Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 May 10.
Published in final edited form as: Curr Opin Endocrinol Diabetes Obes. 2011 Aug;18(4):235–240. doi: 10.1097/MED.0b013e32834803ae

2011 Update: Antigen Specific Therapy in Type 1 Diabetes

Aaron W Michels 1,, Matthias von Herrath 2
PMCID: PMC4016762  NIHMSID: NIHMS577694  PMID: 21844706

Abstract

Purpose of Review

Update on the clinical trials using antigen specific therapies in autoimmune diabetes.

Recent Findings

Type 1 diabetes is now a predictable disease with the measurement of islet autoantibodies, and the incidence is increasing dramatically. Safe and effective interventions are needed to stop the underlying autoimmune destruction of insulin producing beta cells. Beta cell antigens, insulin and glutamic acid decarboxylase, are being used to preserve endogenous insulin production in individuals with new onset diabetes and to prevent diabetes. The results of antigen specific immune intervention trials are reviewed and consideration is given to future directions for inducing tolerance in type 1 diabetes.

Summary

Antigen specific immune therapies act by enhancing regulatory T cell function, in animal models often locally and selectively in islets or pancreatic lymph nodes, while inhibiting effector T cells. This therapeutic pathway provides a safe treatment to preserve beta cell function in new onset diabetic individuals with the GAD-Alum vaccine being the most extensively studied therapy. Insulin is being used in many forms to prevent diabetes and stop the underlying autoimmune process. For the future, combination immune therapies targeting different pathways in the immune system will be needed to effectively induce sustained tolerance in type 1 diabetes.

Keywords: diabetes, autoimmunity, islet antigens, therapies

Introduction

Type 1A diabetes (T1D), the immune mediated form of diabetes, is a chronic autoimmune disease in which there is specific immune destruction of the insulin producing pancreatic β-cells. T cells as well as other mononuclear cells cause insulitis ultimately resulting in β-cell death, decreased insulin production, and a lifelong requirement for insulin therapy (1). Despite treatment with insulin therapy long-term complications, including nephropathy, retinopathy, neuropathy, and cardiovascular disease, can result (2; 3). Over the last two decades, the incidence of T1D has doubled especially in children less than five years of age (4; 5). The Diabetes Control and Complications Trial (DCCT) found that 20% of patients studied, who were within 5 years of diagnosis, had remaining insulin production (0.2–0.5 pmol/ml) (6); at this time immunologic intervention can potentially save beta cell function and reduce reliance on insulin administration. Even partial beta cell function is beneficial as patients that maintain endogenous insulin production have better metabolic control than those who rely solely on exogenous insulin (7), and improved metabolic control reduces the long-term complications from diabetes (8). Therapies that halt beta cell destruction can result in continued endogenous insulin production, greatly improving metabolic control, reducing hypoglycemia, and decreasing the prevalence of complications in T1D. Therapies aimed at altering the underlying autoimmune process in T1D are actively being investigated with monoclonal antibodies to anti-CD3 (912) and anti-CD20 (13) showing preservation of beta cell function after a year. Treatment with a single course of an anti-CD3 monoclonal antibody can result in endogenous insulin production five years later (14). However these therapies are not without side effects and concerns regarding immune suppression. Antigen specific therapies offer an excellent safety profile while still having efficacy, because they can either selectively eliminate islet-reactive T cells, or, alternatively or in addition, induce islet-specific regulatory T cells that can act locally within pancreatic lymph nodes or islets without affecting general immune competence. This review focuses on the mechanism of action of antigen specific therapies and those agents currently being studied in T1D.

