are monomers or
ment group differences in pulmonary function tests, which
dimers, and large immune complexes are not formed. Small
develop early after treatment initiation, are not associated
immune complexes do not readily bind to C1q complement
with clinical sequelae, are nonprogressive with up to 2 yr of
and are, therefore, not readily cleared by the reticuloendo-
therapy, and are reversible with treatment discontinuation
thelial system of the liver or spleen (190). Small immune
(63, 318). This pulmonary function test change does not,
complexes have a similar half-life to circulating Ig molecules,
however, appear to be mediated by the immunological re-
which explains the high levels of circulating total insulin
sponse to inhaled insulin. Teeter and Riese (63) showed that
(antibody bound) in patients with high levels of IAs (see
the treatment group difference in forced expiratory volume
Section IV.E).
in 1 sec (FEV1) is manifest as early as 2 wk after inhaled
insulin initiation, at which time the mean IA response has not G. Buffering effect of insulin antibodies
yet evolved. Furthermore, Fineberg et al. (58) showed no
correlation between IA levels and FEV1 in pooled analyses of Both glycemic stability and instability have been attrib-
inhaled insulin (Exubera) clinical trials. uted to the presence of IAs. Instability has been described
642 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

only in case reports and is characterized either by both hy- T cell clones have been generated from the draining pan-
perglycemia and hypoglycemia occurring within a single creatic lymph nodes of patients with type 1 diabetes that
24-h interval or by periods lasting days to weeks of hyper- were found to be responsive to insulin (356). These data
glycemia alternating with periods of hypoglycemia of similar indicate that insulin may be one of the initial antigens rec-
duration (49, 176, 178, 223, 229, 239, 290, 291, 341, 342). Some ognized by autoreactive T cells before the onset of clinical
of these case reports describe patients with relative overnight type 1 diabetes and exogenous insulin treatment.
hypoglycemia and daytime hyperglycemia, although there is A few studies have evaluated insulin-specific T cell re-
no clear mechanistic explanation for such diurnal patterns sponses in patients with diabetes being treated with insulin.
(see Section V.B.3.c). Because these glycemic instability syn- The most commonly used assay to measure insulin-
dromes are rare and variable in nature and because in vitro responsive T cells involves coincubating peripheral blood
parameters are not predictive of clinical findings (see Section mononuclear cells with or without insulin for 6 to 10 d. The
V.B), it is difficult to establish cause and effect relationships proliferative response is measured by the incorporation of
between the antibodies and such glucose variability radio-labeled nucleotide (357). T cell responses in patients
syndromes. with type 1 diabetes treated with porcine and bovine insulin
Conversely, because studies in patients being treated with or porcine-bovine mixtures were first reported in 1975 (358,
insulin—as well as those in individuals with insulin auto- 359). Patients with recent-onset diabetes (32%) and those
immune-hypoglycemia syndrome— have suggested that the with long-standing illness (47%) were found to have positive
presence of high levels of IAs is associated with a retarded T cell responses to human insulin (360). Since T cell help is
disappearance rate of insulin, some authors have suggested required for B cell development and antibody production, IA
that antibodies may serve as a “buffer” to glucose variability levels might be expected to correlate with T cell responsive-
(see Section V.B.1) (6, 178, 223, 286, 343, 344). Limited studies ness. However, poor correlations between IA and/or IAA
demonstrated that this slowed rate of insulin disappearance levels and T cell responsiveness have been observed (360 –
appeared to decrease the likelihood of diabetic ketoacidosis, 362). Patients with high IAs or IAAs had very low T cell
contributing to stability of glycemic control (269, 345–347). responses, and patients with low IA or IAA levels had high
However, for the majority of patients, the levels of antibodies T cell responses. The relatively infrequent finding of a strong
seen with insulin therapy are unlikely to result in significant cellular immune response to insulin in patients with type 1
effects on glycemic variability (348). diabetes treated with exogenous insulin may be partly ex-
plained by the activation of regulatory T cells (75).
Additional investigations suggest that T cells respond to
H. Insulin antibodies and risk for diabetes different regions (epitopes) of the insulin molecule. T cell
1. IAs and ␤-cell loss. Although the spontaneous appearance responses are greater with the B chain than the A chain of
of IAA in nondiabetic patients is known to be predictive of insulin (363, 364). Theoretically, insulin-specific T cells mea-
type 1 diabetes development (349, 350), there is no evidence sured in these studies may be autoreactive T cells generated
that IAAs or IAs themselves causally mediate ␤-cell destruc- by endogenous insulin, autoreactive T cells expanded by
tion. Support for this comes from studies in which nondia- exogenous insulin, and/or T cells initially generated by ex-
betic patients with circulating antibodies to insulin were ogenous insulin; however, based on the assays used, it is not
followed for the onset of diabetes (351–354). For example, possible to differentiate among these possibilities. T cell re-
Bock et al. (351) investigated whether insulin treatment of sponses with exogenous insulin treatment likely will be fur-
patients without diabetes who were undergoing insulin ther investigated if insulin treatment is found to prevent the
shock therapy for psychiatric disorders would be at in- onset of type 1 diabetes in humans, as observed in NOD mice.
creased risk for the development of diabetes. In their retro-
spective analysis of 481 patients observed for an average of 3. Exogenous insulin and prevention of type 1 diabetes in clinical
22 yr, one patient developed type 1 diabetes, and 12 devel- studies. Results from the NOD mouse studies led to the Di-
oped type 2 diabetes. These instances did not differ from the abetes Prevention Trial Type 1, which was designed to test
background population. Only two of 27 patient samples ex- the ability of sc and orally administered insulin to prevent
amined were positive for IAs, and none was positive for islet type 1 diabetes in subjects known to be at risk based on the
cell antibodies. The researchers concluded that exogenous presence of autoimmune markers (e.g., IAAs and islet cell
insulin used in diabetes prevention trials was safe and would antibodies) (365). The sc insulin administration arm of the
not increase the risk for diabetes. trial showed no acceleration or delay of type 1 diabetes onset.
The same was true for the oral insulin treatment arm when
2. Cellular immune response to exogenous insulin. Most of the all patients were included (366). A subset analysis of the oral
reports on insulin-specific T cell responses have focused on insulin treatment group, however, demonstrated a signifi-
the autoreactive T cells involved in the pathogenesis of type cantly lower annualized rate of diabetes onset in patients
1 diabetes. Studies using NOD mice have demonstrated the with baseline IAA levels of at least 80 nU/ml treated with
presence of CD4⫹ and CD8⫹ cells that recognize insulin and oral insulin compared with patients treated with placebo.
lead to the destruction of ␤-cells in this model of spontaneous Additional studies showed no effect of oral insulin on
autoimmune diabetes (355). However, multiple interven- residual ␤-cell function in patients with new onset type 1
tions, including parenteral, oral, or aerosolized insulin treat- diabetes (103, 104). More recently, 38 children at risk for type
ment (see Sections III.A and III.B) have been shown to delay 1 diabetes were treated with intranasal insulin and showed
the onset of diabetes in these mice. no evidence for accelerated loss of ␤-cell function (367). Fur-
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 643

thermore, IA responses to intranasal insulin were demon- unregulated appearance of free insulin in the circulation
strated, as were immune changes consistent with mucosal (178). Clinical hypoglycemia in this setting may be multi-
tolerance to insulin. A small trial suggested that low-dose sc factorial in etiology, involving deficient glucose counterregu-
insulin may have favorable immunomodulatory effects in lation coupled with a prolonged half-life of free insulin (213,
adult patients with latent autoimmune diabetes (368). 300). Most studies published since 1980 have shown no ev-
idence for a significant relationship between IA and average
glycemia.
Some early studies suggested that IAs were associated
VI. Conclusion
with increases in neonatal morbidity, including hypoglyce-
Regardless of purity and origin, therapeutic insulins con- mia, respiratory distress syndrome, and hypocalcemia (304,
tinue to be immunogenic in humans; however, severe im- 310). However, studies in which pregnant women with di-
munological complications occur rarely, and less severe abetes were monitored for glycemic control argue against a
events affect just a small minority of patients. Today’s human connection between IAs and fetal risk. No distinguishing
insulins are free of noninsulin peptides and variations in clinical characteristics of neonatal hypoglycemia associated
insulin structure that contributed to the antigenicity seen with IAs, such as unusual prolongation, have been described
with pancreatic insulins in the past; however, insulins must in babies born to mothers with IAs.
be manufactured, stored, and delivered by nonphysiological Human studies have not shown consistent links between
means, possibly contributing to the humoral and cellular IAs and diabetic complications. Although studies have
immune responses seen in patients treated with insulin. shown increased levels of immune complexes in patients
IAs may be detectable in insulin-naive individuals who with diabetic microangiopathic complications (320 –322),
have a high likelihood of developing type 1 diabetes, have these immune complexes often do not contain insulin or IAs
had viral disorders, have been treated with various drugs, or (321–324). Neither does insulin administration contribute to
have autoimmune disorders or paraneoplastic syndromes. immune complex formation (325). The majority of studies
This suggests that under certain circumstances immune tol- have shown no relationship between IAs and diabetic an-
erance to insulin can be overcome. Factors that can lead to giopathic complications, including nephropathy, retinopa-
more or less susceptibility to humoral responses to exoge- thy, and neuropathy.
nous insulin include the recipient’s immune response genes, The absence of a pathogenic effect may be related to the
age, the presence of sufficient circulating autologous insulin, properties of the immune complex. Researchers have ob-
and the site of insulin delivery. Down-regulation of immune served that insulin and IA-immune complexes do not pre-
responses to self- and nonself-antigens involves a subset of cipitate (at antigen or antibody excess) and cannot be de-
regulatory T lymphocytes that may be critical in warding off tected by standard immunodiffusion methods (340). In
or terminating autoimmunity (67– 69). These regulatory cells addition, binding data suggested that insulin binding is uni-
may also play a role in the development and levels of im- valent, which limits the formation of large immune com-
mune responses to exogenous insulin. plexes. Small immune complexes do not readily bind to C1q
Researchers have postulated that IAs can be associated complement and are not, therefore, readily cleared by the
with insulin resistance, which would be detected clinically as reticuloendothelial system of the liver or spleen (190). Small
increases in insulin dose requirement. However, most stud- immune complexes have a similar half-life to circulating Ig
ies have shown no relationship between dose and IAs (58, 99, molecules. This explains the high levels of circulating total
114, 241, 247, 250, 252, 255, 259, 260, 262, 265, 266, 269 –273). insulin (antibody bound) in patients with high levels of IAs.
In long-term, follow-up studies of children with type 1 di- In conclusion, humoral antibody responses to exogenous
abetes, neither the presence of IAAs nor the development of insulin continue to be largely unavoidable. Little proof exists
IAs affected insulin dose requirements (114). that the development of antibodies to exogenous insulin
Case reports indicate that high levels of IAs can be asso- therapy affects glycemic control, insulin dose requirements,
ciated with prolonged or recurrent hypoglycemia, but stud- and hypoglycemia, or contributes to ␤-cell failure or to the
ies of large populations have failed to establish a relationship long-term complications of diabetes. Current human insulin
between IAs and hypoglycemia event rates. Reports describe and insulin analog therapies have resulted in decreased IA
patients with hypoglycemia syndromes who are found to levels when contrasted with animal insulins. Local reactions
have markedly high levels of IAs, but it is difficult to deter- to the most recent formulations of insulin continue to be
mine from the literature how many patients with high an- observed but are infrequent, and systemic reactions are rare.
tibody levels do not have hypoglycemia. Until therapeutic insulin can be delivered in a physiological
It is not clear what factors may precipitate the hypothe- manner, we are likely to continue to observe infrequent im-
sized unregulated dissociation of antibody-insulin com- munological sequelae. The development of IAs to exogenous
plexes. Because some patients are reported to have nocturnal insulin reflects the exquisite sensitivity of the immune sys-
hypoglycemia, researchers often presume that binding equi- tem to even minor perturbations.
librium is shifted toward antibody-insulin dissociation when
free insulin concentrations decline. Clinical trial data suggest Acknowledgments
that hypoglycemia does not correlate with IAs in large pop-
ulations of patients; however, it cannot be excluded that a Address all correspondence and requests for reprints to: S. Edwin
small number of patients with high antibody levels develop Fineberg, 3504 Mountain Lane, Mountain Brook, Alabama 35213. E-mail:
hypoglycemia syndromes owing in part to prolonged or efineber@iupui.edu
644 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

Disclosure Statement: S.E.F. serves on advisory boards for Pfizer Inc., from 17th European Peptide Symposium in Prague. Berlin: Walter
Eli Lilly and Company, and MannKind Corporation. T.T.K., D.F.-K., de Gruter; 387–394
R.J.F., G.L.F., and A.S.K. are employees of Pfizer Inc. 24. Ege H 1985 Sources and characteristics of the human insulins. In:
Statistical support was provided by Pamela F. Schwartz of Pfizer Inc. Breimer DD, Speiser P, eds. Topics in pharmaceutical sciences. New
Editorial support was provided by Mark Poirier, BA, of PAREXEL and York: Elsevier Science Publishers; 57–70
was funded by Pfizer Inc. 25. Chance RE, Hoffman JA 1981 The production of human insulin
using recombinant DNA technology and a new chain combinations
procedure. In: Rick DH, Gross E, eds. Peptides: synthesis-structure-
References function. Proceedings of the Seventh American Peptide Sympo-
sium. Rockford, IL: Pierce Chemical Company; 721–728
1. Banting FG, Best CH 1922 Pancreatic extracts. J Lab Clin Med 26. Schernthaner G, Borkenstein M, Fink M, Mayr WR, Menzel J,
7:3–11 Schober E 1983 Immunogenicity of human insulin (Novo) or pork
2. Bliss M 1982 The discovery of insulin. Chicago: University of monocomponent insulin in HLA-DR-typed insulin-dependent di-
Chicago Press abetic individuals. Diabetes Care 6(Suppl 1):43– 48
3. Williams JR 1922 A clinical study of the effects of insulin in severe 27. Heding LG, Marshall MO, Persson B, Dahlquist G, Thalme B,
diabetes. J Metab Res 2:729 –751 Lindgren F, Akerblom HK, Rilva A, Knip M, Ludvigsson J 1984
4. Tuft L 1928 Insulin hypersensitivities: immunologic considerations Immunogenicity of monocomponent human and porcine insulin in
and case reports. Am J Med Sci 176:707–720 newly diagnosed type 1 (insulin-dependent) diabetic children. Dia-
5. Lowell FC 1944 Immunologic studies in insulin resistance. J Clin betologia 27(Suppl):96 –98
Invest 23:233–240 28. Fineberg SE, Galloway JA, Fineberg NS, Rathbun MJ, Hufferd S
6. Berson SA, Yalow RS, Bauman A, Rothschild MA, Newerly K 1983 Immunogenicity of recombinant DNA human insulin. Dia-
1956 Insulin-I131 metabolism in human subjects: demonstration of betologia 25:465– 469
insulin binding globulin in the circulation of insulin treated sub- 29. Galloway JA, deShazo R 1983 The clinical use of insulin and the
jects. J Clin Invest 35:170 –190 complications of insulin therapy. In: Ellenberg M, Rifkin H, eds.
7. Carpenter FH 1958 Partition column chromatography of insulin in Diabetes mellitus, theory and practice. New Hyde Park, NY: Med-
2-butanol-aqueous acid systems. Arch Biochem Biophys 78:539 – ical Examination Publishing; 519
545 30. Ganz MA, Unterman T, Roberts M, Uy R, Sahgal S, Samter M,
8. Thompson EOP, O’Donnell IJ 1960 The chromatography of in- Grammer LC 1990 Resistance and allergy to recombinant human
sulin on DEAE-cellulose in buffers containing 8 M urea. Aust J Bio insulin. J Allergy Clin Immunol 86:45–51
Sci 13:393– 400 31. Bodtger U, Wittrup M 2005 A rational clinical approach to sus-
9. Mirsky IA, Kawamura K 1966 Heterogeneity of crystalline insulin. pected insulin allergy: status after five years and 22 cases. Diabet
Endocrinology 78:1115–1119 Med 22:102–106
10. Brange J 1987 Galenics of insulin: the physico-chemical and phar- 32. Leonet J, Malaise J, Goffin E, Lefebvre C, Tennstedt D, Van-
maceutical aspects of insulin and insulin preparations. New York: deleene B, Buysschaert M, Squifflet JP 2006 Solitary pancreas
Springer-Verlag transplantation for life-threatening allergy to human insulin.
11. Paley RG, Turnbridge RE 1952 Dermal reactions to insulin ther- Transplant Int 19:474 – 477
apy. Diabetes 1:22 33. Beltrand J, Guilmin-Crepon S, Castanet M, Peuchmaur M,
12. Patterson R, Lucena G, Metz R, Roberts M 1969 Reaginic antibody Czernichow P, Levy-Marchal C 2006 Insulin allergy and extensive
against insulin: demonstration of antigenic distinction between
lipoatrophy in child with type 1 diabetes. Horm Res 65:253–260
native and extracted insulin. J Immunol 103:1061–1071
34. Castera V, Dutour-Meyer A, Koeppel M, Petitjean C, Darmon P
13. Yagi Y, Maier P, Pressman D, Arbesman CE, Reisman RE,
2005 Systemic allergy to human insulin and its rapid and long
Lenzner AR 1963 Multiplicity of insulin-binding antibodies in hu-
acting analogs: successful treatment by continuous subcutaneous
man sera: presence of antibody activity in ␥-, ␤-2a, and ␤-2m-
insulin lispro infusion. Diabetes Metab 31(4 Pt 1):391– 400
globulins. J Immunol 90:760 –769
35. Kang S, Brange J, Burch A, Volund A, Owens DR 1991 Subcu-
14. Shipp JC, Cunningham RW, Russell RO, Marble A 1965 Insulin
resistance: clinical features, natural course and effects of adrenal taneous insulin absorption explained by insulin’s physicochemical
steroid treatment. Medicine (Baltimore) 44:165–186 properties: evidence from absorption studies of soluble human
15. Oakley WG, Jones VE, Cunliffe AC 1967 Insulin resistance. Br insulin and insulin analogues in humans. Diabetes Care 14:942–948
Med J 2:134 –138 36. Mooradian AD, Bernbaum M, Albert SG 2006 Narrative review:
16. Steiner DF, Hallund O, Rubenstein A, Cho S, Bayliss C 1968 a rational approach to starting insulin therapy. Ann Intern Med
Isolation and properties of proinsulin, intermediate forms, and 145:125–134
other minor components from crystalline bovine insulin. Diabetes 37. Vajo Z, Fawcett J, Duckworth WC 2001 Recombinant DNA tech-
17:725–736 nology in the treatment of diabetes: insulin analogs. Endocr Rev
17. Damgaard U, Markussen J 1979 Analysis of insulins and related 22:706 –717
compounds by HPLC. Horm Metab Res 11:580 –581 38. Robinson DM, Wellington K 2006 Insulin glulisine. Drugs 66:
18. Welinder BS, Sorensen HH, Hansen B 1986 Reversed-phase high- 861– 869
performance liquid chromatography of insulin: resolution and re- 39. Dailey G, Rosenstock J, Moses RG, Ways K 2004 Insulin glulisine
covery in relation to column geometry and buffer components. provides improved glycemic control in patients with type 2 dia-
J Chromatogr 361:357–367 betes. Diabetes Care 27:2363–2368
19. Klaff LJ, Vinik AI, Berelowitz M, Jackson WP 1978 Circulating 40. Fineberg NS, Fineberg SE, Anderson JH, Birkett MA, Gibson RG,
antibodies in diabetics treated with conventional and purified in- Hufferd S 1996 Immunologic effects of insulin lispro [Lys (B28),
sulins. S Afr Med J 54:149 –153 Pro (B29) human insulin] in IDDM and NIDDM patients previously
20. Kahn CR, Rosenthal AS 1979 Immunologic reactions to insulin: treated with insulin. Diabetes 45:1750 –1754
insulin allergy, insulin resistance, and the autoimmune insulin 41. Fineberg SE, Huang J, Brunelle R, Gulliya KS, Anderson Jr JH
syndrome. Diabetes Care 2:283–295 2003 Effect of long-term exposure to insulin lispro on the induction
21. Chance RE, Root MA, Galloway JA 1976 The immunogenicity of of antibody response in patients with type 1 or type 2 diabetes.
insulin preparations. Acta Endocrinol Suppl (Copenh) 205:185–198 Diabetes Care 26:89 –96
22. Fineberg SE, Galloway JA, Fineberg NS, Goldman J 1983 Effects 42. Lindholm A, Jensen LB, Home PD, Raskin P, Boehm BO, Rastam
of species of origin, purification levels, and formulation on insulin J 2002 Immune responses to insulin aspart and biphasic insulin
immunogenicity. Diabetes 32:592–599 aspart in people with type 1 and type 2 diabetes. Diabetes Care
23. Markussen J, Schaumberg K 1983 Reaction mechanism in trypsin 25:876 – 882
catalyzed synthesis of human insulin studied by 17 O-NMR spec- 43. Urdaneta F, Lobato EB, Kirby RR, Horrow JC 1999 Noncardio-
troscopy. In: Blakha K, Malon P, eds. Peptides 1982. Proceedings genic pulmonary edema associated with protamine administration
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 645

during coronary artery bypass graft surgery. J Clin Anesth 11:675– 63. Teeter JG, Riese RJ 2006 Dissociation of lung function changes
681 with humoral immunity during inhaled human insulin therapy.
44. Weiler JM, Gellhaus MA, Carter JG, Meng RL, Benson PM, Hottel Am J Respir Crit Care Med 173:1194 –1200
RA, Schillig KB, Vegh AB, Clarke WR 1990 A prospective study 64. An B, Reinhardt RR 2003 Effects of different durations of breath
of the risk of an immediate adverse reaction to protamine sulfate holding after inhalation of insulin using the AERx insulin diabetes
during cardiopulmonary bypass surgery. J Allergy Clin Immunol management system. Clin Ther 25:2233–2244
85:713–719 65. Guermonprez P, Valladeau J, Zitvogel L, Thery C, Amigorena S
45. Blumer IR 2006 Severe injection site reaction to insulin detemir. 2002 Antigen presentation and T cell stimulation by dendritic cells.
Diabetes Care 29:946 Annu Rev Immunol 20:621– 667
46. Darmon P, Castera V, Koeppel MC, Petitjean C, Dutour 2005 A 66. Chatenoud L, Bach JF 2006 Adaptive human regulatory T cells:
type III allergy to insulin detemir. Diabetes Care 28:2980 myth or reality? J Clin Invest 116:2325–2327
47. Pieber TR, Eugene-Jolchine I, Derobert E 2000 Efficacy and safety 67. Bluestone JA, Abbas AK 2003 Natural versus adaptive regulatory
of HOE 901 versus NPH insulin in patients with type 1 diabetes: T cells. Nat Rev Immunol 3:253–257
the European Study Group of HOE 901 in type 1 diabetes. Diabetes 68. Piccirillo CA, Tritt M, Sgouroudis E, Albanese A, Pyzik M, Hay
Care 23:157–162 V 2005 Control of type 1 autoimmune diabetes by naturally oc-
48. Dahl-Jorgensen K, Torjesen P, Hanssen KF, Sandvik L, Aagenaes curring CD4⫹CD25⫹ regulatory T lymphocytes in neonatal NOD
O 1987 Increase in insulin antibodies during continuous subcuta- mice. Ann NY Acad Sci 1051:72– 87
neous insulin infusion and multiple-injection therapy in contrast to 69. Sakaguchi S 2005 Naturally arising Foxp3-expressing
conventional treatment. Diabetes 36:1–5 CD25⫹CD4⫹ regulatory T cells in immunological tolerance to self
49. Jeandidier N, Boivin S, Sapin R, Rosart-Ortega F, Uring-Lambert and non-self. Nat Immunol 6:345–352
B, Reville P, Pinget M 1995 Immunogenicity of intraperitoneal 70. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady
insulin infusion using programmable implantable devices. Diabe- G, Wahl SM 2003 Conversion of peripheral CD4⫹CD25-naive T
tologia 38:577–584 cells to CD4⫹CD25⫹ regulatory T cells by TGF-␤ induction of
50. Olsen CL, Chan E, Turner DS, Iravani M, Nagy M, Selam JL, transcription factor Foxp3. J Exp Med 198:1875–1886
Wong ND, Waxman K, Charles MA 1994 Insulin antibody re- 71. Weigle WO 1981 Self-nonself recognition by T and B lymphocytes
sponses after long-term intraperitoneal insulin administration via and their roles in autoimmune phenomena. Arthritis Rheum 24:
implantable programmable insulin delivery systems. Diabetes 1044 –1053
Care 17:169 –176 72. Whiteley PJ, Lake JP, Selden RF, Kapp JA 1989 Tolerance induced
51. Jeandidier N, Boullu S, Busch-Brafin MS, Chabrier G, Sapin R, by physiological levels of secreted proteins in transgenic mice ex-
Gasser F, Pinget M 2002 Comparison of antigenicity of Hoechst pressing human insulin. J Clin Invest 84:1550 –1554
21PH insulin using either implantable intraperitoneal pump or 73. Alard P, Manirarora JN, Parnell SA, Hudkins JL, Clark SL,
subcutaneous external pump infusion in type 1 diabetic patients. Kosiewicz MM 2006 Deficiency in NOD antigen-presenting cell
Diabetes Care 25:84 – 88 function may be responsible for suboptimal CD4⫹CD25⫹ T-cell-
52. Diabetes Control and Complications Trial Research Group 1993
mediated regulation and type 1 diabetes development in NOD
The effect of intensive treatment of diabetes on the development
mice. Diabetes 55:2098 –2105
and progression of long-term complications in insulin-dependent
74. Lepault F, Gagnerault MC 2000 Characterization of peripheral
diabetes mellitus. N Engl J Med 329:977–986
regulatory CD4⫹ T cells that prevent diabetes onset in nonobese
53. Korytkowski M 2002 When oral agents fail: practical barriers to
diabetic mice. J Immunol 164:240 –247
starting insulin. Int J Obes Relat Metab Disord 26(Suppl 3):S18 –S24
75. Tiittanen M, Huupponen JT, Knip M, Vaarala O 2006 Insulin
54. Riddle MC 2002 The underuse of insulin therapy in North Amer-
treatment in patients with type 1 diabetes induces upregulation of
ica. Diabetes Metab Res Rev 18(Suppl 3):S42–S49
55. Patton JS, Bukar J, Nagarajan S 1999 Inhaled insulin. Adv Drug regulatory T-cell markers in peripheral blood mononuclear cells
Deliv Rev 35:235–247 stimulated with insulin in vitro. Diabetes 55:3446 –3454
56. White S, Bennett DB, Cheu S, Conley PW, Guzek DB, Gray S, 76. Jensen PE, Pierce CW, Kapp JA 1984 Regulatory mechanisms in
Howard J, Malcolmson R, Parker JM, Roberts P, Sadrzadeh N, immune responses to heterologous insulins. II. Suppressor T cell
Schumacher JD, Seshadri S, Sluggett GW, Stevenson CL, Harper activation associated with nonresponsiveness in H-2b mice. J Exp
NJ 2005 EXUBERA: pharmaceutical development of a novel prod- Med 160:1012–1026
uct for pulmonary delivery of insulin. Diabetes Technol Ther 77. Jensen PE, Kapp JA 1985 Stimulation of helper T cells and dom-
7:896 –906 inant suppressor T cells that recognize autologous insulin. J Mol
57. Fineberg SE 2006 Diabetes therapy trials with inhaled insulin. Cell Immunol 2:133–139
Expert Opin Investig Drugs 15:743–762 78. Schernthaner G 1993 Immunogenicity and allergenic potential of
58. Fineberg SE, Kawabata T, Finco-Kent D, Liu C, Krasner A 2005 animal and human insulins. Diabetes Care 16(Suppl 3):155–165
Antibody response to inhaled insulin in patients with type 1 or type 79. Van Haeften TW 1989 Clinical significance of insulin antibodies in
2 diabetes: an analysis of initial phase II and III inhaled insulin insulin-treated diabetic patients. Diabetes Care 12:641– 648
(Exubera) trials and a two-year extension trial. J Clin Endocrinol 80. Fineberg SE, Galloway JA, Fineberg NS, Rathbun MJ 1982 Im-
Metab 90:3287–3294 munologic improvement resulting from the transfer of animal in-
59. Heise T, Bott S, Tusek C, Stephan JA, Kawabata T, Finco-Kent D, sulin-treated diabetic subjects to human insulin (recombinant
Liu C, Krasner A 2005 The effect of insulin antibodies on the DNA). Diabetes Care 5(Suppl 2):107–113
metabolic action of inhaled and subcutaneous insulin: a prospec- 81. Raskin P, Guthrie RA, Leiter L, Riis A, Jovanovic L 2000 Use of
tive randomized pharmacodynamic study. Diabetes Care 28:2161– insulin aspart, a fast-acting insulin analog, as the mealtime insulin
2169 in the management of patients with type 1 diabetes. Diabetes Care
60. Hermansen K, Ronnemaa T, Petersen AH, Bellaire S, Adamson 23:583–588
U 2004 Intensive therapy with inhaled insulin via the AERx insulin 82. Marshall MO, Heding LG, Villumsen J, Akerblom HK, Baevre H,
diabetes management system: a 12-week proof-of-concept trial in Dahlquist G, Kjaergaard JJ, Knip M, Lindgren F, Ludvigsson J
patients with type 2 diabetes. Diabetes Care 27:162–167 1988 Development of insulin antibodies, metabolic control and
61. Brunner GA, Balent B, Ellmerer M, Schaupp L, Siebenhofer A, B-cell function in newly diagnosed insulin dependent diabetic chil-
Jendle JH, Okikawa J, Pieber TR 2001 Dose-response relation of dren treated with monocomponent human insulin or monocom-
liquid aerosol inhaled insulin in type I diabetic patients. Diabeto- ponent porcine insulin. Diabetes Res 9:169 –175
logia 44:305–308 83. Fineberg SE, Fineberg NS, Galloway JA, Rabin BS, Fireman P,
62. Rave K, Bott S, Heinemann L, Sha S, Becker RHA, Willavize SA, Irvine WJ 1984 What factors regulate the immune response to
Heise T 2005 Time-action profile of inhaled insulin in comparison insulin in type 1 and 2 diabetes? Diabetes 33(Suppl 1):157A (Ab-
with subcutaneously injected insulin lispro and regular human stract)
insulin. Diabetes Care 28:1077–1082 84. Goldman J, Baldwin D, Pugh W, Rubenstein AH 1978 Equilib-
646 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

rium binding assay and kinetic characterization of insulin anti- Gepner C, Bouhanick B, Mogenet A, Nicolino M, Carel JC,
bodies. Diabetes 27:653– 660 Lecomte P, Marechaud R, Bougneres P, Charbonnel B, Sai P 2000
85. Fineberg SE, Fineberg NS, Estridge T, Hufford S 1988 Long term Oral insulin administration and residual ␤-cell function in recent-
sequelae of human vs. pork vs. mixed beef pork insulins in type 1 onset type 1 diabetes: a multicentre randomised controlled trial.
and 2 diabetes in a two year double blind clinical trial. Diabetes Diabete Insuline Orale group. Lancet 356:545–549
37(Suppl 1):P24A (Abstract) 105. Garg S, Rosenstock J, Silverman BL, Sun B, Konkoy CS, de la
86. Thomas JW, Virta VJ, Nell LJ 1985 Heterogeneity and specificity Pena A, Muchmore DB 2006 Efficacy and safety of preprandial
of human anti-insulin antibodies determined by isoelectric focus- human insulin inhalation powder versus injectable insulin in pa-
ing. J Immunol 134:1048 –1052 tients with type 1 diabetes. Diabetologia 49:891– 899
87. Maneschi F, Fineberg SE, Kohner EM 1982 Successful treatment 106. Karjalainen J, Knip M, Mustonen A, Akerblom HK 1988 Insulin
of immune-mediated insulin resistance by human insulin (recom- autoantibodies at the clinical manifestation of type 1 (insulin-de-
binant DNA). Diabetes Care 5(Suppl 2):175–179 pendent) diabetes—a poor predictor of clinical course and antibody
88. Grammer LC, Roberts M, Patterson R 1985 IgE and IgG antibody response to exogenous insulin. Diabetologia 31:129 –133
against human (recombinant DNA) insulin in patients with sys- 107. Kahn CR, Mann D, Rosenthal AS, Galloway JA, Johnson AH,
temic insulin allergy. J Lab Clin Med 105:108 –113 Mendell N 1982 The immune response to insulin in man: inter-
89. Schlichtkrull J, Brange J, Christiansen AH, Hallund O, Heding action of HLA alloantigens and the development of the immune
LG, Jorgensen KH 1972 Clinical aspects of insulin—antigenicity. response. Diabetes 31:716 –723
Diabetes 21(Suppl):56 108. Reeves WG, Gelsthorpe K, Van der Minne P, Torensma R, Tat-
90. Feinglos MN, Jegasothy BV 1979 “Insulin” allergy due to zinc. tersall RB 1984 HLA phenotype and insulin antibody production.
Lancet 1:122–124 Clin Exp Immunol 57:443– 448
91. Stewart WJ, McSweeney SM, Kellett MA, Faxon DP, Ryan TJ 1984 109. McEvoy RC, Witt ME, Ginsberg-Fellner F, Rubinstein P 1986
Increased risk of severe protamine reactions in NPH insulin-de- Anti-insulin antibodies in children with type I diabetes mellitus:
pendent diabetics undergoing cardiac catheterization. Circulation genetic regulation of production and presence at diagnosis before
70:788 –792 insulin replacement. Diabetes 35:634 – 641
92. Robbins DC, Mead PM 1987 Free covalent aggregates of thera- 110. Sklenar I, Neri TM, Berger W, Erb P 1982 Association of specific
peutic insulin in blood of insulin-dependent diabetics. Diabetes immune response to pork and beef insulin with certain HLA-DR
36:147–151 antigens in type 1 diabetes. Br Med J (Clin Res Ed) 285:1451–1453
93. Ratner RE, Phillips TM, Steiner M 1990 Persistent cutaneous 111. Asplin CM, Dornan TL, Raghu PK, Hansen JA, Palmer JP 1984
insulin allergy resulting from high-molecular-weight insulin ag- The antibody response to insulin therapy: a prospective study in
gregates. Diabetes 39:728 –733 HLA-typed insulin-dependent diabetic subjects. Diabetes 33:966 –
94. Micossi P, Galimberti G, Librenti MC, Petrella G, Dozio N, 969
Cristallo M, Sodoyez JC, Sodoyez-Goffaux F, Grau U, Zolto- 112. Nakao Y, Matsumoto H, Miyazaki T, Mizuno N, Arima N,
brocki M 1988 Glycerol-insulin raises circulating insulin antibodies Wakisaka A, Okimoto K, Akazawa Y, Tsuji K, Fujita T 1981 IgG
in type I diabetic patients treated with permanently implanted heavy-chain (Gm) allotypes and immune response to insulin in
devices. Metabolism 37:1029 –1032 insulin-requiring diabetes mellitus. N Engl J Med 304:407– 409
95. Jeandidier N, Boullu S, Delatte E, Sapin R, Steibel J, Meyer P, Uhl 113. Sutton M, Klaff LJ, Asplin CM, Clemons P, Tatpati O, Lyen K,
C, Pinget M 2001 High antigenicity of intraperitoneal insulin in- Raghu P, Baker L, Guthrie R, Sperling M 1988 Insulin autoanti-
fusion via implantable devices: preliminary rat studies. Horm bodies at diagnosis of insulin-dependent diabetes: effect on the
Metab Res 33:34 –38 antibody response to insulin treatment. Metab Clin Exp 37:1005–
96. Robbins PF, Thomas JW, Jensen PE, Kapp JA 1984 Insulin-specific 1007
tolerance induction. I. Abrogation of helper T cell activity is con- 114. Salardi S, Cacciari E, Steri L, Donati S, Mandini M, Gualandi S,
trolled by H-2-linked Ir genes. J Immunol 132:43– 49 Mantovani W, Zucchini S 1995 An 8-year follow-up of anti-insulin
97. Lalor PA, Morahan G 1990 The peritoneal Ly-1 (CD5) B cell rep- antibodies in diabetic children: relation to insulin autoantibodies,
ertoire is unique among murine B cell repertoires. Eur J Immunol HLA type, ␤-cell function, clinical course and type of insulin ther-
20:485– 492 apy. Acta Paediatr 84:639 – 645
98. Overkamp D, Mohammed-Ali S, Cartledge C, Landon J 1988 115. Davis SN, Thompson CJ, Peak M, Brown MD, Alberti KGMM
Production of polyclonal antibodies in ascitic fluid of mice: tech- 1992 Effects of human insulin on insulin binding antibody pro-
nique and applications. J Immunoassay 9:51– 68 duction in nondiabetic subjects. Diabetes Care 15:124 –126
99. Lassmann-Vague V, Belicar P, Raccah D, Vialettes B, Sodoyez JC, 116. Vardi P, Modan-Mozes D, Ish-Shalom S, Soloveitzik L, Barzilai
Vague P 1995 Immunogenicity of long-term intraperitoneal insulin D, Modan M 1993 Low titer, competitive insulin autoantibodies are
administration with implantable programmable pumps: metabolic spontaneously produced in autoimmune diseases of the thyroid.
consequences. Diabetes Care 18:498 –503 Diabetes Res Clin Pract 21:161–166
100. Duckworth WC, Saudek CD, Giobbie-Hurder A, Henderson WG, 117. Miller RA 1996 The aging immune system: primer and prospectus.
Henry RR, Kelley DE, Edelman SV, Zieve FJ, Adler RA, Anderson Science 273:70 –74
JW, Anderson RJ, Hamilton BP, Donner TW, Kirkman MS, Mor- 118. Schmucker DL, Heyworth MF, Owen RL, Daniels CK 1996 Impact
gan NA 1998 The Veterans Affairs Implantable Insulin Pump of aging on gastrointestinal mucosal immunity. Dig Dis Sci 41:
Study: effect on cardiovascular risk factors. Diabetes Care 21:1596 – 1183–1193
1602 119. McElhaney JE, Meneilly GS, Beattie BL, Helgason CD, Lee SF,
101. Chute EP, Rupp WM, Barbosa JJ, Rohde TD, Bordewich P, Hitch- Devine RD, Bleackley RC 1992 The effect of influenza vaccination
cock CR, Goldenberg FJ, Buchwald H 1985 Anti-insulin antibody on IL2 production in healthy elderly: implications for current vac-
titers after one year of intravenous insulin infusion by implantable cination practices. J Gerontol 47:M3–M8
pump. Surg Forum 36:189 –191 120. Gorus FK, Vandewalle CL, Dorchy H, Van Crombrugge P, Schuit
102. Liebl A, Renner R, Hepp KD 2000 Reduction of anti-insulin an- FC, Pipeleers DG 1994 Influence of age on the associations among
tibodies in type 1 diabetic patients treated with continuous intra- insulin autoantibodies, islet cell antibodies, and HLA DAQ1*0301-
peritoneal insulin infusion (CIPII) via the umbilical vein (CIUII). DQB1*0302 in siblings of patients with type 1 (insulin-dependent)
Diabetes 49(Suppl 1):A115 (Abstract) diabetes mellitus. Belgian Diabetes Registry. J Clin Endocrinol
103. Pozzilli P, Pitocco D, Visalli N, Cavallo MG, Buzzetti R, Crino A, Metab 78:1172–1178
Spera S, Suraci C, Multari G, Cervoni M, Manca Bitti ML, Mat- 121. Fineberg NS, Fineberg SE, Linarelli LG, Mahler J 1984 Differences
teoli MC, Marietti G, Ferrazzoli F, Cassone Faldetta MR, Gior- in recognition of insulin as an immunogen in type 1 and type 2
dano C, Sbriglia M, Sarugeri E, Ghirlanda G 2000 No effect of oral diabetes. Diabetes 33(Suppl 1):39A (Abstract)
insulin on residual ␤-cell function in recent-onset type I diabetes 122. Fineberg NS, Fineberg SE, Galloway J 1992 Does age at initiation
(the IMDIAB VII): IMDIAB Group. Diabetologia 43:1000 –1004 of insulin therapy determine who will develop an immune re-
104. Chaillous L, Lefevre H, Thivolet C, Boitard C, Lahlou N, Atlan- sponse? Diabetes 41(Suppl 1):191A (Abstract)
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 647

123. Zwickl CM, Smith HW, Zimmermann JL, Wierda D 1995 Immu- R, Krischer J, Maclaren N 1995 Insulin immunization of nonobese
nogenicity of biosynthetic human LysPro insulin compared to na- diabetic mice induces a protective insulitis characterized by di-
tive-sequence human and purified porcine insulins in rhesus mon- minished intraislet interferon-␥ transcription. J Clin Invest 95:628 –
keys immunized over a 6-week period. Arzneimittelforschung 45: 634
524 –528 143. Daniel D, Wegmann DR 1996 Protection of nonobese diabetic mice
124. Rosenberg AS, Worobec A A risk-based approach to immunoge- from diabetes by intranasal or subcutaneous administration of
nicity concerns of therapeutic protein products, part 3: effects of insulin peptide B-(9 –23). Proc Natl Acad Sci USA 93:956 –960
manufacturing changes in immunogenicity and the utility of ani- 144. Hancock WW, Polanski M, Zhang J, Blogg N, Weiner HL 1995
mal immunogenicity studies. BioPharm International. Available at: Suppression of insulitis in non-obese diabetic (NOD) mice by oral
http://www.biopharminternational.com/biopharm/article/articleDe- insulin administration is associated with selective expression of
tail.jsp?id⫽146216. Accessed October 30, 2006 interleukin-4 and -10, transforming growth factor-␤, and prostag-
125. Ottesen JL, Nilsson P, Jami J, Weilguny D, Duhrkop M, Bucchini landin-E. Am J Pathol 147:1193–1199
D, Havelund S, Fogh JM 1994 The potential immunogenicity of 145. Yu L, Robles DT, Abiru N, Kaur P, Rewers M, Kelemen K,
human insulin and insulin analogues evaluated in a transgenic Eisenbarth GS 2000 Early expression of antiinsulin autoantibodies
mouse model. Diabetologia 37:1178 –1185 of humans and the NOD mouse: evidence for early determination
126. McConnell WR, Finch G, Elwell M, Kawabata T, Moutvic R, of subsequent diabetes. Proc Natl Acad Sci USA 97:1701–1706
Shaw M, Stammberger I 2006 Toxicological investigations on in- 146. Flynn SD, Keren DF, Torretti B, Dieterle RC, Grauds S 1981
haled insulin. Toxicologist 90:127 Factors affecting enzyme-linked immunosorbent assay (ELISA) for
127. Wolff RK, Vick AM, Wroblewski VJ, Engelhardt JA, Deaver DR, insulin antibodies in serum. Clin Chem 27:1753–1757
Shaw ME 2004 Thirteen-week inhalation study to characterize the 147. Kobayashi N, Terada E, Ghoda A, Kitagaki K, Okudaira H, Ogita
toxicity and PK/PD of a human inhaled insulin powder (HIIP) in T, Shimabukuro K, Miyamoto T 1985 A solid-phase enzyme im-
beagle dogs. Diabetes 53(Suppl 2):A115–A116 (Abstract) munoassay for anti-insulin antibody in diabetes mellitus patients.
128. Hammad H, Lambrecht BN 2006 Recent progress in the biology of J Immunol Methods 84:245–250
airway dendritic cells and implications for understanding the reg- 148. Wilkin T, Nicholson S, Casey C 1985 A micro enzyme-linked
ulation of asthmatic inflammation. J Allergy Clin Immunol 118: immunosorbent assay for insulin antibodies in serum. J Immunol
331–336 Methods 76:185–194
129. Harrison LC, Dempsey-Collier M, Kramer DR, Takahashi K 1996 149. Dib SA, Freire MB, Miranda WL, Russo EM 1994 Detection of
Aerosol insulin induces regulatory CD8 ␥ ␦ T cells that prevent insulin antibodies by radioassay and ELISA: interrelation and cor-
murine insulin-dependent diabetes. J Exp Med 184:2167–2174 relation with metabolic control in type I diabetes. Braz J Med Bio
130. Atkinson MA, Maclaren NK, Luchetta R 1990 Insulitis and dia- Res 27:1167–1180
betes in NOD mice reduced by prophylactic insulin therapy. Di- 150. Sodoyez-Goffaux F, Koch M, Dozio N, Brandenburg D, Sodoyez
abetes 39:933–937 JC 1988 Advantages and pitfalls of radioimmune and enzyme
131. Bowman MA, Campbell L, Darrow BL, Ellis TM, Suresh A, At- linked immunosorbent assays of insulin antibodies. Diabetologia
kinson MA 1996 Immunological and metabolic effects of prophy- 31:694 –702
lactic insulin therapy in the NOD-scid/scid adoptive transfer 151. Stumpo RR, Llera AS, Cardoso AI, Poskus E 1994 Solid versus
model of IDDM. Diabetes 45:205–208 liquid phase assays in detection of insulin antibodies: influence of
132. Gotfredsen CF, Buschard K, Frandsen EK 1985 Reduction of di- iodination site on labelled insulin binding. J Immunol Methods
abetes incidence of BB Wistar rats by early prophylactic insulin 169:241–249
treatment of diabetes-prone animals. Diabetologia 28:933–935 152. Greenbaum CJ, Palmer JP, Kuglin B, Kolb H 1992 Insulin auto-
133. Gottlieb PA, Handler ES, Appel MC, Greiner DL, Mordes JP, antibodies measured by radioimmunoassay methodology are more
Rossini AA 1991 Insulin treatment prevents diabetes mellitus but related to insulin-dependent diabetes mellitus than those measured
not thyroiditis in RT6-depleted diabetes resistant BB/Wor rats. by enzyme-linked immunosorbent assay: results of the Fourth In-
Diabetologia 34:296 –300 ternational Workshop on the Standardization of Insulin Autoan-
134. Vlahos WD, Seemayer TA, Yale JF 1991 Diabetes prevention in BB tibody Measurement. J Clin Endocrinol Metab 74:1040 –1044
rats by inhibition of endogenous insulin secretion. Metabolism 153. Diaz JL, Wilkin T 1986 Comparison of liquid phase radiobinding
40:825– 829 assay and solid phase ELISA for the measurement of insulin an-
135. Bergerot I, Fabien N, Maguer V, Thivolet C 1994 Oral adminis- tibodies and insulin autoantibodies in serum. Diabetic Med 3:316 –
tration of human insulin to NOD mice generates CD4⫹ T cells that 321
suppress adoptive transfer of diabetes. J Autoimmun 7:655– 663 154. Bingley PJ, Bonifacio E, Mueller PW 2003 Diabetes Antibody
136. Muir A, Schatz D, Maclaren N 1993 Antigen-specific immuno- Standardization Program: first assay proficiency evaluation. Dia-
therapy: oral tolerance and subcutaneous immunization in the betes 52:1128 –1136
treatment of insulin-dependent diabetes. Diabetes Metab Rev 155. Achenbach P, Schlosser M, Williams AJ, Yu L, Mueller PW,
9:279 –287 Bingley PJ, Bonifacio E 2007 Combined testing of antibody titer
137. Ploix C, Bergerot I, Fabien N, Perche S, Moulin V, Thivolet C 1998 and affinity improves insulin autoantibody measurement: Diabetes
Protection against autoimmune diabetes with oral insulin is asso- Antibody Standardization Program. Clin Immunol 122:85–90
ciated with the presence of IL-4 type 2 T-cells in the pancreas and 156. Maniatis AK, Yu L, Miao D, Nelson K, Eisenbarth GS 2001 Rapid
pancreatic lymph nodes. Diabetes 47:39 – 44 assays for detection of anti-islet autoantibodies: implications for
138. Polanski M, Melican NS, Zhang J, Weiner HL 1997 Oral admin- organ donor screening. J Autoimmun 16:71–76
istration of the immunodominant B-chain of insulin reduces dia- 157. Andersen OO 1976 Clinical significance of anti-insulin-antibodies.
betes in a co-transfer model of diabetes in the NOD mouse and is Acta Endocrinol 205(Suppl):231–240
associated with a switch from Th1 to Th2 cytokines. J Autoimmun 158. Devlin JG 1966 Evidence for the existence of an IgM immuno-
10:339 –346 globulin to insulin. Ir J Med Sci 6:507–510
139. Von Herrath MG, Dyrberg T, Oldstone MB 1996 Oral insulin 159. Faulk WP, Karam JH, Fudenberg HH 1971 Human anti-insulin
treatment suppresses virus-induced antigen-specific destruction of antibodies. J Immunol 106:1112–1116
␤ cells and prevents autoimmune diabetes in transgenic mice. J Clin 160. Reisman RE, Wicher K, Arbesman CE 1969 Immunotherapy with
Invest 98:1324 –1331 antigen E. J Allergy 44:82–95
140. Zhang ZJ, Davidson L, Eisenbarth G, Weiner HL 1991 Suppres- 161. Patterson R, O’Rourke J, Roberts M, Suszko I 1973 Immunologic
sion of diabetes in nonobese diabetic mice by oral administration reactions against insulin. I. IgG anti-insulin and insulin resistance.
of porcine insulin. Proc Natl Acad Sci USA 88:10252–10256 J Immunol 110:1126 –1134
141. Karounos DG, Bryson JS, Cohen DA 1997 Metabolically inactive 162. Kniker WT, Grodsky G, Forsham P 1967 Immunogenic vasculitis:
insulin analog prevents type I diabetes in prediabetic NOD mice. is it part of diabetes mellitus? In: Kimura SJ, Caygill WM, eds.
J Clin Invest 100:1344 –1348 Vascular complications of diabetes mellitus. St. Louis, MO: C.V.
142. Muir A, Peck A, Clare-Salzler M, Song YH, Cornelius J, Luchetta Mosby Company; 184
648 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

163. Dolovich J, Schnatz JD, Reisman RE, Yagi Y, Arbesman CE 1970 2005 Epitope analysis of insulin autoantibodies using recombinant
Insulin allergy and insulin resistance: case report with immuno- Fab. Clin Exp Immunol 140:564 –571
logic studies. J Allergy 46:127–137 186. Devendra D, Galloway TS, Horton SJ, Evenden A, Keller U,
164. Grammer LC, Roberts M, Buchanan TA, Fitzsimons R, Metzger Wilkin TJ 2003 The use of phage display to distinguish insulin
BE, Patterson R 1987 Specificity of immunoglobulin E and immu- autoantibody (IAA) from insulin antibody (IA) idiotypes. Diabe-
noglobulin G against human (recombinant DNA) insulin in human tologia 46:802– 809
insulin allergy and resistance. J Lab Clin Med 109:141–146 187. Devendra D, Galloway TS, Horton SJ, Wilkin TJ 2004 Exploring
165. Hamilton RG, Rendell M, Adkinson Jr NF 1980 Serological anal- the idiotypes of insulin antibodies as markers for remission in type
ysis of human IgG and IgE anti-insulin antibodies by solid-phase 1 diabetes. Diabet Med 21:1316 –1324
radioimmunoassays. J Lab Clin Med 96:1022–1036 188. Devendra D, Franke B, Galloway TS, Horton SJ, Knip M, Wilkin
166. Kumar D 1977 Anti-insulin IgE in diabetics. J Clin Endocrinol TJ 2004 Distinct idiotypes of insulin autoantibody in autoimmune
Metab 45:1159 –1164 polyendocrine syndrome type 2 and childhood onset type 1 dia-
167. Patterson R, Mellies CJ, Roberts M 1973 Immunologic reactions betes. J Clin Endocrinol Metab 89:5266 –5270
against insulin. II. IgE anti-insulin, insulin allergy and combined 189. Petersen KG, Storch MJ, Rother K, Licht T, Kerp L 1985 Insulin
IgE and IgG immunologic insulin resistance. J Immunol 110:1135– and anti-insulin antibody interaction: evidence for the formation of
1145 7 S and 10 S structures. Diabetes 34:799 – 802
168. Patterson R, Roberts M, Grammer LC 1990 Insulin allergy: re- 190. Schifferli JA, Taylor RP 1989 Physiological and pathological as-
evaluation after two decades. Ann Allergy 64:459 – 462 pects of circulating immune complexes. Kidney Int 35:993–1003
169. Grammer L 1986 Insulin allergy. Clin Rev Allergy 4:189 –200 191. Nagaoka K, Chinen Y, Takegami T, Nabeya N, Sakurami T, Imura
170. deShazo RD, Mather P, Grant W, Carrington D, Frentz JM, Lueg H, Kuno S, Tasaka K, Hamajima Y 1979 Circulating immune
M, Lauritano AA, Falholt K 1987 Evaluation of patients with local complexes in the serum of diabetes mellitus in childhood by a
reactions to insulin with skin tests and in vitro techniques. Diabetes modified 125I-C1q binding test. Endocrinol Jpn 26:527–532
Care 10:330 –336 192. Federlin K, Kelcorsky H, Maser E 1981 Clinical aspects of immu-
171. Lieberman P, Patterson R, Metz R, Ucena G 1971 Allergic reactions nity to insulin. In: Keck K, Erb P, eds. Basic and clinical aspects of
to insulin. JAMA 215:1106 –1112 immunity to insulin. New York: Walter de Gruyter; 250
172. Finco-Kent D, Krasner A, Williams D, Duggan W, Kawabata T 193. Hanauer A, Batson J 1961 Anaphylactic shock following insulin
2006 IgG subclass analysis of insulin antibodies in patients with injection: case report and review of the literature. Diabetes 10:105–
type 1 diabetes treated with inhaled human insulin (Exubera) and 109
subcutaneous insulin. Diabet Med 23(Suppl 4):325 (Abstract) 194. deShazo RD, Boehm TM, Kumar D, Galloway JA, Dvorak HF
173. Füchtenbusch M, Kredel K, Bonifacio E, Schnell O, Ziegler AG 1982 Dermal hypersensitivity reactions to insulin: correlations of
2000 Exposure to exogenous insulin promotes IgG1 and the T- three patterns to their histopathology. J Allergy Clin Immunol
helper 2-associated IgG4 responses to insulin but not to other islet 69:229 –237
autoantigens. Diabetes 49:918 –925 195. Heding LG, Falholt K, Kristensen JS 1984 IgE specific for insulin—
174. Brooks-Worrell BM, Nielson D, Palmer JP 1999 Insulin autoan- formation and clinical significance. In: Andreani D, Di Mario U,
tibodies and insulin antibodies have similar binding characteristics. Federlin KF, Heding LG, eds. Immunology in diabetes. London:
Proc Assoc Am Physicians 111:92–96 Kimpton; 235–244
175. Baxter RC, Yue DK, Turtle JR 1976 Equilibrium binding studies of 196. Arkins JA, Engbring NH, Lennon EJ 1962 The incidence of skin
insulin antibodies in diabetic subjects. Clin Chem 22:1089 –1094 reactivity to insulin diabetic patients. J Allergy 33:69 –72
176. Albert SG, Popp DA 1984 Hypoglycemia due to serum-complexed 197. Van Haeften TW, Gerich JE 1990 Complications of insulin therapy.
insulin in a patient with diabetes mellitus. Diabetes Care 7:285–290 In: Becker KL, ed. Principles and practices of endocrinology and
177. Kim MR, Sheeler LR, Mansharamani N, Haug MT, Faiman C, metabolism. Philadelphia: J. B. Lippincott Company; 1112–1118
Gupta MK 1997 Insulin antibodies and hypoglycemia in diabetic 198. Grammer LC, Chen PY, Patterson R 1983 Evaluation and man-
patients. Can a quantitative analysis of antibody binding predict agement of insulin allergy. J Allergy Clin Immunol 71:250 –254
the risk of hypoglycemia? Endocrine 6:285–291 199. Reeves WG, Allen BR, Tattersall RB 1980 Insulin-induced lipoat-
178. Van Haeften TW, Krom BA, Gerich JE 1987 Prolonged fasting rophy: evidence for an immune pathogenesis. Br Med J 280:1500 –
hypoglycemia due to insulin antibodies in patient with non-insu- 1503
lin-dependent diabetes mellitus: effect of insulin withdrawal on 200. Raile K, Noelle V, Landgraf R, Schwarz HP 2001 Insulin anti-
insulin-antibody-binding kinetics. Diabetes Care 10:160 –163 bodies are associated with lipoatrophy but also with lipohyper-
179. Goldman J, Baldwin D, Rubenstein AH, Klink DD, Blackard trophy in children and adolescents with type 1 diabetes. Exp Clin
WG, Fisher LK, Roe TF, Schnure JJ 1979 Characterization of cir- Endocrinol Diabetes 109:393–396
culating insulin and proinsulin-binding antibodies in autoimmune 201. deShazo RD 1978 Insulin allergy and insulin resistance: two im-
hypoglycemia. J Clin Invest 63:1050 –1059 munologic reactions. Postgrad Med 63:85–92
180. Eguchi Y, Uchigata Y, Yao K, Yokoyama H, Hirata Y, Omori Y 1994 202. Paley R, Turnbridge R 1952 Dermal reactions to insulin therapy.
Longitudinal changes of serum insulin concentration and insulin Diabetes 1:22–27
antibody features in persistent insulin autoimmune syndrome 203. Nakagawa S, Nakayama H, Sasaki T, Yoshino K, Yu YY 1973 A
(Hirata’s disease). Autoimmunity 19:279 –284 simple method for the determination of serum free insulin levels
181. Berzofsky JA, Berkower IJ, Epstein SL 1999 Antigen-antibody in insulin-treated patients. Diabetes 22:590 – 600
interactions and monoclonal antibodies. In: Paul WE, ed. Funda- 204. Sapin R 2003 Insulin assays: previously known and new analytical
mental immunology. Philadelphia: Lippincott-Raven Publishers; features. Clin Lab 49:113–121
75–110 205. Hayford JT, Thompson RG 1982 Free and total insulin integrated
182. Reeves WG 1983 Insulin antibody determination: theoretical and concentrations in insulin dependent diabetes. Metabolism 31:387–
practical considerations. Diabetologia 24:399 – 403 397
183. Heise T, Nosek L, Ronn BB, Endahl L, Heinemann L, Kapitza C, 206. Gray RS, Cowan P, Di Mario U, Elton RA, Clarke BF, Duncan LJ
Draeger E 2004 Lower within-subject variability of insulin detemir 1985 Influence of insulin antibodies on pharmacokinetics and bio-
in comparison to NPH insulin and insulin glargine in people with availability of recombinant human and highly purified beef insu-
type 1 diabetes. Diabetes 53:1614 –1620 lins in insulin dependent diabetics. Br Med J Clin Res Ed 290:1687–
184. Garcia CA, Prabakar KR, Diez J, Cao ZA, Allende G, Zeller M, 1691
Dogra R, Mendez A, Rosenkranz E, Dahl U, Ricordi C, Hanahan 207. Van Haeften TW, Bolli GB, Dimitriadis GD, Gottesman IS, Hor-
D, Pugliese A 2005 Dendritic cells in human thymus and periphery witz DL, Gerich JE 1986 Effect of insulin antibodies and their
display a proinsulin epitope in a transcription-dependent, capture- kinetic characteristics on plasma free insulin dynamics in patients
independent fashion. J Immunol 175:2111–2122 with diabetes mellitus. Metabolism 35:649 – 656
185. Padoa CJ, Crowther NJ, Thomas JW, Hall TR, Bekris LM, Torn C, 208. Francis AJ, Hanning I, Alberti KG 1985 The influence of insulin
Landin-Olsson M, Ortqvist E, Palmer JP, Lernmark A, Hampe CS antibody levels on the plasma profiles and action of subcutane-
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 649

ously injected human and bovine short acting insulins. Diabeto- 228. Rhie FH, Ganda OP, Bern MM, Soeldner JS, Gerson B, Azizi F
logia 28:330 –334 1981 Insulin resistance and monoclonal gammopathy. Metabolism
209. Peters A, Klose O, Hefty R, Keck F, Kerner W 1995 The influence 30:41– 45
of insulin antibodies on the pharmacokinetics of NPH insulin in 229. Renie A, Hamilton RG, Adkinson Jr NF, Rendell MS 1981 Hy-
patients with type 1 diabetes treated with human insulin. Diabet perlabile diabetes accompanied by insulin resistance. Clin Chem
Med 12:925–930 27:1463–1464
210. Van Haeften TW, Heiling VJ, Gerich JE 1987 Adverse effects of 230. Burrill K, Karam JH, Ching KN, Grodsky GM, Forsham PH 1971
insulin antibodies on postprandial plasma glucose and insulin pro- The use of fish insulin: a simple test to evaluate the role of insulin
files in diabetic patients without immune insulin resistance: im- antibodies in insulin resistance. Diabetes 20(Suppl 1):344 (Abstract)
plications for intensive insulin regimens. Diabetes 36:305–309 231. Kumar D, Miller LV 1970 Pork insulin resistance treated with
211. Waldhausl WK, Bratusch-Marrain P, Kruse V, Jensen I, Nowotny dealanated insulin. Diabetes 19(Suppl 1):392
P, Vierhapper H 1985 Effect of insulin antibodies on insulin phar- 232. Naquet P, Ellis J, Kenshole A, Semple JW, Delovitch TL 1989
macokinetics and glucose utilization in insulin-dependent diabetic Sulfated beef insulin treatment elicits CD8⫹ T cells that may ab-
patients. Diabetes 34:166 –173 rogate immunologic insulin resistance in type I diabetes. J Clin
212. Bolli G, de Feo P, Compagnucci P, Cartechini MG, Angeletti G, Invest 84:1479 –1487
Santeusanio F, Brunetti P, Gerich JE 1983 Abnormal glucose coun- 233. Karam JH, Grodsky GM, Forsham PH 1969 Insulin-resistant di-
terregulation in insulin-dependent diabetes mellitus: interaction of abetes with autoantibodies induced by exogenous insulin: success-
anti-insulin antibodies and impaired glucagon and epinephrine ful treatment by insulin withdrawal. Diabetes 18:445– 454
secretion. Diabetes 32:134 –141 234. Nathan DM, Axelrod L, Flier JS, Carr DB 1981 U-500 insulin in the
213. Bolli GB, Dimitriadis GD, Pehling GB, Baker BA, Haymond MW, treatment of antibody-mediated insulin resistance. Ann Intern Med
Cryer PE, Gerich JE 1984 Abnormal glucose counterregulation 94:653– 656
after subcutaneous insulin in insulin-dependent diabetes mellitus. 235. Rendell M, Slevin D, Meltz G, Simpson J, Barquet A 1979 A case
N Engl J Med 310:1706 –1711 of maturity-onset diabetes mellitus resistant to insulin but respon-
214. Gardner DF, Wilson HK, Podet EJ, Arakaki RF, Nell LJ, Thomas sive to tolbutamide. Ann Intern Med 90:195–197
JW, Crane MM, Field JB 1986 Prolonged action of regular insulin 236. Oakley W, Field JB, Sowton GE, Rigby B, Cunliffe AC 1959
in diabetic patients: lack of relationship to circulating insulin an- Action of prednisone in insulin-resistant diabetes. Br Med J 1:1601–
tibodies. J Clin Endocrinol Metab 62:621– 627 1606
215. Heinemann L 2002 Variability of insulin absorption and insulin 237. Micic D, Brkic S, Kendereski A, Popovic V, Zoric S, Nikolic JA,
action. Diabetes Technol Ther 4:673– 682 Igrutinovic L, Ivanoska D, Manojlovic D, Micic J 1993 Immuno-
216. Ziel FH, Davidson MB, Harris MD, Rosenberg CS 1988 The vari- logical resistance to human biosynthetic insulin– effects of immu-
ability in the action of unmodified insulin is more dependent on nosuppression and plasmapheresis. Diabetes Res Clin Pract 19:
changes in tissue insulin sensitivity than on insulin absorption. 83– 89
Diabet Med 5:662– 666 238. Merimee TJ 1965 Insulin resistance: study of effect of 6-mercap-
217. Grunfeld C 1984 Insulin resistance: pathophysiology, diagnosis, topurine. Lancet 42:69 –71
and therapeutic implications. Special Top Endocrinol Metabol 239. Matsuyoshi A, Shimoda S, Tsuruzoe K, Taketa K, Chirioka T,
6:193–240 Sakamoto F, Sakakida M, Miyamura N, Araki E 2006 A case of
218. Davidson JK, DeBra DW 1978 Immunologic insulin resistance. slowly progressive type 1 diabetes with unstable glycemic control
Diabetes 27:307–318 caused by unusual insulin antibody and successfully treated with
219. Witters LA, Ohman JL, Weir GC, Raymond LW, Lowell FC 1977 steroid therapy. Diabetes Res Clin Pract 72:238 –243
Insulin antibodies in the pathogenesis of insulin allergy and re- 240. Partamian J, Whitehouse F 1978 The role of insulin antibody (IAb)
sistance. Am J Med 63:703–709 affinity in insulin resistance (IR). Diabetes (Suppl 1):262 (Abstract)
220. Asai M, Kodera T, Ishizeki K, Uebori S, Kashiwaya T, Itoh H, 241. Armitage M, McCaughey E, Brooks A, Cove D, Betts P, Casey C,
Makino I 2003 Insulin lispro reduces insulin antibodies in a patient Wilkin TJ 1988 Insulin resistance and insulin antibodies: fact or
with type 2 diabetes with immunological insulin resistance. Dia- hallowed fiction? Pract Diabetes 5:200 –202
betes Res Clin Pract 61:89 –92 242. Tominaga M, Matsumoto M, Igarashi M, Eguchi H, Sekikawa A,
221. Hara K, Tobe K, Uchigata Y, Nakazono M, Yasuda K, Terauchi Y, Sasaki H 1992 Insulin antibody does not cause insulin resistance
Iwamoto Y, Akanuma Y, Kimura S, Kadowaki T 2000 Antibody- during glucose clamping in rats. Diabetes Res Clin Pract 18:143–151
mediated insulin resistance treated by cessation of insulin admin- 243. Thomas JW, Virta VJ, Nell LJ 1986 Idiotypic determinants on
istration. Intern Med 39:143–145 human anti-insulin antibodies are cyclically expressed. J Immunol
222. Jaeger C, Eckhard M, Brendel MD, Bretzel RG 2004 Diagnostic 137:1610 –1615
algorithm and management of immune-mediated complications 244. Moller DE, Flier JS 1991 Insulin resistance–mechanisms, syn-
associated with subcutaneous insulin therapy. Exp Clin Endocrinol dromes, and implications. N Engl J Med 325:938 –948
Diabetes 112:416 – 421 245. Lassmann-Vague V, SanMarco M, LeJeune PJ, Alessis C, Vague
223. Koyama R, Nakanishi K, Kato M, Yamashita S, Kuwahara H, P, Belicar P 1998 Autoimmunity and intraperitoneal insulin treat-
Katori H 2005 Hypoglycemia and hyperglycemia due to insulin ment by programmable pumps: lack of relationship. Diabetes Care
antibodies against therapeutic human insulin: treatment with dou- 21:2041–2044
ble filtration plasmapheresis and prednisolone. Am J Med Sci 329: 246. Bistritzer T, Sack J, Theodor R, Weissglass L, Ben Bassat I, Lahav
259 –264 M 1984 Correlation between HbA1c, purified insulins, diabetic
224. Lahtela JT, Knip M, Paul R, Antonen J, Salmi J 1997 Severe control, and insulin antibodies in diabetic children. Horm Res 20:
antibody-mediated human insulin resistance: successful treatment 178 –185
with the insulin analog lispro: a case report. Diabetes Care 20:71–73 247. Livneh A, Avraham H, Bistritzer T, Weisglass L, Theodor R, Sack
225. Miyoshi H, Yanigasawa K, Obara SKT 2003 Response to insulin J 1990 Deleterious effect of anti-insulin antibodies on diabetes con-
lispro in a case of diabetes complicated by liver cirrhosis that could trol. Isr J Med Sci 26:11–15
not be controlled with high-dose human insulin. J Japan Diab Soc 248. Moises RS, Sa JR, Chacra AR, Russo EM 1990 Relationship be-
46:145–149 tween insulin antibodies and metabolic control in type I diabetes
226. Mizuhashi S, Nakamura K, Mori Y, Noda M, Nakanishi K 2006 mellitus. Braz J Med Biol Res 23:1243–1252
Insulin allergy and immunologic insulin resistance caused by in- 249. Andersen OO 1975 The immunogenic properties of highly purified
terleukin-6 in a patient with lung cancer. Diabetes Care 29:1711– insulin preparations: the clinical importance of insulin-binding
1712 antibodies. Acta Endocrinol 78:723–735
227. Uriuhara A, Morita S, Kuroda S, Fumoto M, Araki S, Aoyama N, 250. Goswami R, Jaleel A, Kochupillai NP 2000 Insulin antibody re-
Amano K, Hayashi Y, Yokono K, Kasuga M 1994 Severe insulin- sponse to bovine insulin therapy: functional significance among
resistant diabetes mellitus associated with hypereosinophilic syn- insulin requiring young diabetics in India. Diabetes Res Clin Pract
drome. Intern Med 33:632– 636 49:7–15
650 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

251. Asplin C, Raghu P, Clemons P, Lyen K, Tatpati O, McKnight B, antibody and hormone dosage in non-resistant diabetes. Postgrad
Baker L, Guthrie R, Sperling M, Palmer J 1987 Randomized pro- Med J Suppl 819
spective trial of pure porcine and conventional bovine/porcine 272. Skom JH, Talmage DW 1958 The role of nonprecipitating insulin
insulin. Diabetes Care 10:337–342 antibodies in diabetes. J Clin Invest 37:787–793
252. Chen JW, Frystyk J, Lauritzen T, Christiansen JS 2005 Impact of 273. Haumont D, Dorchy H, Toussaint D, Despontin M 1982 Exoge-
insulin antibodies on insulin aspart pharmacokinetics and phar- nous insulin needs. Relationship with duration of diabetes, C-
macodynamics after 12-week treatment with multiple daily injec- peptidemia, insulin antibodies, and retinopathy. Helv Paediatr
tions of biphasic insulin aspart 30 in patients with type 1 diabetes. Acta 37:143–150
Eur J Endocrinol 153:907–913 274. Asplin CM, Hartog M, Goldie DJ 1978 Change of insulin dosage,
253. Daneman D, Fishman L, Clarson C 1987 Factors affecting insulin circulating free and bound insulin and insulin antibodies on trans-
antibody binding in children with insulin-dependent diabetes mel- ferring diabetics from conventional to highly purified porcine in-
litus. Clin Invest Med - Medecine Clinique et Experimentale 10: sulin. Diabetologia 14:99 –105
480 – 483 275. Mustaffa BE, Daggett PR, Nabarro JD 1977 Insulin binding ca-
254. Dozio N, Beretta A, Castiglioni M, Rosa S, Scavini M, Belloni C, pacity in patients changed from conventional to highly purified
Poloniato A 1996 Insulin antibodies do not preclude optimization insulins: an indicator of likely response. Diabetologia 13:311–315
of metabolic control in women with IDDM during pregnancy. 276. Davidson JK 1989 Transferring patients with insulin-dependent
Diabetes Care 19:979 –982 diabetes mellitus from animal-source insulins to recombinant DNA
255. Fernandez Castaner M, Perez M, Maravall J, Montana E, Soler J human insulin: clinical experience. Clinical Ther 11:319 –330
1996 Lack of relationship between insulin antibodies, metabolic 277. Fletcher JA, Barnett AH, Pyke DA, Volkmann HP, Hartog M,
control and insulin requirements in type 1 diabetes [letter; com- Perrett AD, Bhakri HL, Leatherdale B, Harrison HE 1990 Transfer
ment]. Diabet Med 13:686 – 687 from animal insulins to semisynthetic human insulin: a study in
256. Francis AJ, Home PD, Walford S, Alberti KG, Mann N, Reeves four centres. Diabetes Res 14:151–158
WG 1985 Prevalence of morning hyperglycaemia: determinants of 278. Lam HC, Ho LT, Tang KT, Wang LM, Kwok CF, Liu YF, Hsiao
fasting blood glucose concentrations in insulin-treated diabetics. LJ, Li SH 1989 Immunogenicity of highly purified porcine and
Diabet Med 2:89 –94 human insulins in diabetic patients with high titers of insulin an-
257. Gonen B, Goldman J, Baldwin D, Goldberg RB, Ryan WG, Blix tibody. Taiwan I Hsueh Hui Tsa Chih - J Formosan Med Assoc
PM, Schanzlin D, Fritz KJ, Rubenstein AH 1979 Metabolic control 88:346 –351
in diabetic patients. Effect of insulin-secretory reserve (measured 279. Makela AL, Akerblom HK, Makela P 1982 Insulin antibodies in
by plasma C-peptide levels) and circulating insulin antibodies. diabetic children treated with highly purified porcine NPH insulin.
Diabetes 28:749 –753 Comparison of results obtained by three methods of insulin anti-
258. Gray RS, Borsey DQ, Kurtz A, Rainbow S, Smith AF, Elton RA, body determination. Diabete Metab 8:341–348
Duncan LJ, Clarke BF 1981 Relationship of glycosylated haemo- 280. Wilson RM, Douglas CA, Tattersall RB, Reeves WG 1985 Immu-
globin to C-peptide secretory status and antibody binding of in- nogenicity of highly purified bovine insulin: a comparison with
sulin in insulin-dependent diabetes. Horm Metab Res 13:599 – 603 conventional bovine and highly purified human insulins. Diabe-
259. Hubinger A, Becker A, Gries FA 1988 Total insulin levels in type tologia 28:667– 670
1 diabetic patients with insulin antibodies and their effect on insulin 281. Hirata Y, Ishizu H, Ouchi N, Motomura S, Abe M, Hara Y, Waka-
requirement and metabolic control. Diabetes Res 7:65– 69 sugi H, Takahashi I, Sakani H, Tanaka M, Kawano H,. Kanesaka
260. Ionescu-Tirgoviste C, Mirodon Z, Cheta D, Santu E, Simionescu T 1970 Insulin autoimmunity in a case of spontaneous hypogly-
M, Nicolau A, Mincu I 1989 Comparative study of insulin anti- cemia. J Japan Diab Soc 13:312–320
bodies in diabetic patients with primary insulin-dependence (type 282. Uchigata Y, Kuwata S, Tsushima T, Tokunaga K, Miyamoto M,
I) and secondary insulin-dependence (type II). Medecine Interne Tsuchikawa K, Hirata Y, Juji T, Omori Y 1993 Patients with Graves
27:303–311 disease who developed insulin autoimmune syndrome (Hirata dis-
261. Kabadi UM, Birkenholz M 1992 Metabolic control following trans- ease) possess HLA-Bw62/Cw4/DR4 carrying DRB1*0406. J Clin
fer from mixed bovine-porcine insulin to human insulin in subjects Endocrinol Metab 77:249 –254
with IDDM: influence of the presence of insulin antibodies. Dia- 283. Burch HB, Clement S, Sokol MS, Landry F 1992 Reactive hypo-
betes Res 19:187–193 glycemic coma due to insulin autoimmune syndrome: case report
262. Sakata S, Matsuda M, Komaki T, Miura K 1986 Effect of anti- and literature review. Am J Med 92:681– 685
insulin antibodies on glycemic control in insulin treated diabetic 284. Takayama-Hasumi S, Eguchi Y, Sato A, Morita C, Hirata Y 1990
patients. Immunol Invest 15:791–799 Insulin autoimmune syndrome is the third leading cause of spon-
263. Virgili F, Frigato F, Magnanini PG, Coronel GA, Iavicoli M 1988 taneous hypoglycemic attacks in Japan. Diabetes Res Clin Pract
Reduction in insulin antibodies 42 months after transfer from por- 10:211–214
cine to human monocomponent insulins. Diabetes Res 9:19 –20 285. Hirata Y, Uchigata Y 1994 Insulin autoimmune syndrome in Japan.
264. Walford S, Allison SP, Reeves WG 1982 The effect of insulin Diabetes Res Clin Pract 24(Suppl):S153–S157
antibodies on insulin dose and diabetic control. Diabetologia 22: 286. Dozio N, Scavini M, Beretta A, Sarugeri E, Sartori S, Belloni C,
106 –110 Dosio F, Savi A, Fazio F, Sodoyez JC, Pozza G 1998 Imaging of the
265. Wredling R, Lins PE, Adamson U 1990 Prevalence of anti-insulin buffering effect of insulin antibodies in the autoimmune hypogly-
antibodies and its relation to severe hypoglycaemia in insulin- cemic syndrome. J Clin Endocrinol Metab 83:643– 648
treated diabetic patients. Scand J Clin Lab Invest 50:551–557 287. Basu A, Service FJ, Yu L, Heser D, Ferries LM, Eisenbarth G 2005
266. Asplin CM, Hartog M, Goldie DJ, Alberti KG, Smythe P, Binder Insulin autoimmunity and hypoglycemia in seven white patients.
C, Fabero OK 1978 A comparison between diabetics receiving a Endocr Pract 11:97–103
high or low daily insulin dosage. Horm Metab Res 10:365–369 288. Harwood R 1960 Insulin-binding antibodies and “spontaneous”
267. Kerp L, Kasemir H 1976 High and low affinity insulin antibodies. hypoglycemia. N Engl J Med 262:978 –979
Acta Endocrinol Suppl 205:211–222 289. Yasuda T, Ishikawa O, Ohigashi H, Furukawa H, Imaoka S,
268. Ludvigsson J 1984 Insulin antibodies in diabetic children treated Iwanaga T, Sasakuma F 1989 Hypoglycemia induced by insulin
with monocomponent porcine insulin from the onset: relationship antibody during postoperative management with intravenous hy-
to B-cell function and partial remission. Diabetologia 26:138 –141 peralimentation–a case report and qualitative analysis of insulin
269. Keilacker H, Rjasanowski I, Ziegler M, Michaelis D, Woltanski antibody. Nippon Geka Gakkai Zasshi 90:949 –952
KP, Besch W 1982 Insulin antibodies in juvenile diabetes mellitus: 290. Hayakawa T 1993 Fasting hypoglycemia and postprandial hyper-
correlations to diabetic stability, insulin requirement and duration glycemia due to insulin antibodies in a patient with non-insulin-
of insulin treatment. Horm Metab Res 14:227–232 dependent diabetes mellitus: evaluation by Scatchard analysis and
270. Martinka E, Strakova J, Mokan M 1998 Insulin antibodies in pa- euglycemic clamp study. J Japan Diab Soc 36:531–537
tients with type 2 diabetes mellitus. Vnitr Lek 44:577–581 291. Sakamoto N, Maruyama M, Tsuruoka A, Tajima N 2002 A type
271. Hurn BA, Farrant PC, Young BA, Grahame A 1969 Insulin-binding 1 diabetic patient with prolonged fasting hypoglycemia and post-
Fineberg et al. • Immunological Responses Endocrine Reviews, October 2007, 28(6):625– 652 651

prandial hyperglycemia due to insulin autoimmune syndrome-like ized trial of human versus animal species insulin in diabetic preg-
insulin antibodies. J Japan Diab Soc 45:471– 476 nant women: improved glycemic control, not fewer antibodies to
292. Suzuki K, Hirayama S, Ito S 1997 A case of a non-insulin depen- insulin, influences birth weight. Am J Obstet Gynecol 167:1325–
dent diabetic patient with regular spontaneous hypoglycemic at- 1330
tacks, which were due to insulin-binding antibodies induced by 313. Wellik SR, de Veciana M, Morgan MA, Berkowitz KM, Arquilla
human insulin therapy. Tohoku J Exp Med 182:163–173 ER 1995 Naturally occurring insulin autoantibodies in neonates of
293. Takahashi S, Oida K, Miyamori I 2000 A 75-year-old type 2 di- normal pregnancies and their relationship to insulinemia and birth
abetes mellitus case responding strikingly troglitazone: possible weight. Am J Obstet Gynecol 173:1878 –1884
mechanism of insulin resistance induced by insulin antibody. Nip- 314. Lindsay RS, Ziegler AG, Hamilton BA, Calder AA, Johnstone FD,
pon Ronen Igakkai Zasshi 37:344 –348 Walker JD 2004 Type 1 diabetes-related antibodies in the fetal
294. Cottrell DA, Henry ML, O’Dorisio TM, Tesi RJ, Ferguson RM, circulation: prevalence and influence on cord insulin and birth
Osei K 1991 Hypoglycemia after successful pancreas transplanta- weight in offspring of mothers with type 1 diabetes. J Clin Endo-
tion in type I diabetic patients. Diabetes Care 14:1111–1113 crinol Metab 89:3436 –3439
295. Tran MP, Larsen JL, Duckworth WC, Ruby EI, Miller SA, Frisbie 315. Weiss PA, Kainer F, Purstner P, Zehetleitner G, Huttner U, Haas
K, Taylor RJ, Stratta RJ 1994 Anti-insulin antibodies are a cause of J 1998 Anti-insulin antibodies and birth weight in pregnancies
hypoglycemia following pancreas transplantation. Diabetes Care complicated by diabetes. Early Hum Dev 53:145–154
17:988 –993 316. Schwartz R, Gruppuso PA, Petzold K, Brambilla D, Hiilesmaa V,
296. Asplin CM, Hartog M, Goldie DJ 1978 The relationship between Teramo KA 1994 Hyperinsulinemia and macrosomia in the fetus
circulating free and bound insulin, insulin antibodies, insulin dos- of the diabetic mother. Diabetes Care 17:640 – 648
age and diabetic control in insulin treated diabetics. Acta Endo- 317. 1992 Manual of clinical laboratory immunology. In: Rose N, ed. 4th
crinol (Copenh) 87:330 –338 ed. Washington, D.C.: American Society of Microbiology
297. Ionescu-Tirgoviste C, Mirodon Z, Paterache E, Cheta D, Mincu I 318. Dreyer M, for the Exubera Phase 3 Study Group 2004 Efficacy and
1991 No relationship between insulin antibodies and hypoglycemia two-year pulmonary safety of inhaled insulin as adjunctive therapy
in insulin-treated diabetic patients. Rom J Intern Med 29:189 –198 with metformin or glibenclamide in type 2 diabetes patients poorly
298. Kure M, Katsura Y, Kosano H, Noritake M, Watanabe T, Iwaki controlled with oral monotherapy. Diabetologia 47(Suppl 1):A44
Y, Nishigori H, Matsuoka T 2005 A trial to assess the amount of (Abstract)
insulin antibodies in diabetic patients by surface plasmon reso- 319. Andreev D, Ditzov S, Dashev G 1970 Diabetes-like vascular le-
nance. Intern Med 44:100 –106 sions in the kidneys of guinea pigs immunized with an insulin-
299. Edgerton DS, Cherrington AD, Williams P, Neal DW, Scott M, adjuvant mixture. Acta Diabetol Lat 7:243–259
Bowen L, Wilson W, Hobbs CH, Leach C, Kuo MC, Strack TR 2006 320. Andreani D, Di Mario U, Galfo C, Ventriglia L, Iavicoli M 1982
Inhalation of human insulin (exubera) augments the efficiency of Circulating immune complexes in diabetics with severe microan-
muscle glucose uptake in vivo. Diabetes 55:3604 –3610 giopathy: evaluation by two different methods. Acta Endocrinol
300. Sjobom NC, Adamson U, Lin PE 1989 The prevalence of impaired (Copenh) 99:239 –244
glucose counterregulation during an insulin-infusion test in insu- 321. Bodansky HJ, Wolf E, Cudworth AG, Dean BM, Nineham LJ,
lin-treated diabetic patients prone to severe hypoglycaemia. Dia- Bottazzo GF, Matthews JA, Kurtz AB, Kohner EM 1982 Genetic
betologia 32:818 – 825 and immunologic factors in microvascular disease in type I insulin-
301. Simister NE 2003 Placental transport of immunoglobulin G. Vac- dependent diabetes. Diabetes 31:70 –74
cine 21:3365–3369 322. Irvine WJ, Di Mario U, Guy K, Iavicoli M, Pozzilli P, Lumbroso
302. Mylvaganam R, Stowers JM, Steel JM, Wallace J, MacHendry JC, B, Andreani D 1978 Immune complexes and diabetic microangi-
Wright AD 1983 Insulin immunogenicity in pregnancy: maternal opathy. J Clin Lab Immunol 1:187–191
and fetal studies. Diabetologia 24:19 –25 323. Di Mario U, Borsey DQ, Contreas G, Prowse CV, Clarke BF,
303. Koczwara K, Bonifacio E, Ziegler AG 2004 Transmission of ma- Andreani D 1986 The relationship of soluble immune complexes,
ternal islet antibodies and risk of autoimmune diabetes in offspring insulin antibodies and insulin-anti-insulin complexes to platelet
of mothers with type 1 diabetes. Diabetes 53:1– 4 and coagulation factors in type 1 diabetic patients with and without
304. Gargiulo P, Di Mario U, Zuccarini O, Troili F, Tiberti C, Nicolini proliferative retinopathy. Clin Exp Immunol 65:57– 65
U, Pachi A, Gerlini G, Fallucca F 1986 Treatment of diabetic preg- 324. Triolo G, Giardina E, Rinaldi A, Bompiani GD 1984 IgA- and
nant women with monocomponent insulins. Acta Endocrinol insulin-containing (C3-fixing) circulating immune complexes in
Suppl (Copenh) 277:60 – 65 diabetes mellitus. Clin Immunol Immunopathol 30:169 –177
305. Heding LG, Persson B, Stangenberg M 1980 B-cell function in 325. Campbell LV, Charlesworth JA, Pasterfield GV, Jenkins A, Pus-
newborn infants of diabetic mothers. Diabetologia 19:427– 432 sell BA 1984 Does insulin administration contribute to immune
306. Murata K, Toyoda N, Sugiyama Y 1990 The effects of insulin complex formation in diabetes? Clin Exp Immunol 58:255–263
antibodies during diabetic pregnancy on newborn infants. Asia 326. Charles MA, Ong K, Slater L, Golden M, Arquilla ER 1982 Im-
Oceania J Obstet Gynaecol 16:115–122 munologic implications of insulin delivery and the role of immune
307. Riskin A, Haney PM Infant of a diabetic mother. Up to Date. complexes in diabetic sequelae. Diabetes Care 5(Suppl 1):88 –92
Available at: http://patients.uptodate.com/topic.asp?file⫽neonatol/ 327. Andersen OO 1976 Anti-insulin-antibodies and late diabetic com-
7268&title⫽Diabetes plications. Acta Endocrinol (Copenh) 83:329 –340
308. Evers IM, de Valk HW, Visser GH 2004 Risk of complications of 328. Virella G, Wohltmann H, Sagel J, Lopes-Virella MF, Kilpatrick
pregnancy in women with type 1 diabetes: nationwide prospective M, Phillips C, Colwell J 1981 Soluble immune complexes in pa-
study in the Netherlands. BMJ 328:915 tients with diabetes mellitus: detection and pathological signifi-
309. Dotta F, Gargiulo P, Tiberti C, Pachi A, Fallucca F, Andreani D, cance. Diabetologia 21:184 –191
Di Mario U 1987 Humoral and cellular immune abnormalities in 329. Triolo G, Davi G, Giardina E, Cardella F, Meli F, Strano A,
neonates of diabetic mothers: any pathological role? Exp Clin En- Bompiani GD 1989 The relationship of insulin antibodies, platelet-
docrinol 89:333–339 fixing immune complexes and platelet-associated IgG to in vitro
310. Fallucca F, Maldonato A, Iavicoli M, Di Rollo G, Di Biase N, platelet aggregation and thromboxane B2 synthesis in childhood
Napoli A, De Vecchis P, Sciullo E, Gerlini G, Pachi A 1989 In- type 1 (insulin-dependent) diabetes mellitus. Diabetes Res 10:1–5
fluence of maternal metabolic control and insulin antibodies on 330. Uchman B, Bang NU, Rathbun MJ, Fineberg NS, Davidson JK,
neonatal complications and B cell function in infants of diabetic Fineberg SE 1988 Effect of insulin immune complexes on human
mothers. Diabetes Res Clin Pract 7:277–284 blood monocyte and endothelial cell procoagulant activity. J Lab
311. Balsells M, Corcoy R, Mauricio D, Morales J, Garcia-Patterson A, Clin Med 112:652– 659
Carreras G, Puig-Domingo M, de Leiva A 1997 Insulin antibody 331. Miller K, Michael AF 1976 Immunopathology of renal extracel-
response to a short course of human insulin therapy in women with lular membranes in diabetes mellitus: specificity of tubular base-
gestational diabetes. Diabetes Care 20:1172–1175 ment-membrane immunofluorescence. Diabetes 25:701–708
312. Jovanovic-Peterson L, Kitzmiller JL, Peterson CM 1992 Random- 332. Deckert T, Egeberg J, Frimodt-Moller C, Sander E, Svejgaard A
652 Endocrine Reviews, October 2007, 28(6):625– 652 Fineberg et al. • Immunological Responses

1979 Basement membrane thickness, insulin antibodies and HLA- 352. Jarvela IY, Juutinen J, Koskela P, Hartikainen AL, Kulmala P,
antigens in long standing insulin dependent diabetics with and Knip M, Tapanainen JS 2006 Gestational diabetes identifies
without severe retinopathy. Diabetologia 17:91–96 women at risk for permanent type 1 and type 2 diabetes in fertile
333. Di Mario U, Ventriglia L, Iavicoli M, Guy K, Andreani D 1983 The age: predictive role of autoantibodies. Diabetes Care 29:607– 612
correlation between insulin antibodies and circulating immune 353. Jennings AM, Spencer KM, Dean BM, Wilson RM, Bottazzo GF,
complexes in diabetics with and without microangiopathy. Clin Ward JD 1989 Islet cell antibodies and insulin autoantibodies in
Exp Immunol 52:575–580 patients treated with oral hypoglycaemic agents. Diabet Med
334. Josephsen PG, Permin H, Juhl F, Enk B, Svehag SE 1986 Circu- 6:434 – 439
lating immune complexes, insulin antibodies, and deposits of im- 354. Nakanishi K, Kobayashi T, Sugimoto T, Murase T, Itoh T, Kosaka
munoglobulins in the skin in diabetes. Dan Med Bull 33:171–174 K 1988 Predictive value of insulin autoantibodies for further pro-
335. Kernell A, Ludvigsson J, Finnstrom K 1990 Vitreous fluoropho- gression of ␤ cell dysfunction in non-insulin-dependent diabetics.
tometry in juvenile diabetics with and without retinopathy in re- Diabetes Res 9:105–109
lation to metabolic control: insulin antibodies and c-peptide levels. 355. Nakayama M, Abiru N, Moriyama H, Babaya N, Liu E, Miao D,
Acta Ophthalmol (Copenh) 68:415– 420 Yu L, Wegmann DR, Hutton JC, Elliott JF, Eisenbarth GS 2005
336. Gray RS, Starkey IR, Rainbow S, Kurtz AB, Abdel-Khalik A, Prime role for an insulin epitope in the development of type 1
Urbaniak S, Elton RA, Duncan LJ, Clarke BF 1982 HLA antigens diabetes in NOD mice. Nature 435:220 –223
and other risk factors in the development of retinopathy in type 1 356. Kent SC, Chen Y, Bregoli L, Clemmings SM, Kenyon NS, Ricordi
diabetes. Br J Ophthalmol 66:280 –285 C, Hering BJ, Hafler DA 2005 Expanded T cells from pancreatic
337. Gilbey SG, Guy RJ, Jones H, Vergani D, Watkins PJ 1986 Diabetes lymph nodes of type 1 diabetic subjects recognize an insulin
and autonomic neuropathy: an immunological association? Diabet epitope. Nature 435:224 –228
Med 3:241–245 357. Nell LJ, Thomas JW 1983 The human immune response to insulin.
338. Scheinin T, Matikainen E, Groop L, Lepantalo M, Koskimies S, I. Kinetic and cellular aspects of lymphocyte proliferative responses
Kontiainen S 1988 HLA-antigens and immunity to insulin in in- in diabetics. J Immunol 131:701–705
sulin-dependent diabetics with or without diabetic neuropathy. 358. Faulk WP, Girard JP, Welscher HD 1975 Cell-mediated immunity
Scand J Clin Lab Invest 48:327–332 to insulin and its polypeptide chains in insulin-treated diabetics. Int
339. Bergamaschini L, Gardinali M, Poli M, Lorini R, Bosi E, Agostoni Arch Allergy Appl Immunol 48:364 –371
A 1991 Complement activation in diabetes mellitus. J Clin Lab 359. MacCuish AC, Jordan J, Campbell CJ, Duncan LJ, Irvine WJ 1975
Immunol 35:121–127 Cell-mediated immunity in diabetes mellitus; lymphocyte trans-
340. Berson SA, Yalow RS 1959 Quantitative aspects of the reaction formation by insulin and insulin fragments in insulin-treated and
between insulin and insulin-binding antibody. J Clin Invest 38: newly-diagnosed diabetes. Diabetes 24:36 – 43
1996 –2016
360. Mayer A, Rharbaoui F, Thivolet C, Orgiazzi J, Madec AM 1999
341. Honda M, Kawashima Y, Kawamura H, Fujikawa H, Kikuchi K,
The relationship between peripheral T cell reactivity to insulin,
Ohashi H, Mori Y, Miyakawa H, Ishibashi M 2006 Acute liver
clinical remissions and cytokine production in type 1 (insulin-
dysfunction complicated with uncontrollable glycemia due to in-
dependent) diabetes mellitus. J Clin Endocrinol Metab 84:2419 –
sulin antibody: successful treatment with glucocorticoid and lispro
2424
insulin. Intern Med 45:1225–1229
361. Dubois-LaForgue D, Carel JC, Bougneres PF, Guillet JG, Boitard
342. Davidson MB, Kumar D, Smith W 1991 Successful treatment of
C 1999 T-cell response to proinsulin and insulin in type 1 and
unusual case of brittle diabetes with sulfated beef insulin. Diabetes
Care 14:1109 –1110 pretype 1 diabetes. J Clin Immunol 19:127–134
343. Vaughan NJ, Matthews JA, Kurtz AB, Nabarro JD 1983 The bio- 362. Kurtz AB, Di Silvio L, Lydyard P 1985 Lymphocyte proliferation
availability of circulating antibody-bound insulin following insulin as a test of the immune response to insulin in diabetics. Diabetes
withdrawal in type 1 (insulin-dependent) diabetes. Diabetologia Res 2:175–178
24:355–358 363. Naquet P, Ellis J, Tibensky D, Kenshole A, Singh B, Hodges R,
344. Bolinger RE, Morris JH, McKnight FG, Diderich DA 1964 Dis- Delovitch TL 1988 T cell autoreactivity to insulin in diabetic and
appearance of I-131-labeled insulin from plasma as a guide to related non-diabetic individuals. J Immunol 140:2569 –2578
management of diabetes. N Engl J Med 270:767–770 364. Nell LJ, Virta VJ, Thomas JW 1985 Recognition of human insulin
345. Dixon K, Exon PD, Malins JM 1975 Insulin antibodies and the in vitro by T cells from subjects treated with animal insulins. J Clin
control of diabetes. Q J Med 44:543–553 Invest 76:2070 –2077
346. Montana E, Fernandez-Castaner M, Rosel P, Gomez JM, Vinzia 365. Diabetes Prevention Trial–Type 1 Diabetes Study Group 2002
C, Soler J 1990 The influence of insulin antibodies on metabolic Effects of insulin in relatives of patients with type 1 diabetes mel-
deterioration after interruption of continuous subcutaneous insulin litus. N Engl J Med 346:1685–1691
infusion. Diabete Metab 16:220 –225 366. Skyler JS, Krischer JP, Wolfsdorf J, Cowie C, Palmer JP, Green-
347. Dixon K, Exon PD, Hughes HR 1972 Insulin antibodies in aetiol- baum C, Cuthbertson D, Rafkin-Mervis LE, Chase HP, Leschek
ogy of labile diabetes. Lancet 1:343–347 E 2005 Effects of oral insulin in relatives of patients with type 1
348. Shima K, Tanaka R, Morishita S, Tarui S, Kumahara Y 1977 diabetes: The Diabetes Prevention Trial–Type 1. Diabetes Care 28:
Studies on the etiology of “brittle diabetes”. Relationship between 1068 –1076
diabetic instability and insulinogenic reserve. Diabetes 26:717–725 367. Harrison LC, Honeyman MC, Steele CE, Stone NL, Sarugeri E,
349. Krischer JP, Cuthbertson DD, Yu L, Orban T, Maclaren N, Jack- Bonifacio E, Couper JJ, Colman PG 2004 Pancreatic ␤-cell function
son R, Winter WE, Schatz DA, Palmer JP, Eisenbarth GS 2003 and immune responses to insulin after administration of intranasal
Screening strategies for the identification of multiple antibody- insulin to humans at risk for type 1 diabetes. Diabetes Care 27:
positive relatives of individuals with type 1 diabetes. J Clin En- 2348 –2355
docrinol Metab 88:103–108 368. Kobayashi T, Nakanishi K, Murase T, Kosaka K 1996 Small doses
350. Verge CF, Gianani R, Kawasaki E, Yu L, Pietropaolo M, Jackson of subcutaneous insulin as a strategy for preventing slowly pro-
RA, Chase HP, Eisenbarth GS 1996 Prediction of type I diabetes gressive ␤-cell failure in islet cell antibody-positive patients with
in first-degree relatives using a combination of insulin, GAD, and clinical features of NIDDM. Diabetes 45:622– 626
ICA512bdc/IA-2 autoantibodies. Diabetes 45:926 –933 369. Cutfield JF, Cutfield SM, Carne A, Emdin SO, Falkmer S 1986 The
351. Bock T, Pedersen CR, Josefsen K, Bottazzo GF, Palmer JP, Bus- isolation, purification and amino-acid sequence of insulin from the
chard K 1992 No risk of diabetes after insulin-shock treatment. teleost fish Cottus scorpius (daddy sculpin). Eur J Biochem
Lancet 339:1504 –1506 158:117–123

Endocrine Reviews is published by The Endocrine Society (http://www.endo-society.org), the foremost professional society serving the
endocrine community.

Вам также может понравиться