Mechanism of Action

Antigen specific therapy involves the administration of islet proteins peptides during various stages of the disease pathogenesis. The currently recognized antigens in human T1D include insulin, proinsulin, B-chain of insulin, glutamic acid decarboxylase, and zinc transporter 8 (ZnT8) proteins (15). Autoantibodies directed towards these peptides can be used to assess disease risk (16; 17). The amount, route of administration, timing of therapy in the disease process, and combination with an adjuvant all contribute to the effectiveness of a given antigen therapy. Most of our understanding regarding the effects of antigen therapy on the immune system comes from studies in the non obese diabetic (NOD) mouse, a spontaneous model of autoimmune diabetes as well as antigen-induced selective transgenic models (18; 19). From these studies we presume that T1D is a T cell mediated disease, at least in its effector phase, in which T cells that produce inflammatory cytokines such as IFN-γ and TNFα in conjunction with innate immunity and other inflammatory factors (such as IL-1β) cause beta cell death (1; 20; 21). Adaptive and natural regulatory T cells can both oppose the actions of autoreactive effector T cells in animal models. Antigen specific therapy can enhance adaptive regulatory T cell responses in humans as it appears that intradermal administration of proinsulin peptide can lead to a peptide specific IL-10 response (22) (regulatory cytokine) similar to inducing tolerance in allergy (23; 24). This IL-10 specific response is also observed when insulin B chain is administered intramuscularly in Incomplete Freund’s Adjuvant (IFA) (25). IL-10 has anti-inflammatory properties, down regulating IFN-γ (26), and inducing T cells to become regulatory T cells in peripheral lymph nodes and lymphoid organs (27; 28). In addition, islet-antigen specific IL-10 responses were detected in healthy individuals while those who progressed to type 1 diabetes had both IFN-γ and IL-10 responses. Interestingly, the individuals with type 1 diabetes who had IL-10 responses developed disease later in life (>20 years of age), underlining the therapeutic potential of an IL-10 response to delay or prevent disease (29). In mice, for example after oral insulin administration, other cytokines such as IL-4 and TGF-beta can be induced (30; 31) – at present it is not fully known, which of these could also play a role after antigen-specific therapy in humans. The concept of preventing or treating a more TH1 mediated disease such as type 1 diabetes by deviating the immune system locally to TH2 still remains therefore rather attractive (32).

A potential major benefit of immunizing with antigens is the result of site-specific immunotherapy, because, at least in animal models, regulatory T cells only act where islet antigens are present, which limits their effect to pancreatic lymph nodes and the islets themselves (33; 34).

Insulin

There is a significant amount of evidence in the NOD mouse model and humans that insulin is a key target of beta-cell autoimmunity leading to T1D. A simple observation is that insulin is specific for beta cells and no other cells are destroyed in T1D, while other identified autoantigens such as GAD65, IA-2, and ZnT8 are not specific to beta cells (35). In the NOD mouse model, mutating a single amino acid in a specific insulin epitope (amino acids 9 to 23 in the B chain of insulin) recognized by T cells prevents all diabetes (36; 37). In humans the insulin gene is the second most important genetic determinant of T1D (polymorphisms of the MHC class II genes provide the most significant genetic risk). The protective genetic variant of insulin results in increased insulin expression in the thymus, likely resulting in deletion of autoreactive insulin T cells during T cell development (3840). Animal studies have shown that decreased insulin expression in the thymus during T cell development leads to increased insulin specific T cells and T1D onset (41).

There have been multiple studies utilizing insulin, insulin peptides, and proinsulin peptides to stop islet autoimmunity and prevent diabetes outlined in table 1 (22; 4245). Twenty years ago, the first diabetes prevention trial (DPT-1) used a low dose of daily oral insulin in order to prevent T1D. In this NIH sponsored study (TrialNet oral insulin) oral insulin was provided to first degree relatives of T1D patients with at least two islet autoantibodies in an attempt to prevent diabetes onset (17; 46). However, the administration of oral insulin did not delay progression to overt diabetes, but in post-hoc analysis individuals with high titer insulin autoanibodies benefited from treatment. In this group of patients, it was estimated that diabetes onset was delayed as much as five years (47). Since the analysis was a post-hoc subgroup analysis, a repeat oral insulin trial sponsored by TrialNet in individuals with multiple islet autoantibodies (including insulin autoantibodies) is currently underway (table 2) (42). One likely issue that negatively affected the success of the DPT-1 oral insulin trial is that, as compared to the mouse models, a very low insulin dose was used (1mg in mice versus 7.5 mg in humans per dose) and that the therapy was given daily, whereas less frequent dosing has shown to be more effective for some mucosal antigens in mouse models (48).

Table 1.

Completed antigen specific trials in type 1 diabetes

Stage of disease Study Title Agent Comment
Prevention TrialNet oral insulin Oral insulin Delay of overt diabetes onset in 1st degree relatives of T1D patients with high titer insulin autoantibodies
Prevention Intranasal insulin Nasal insulin No effect on delay/prevention of type 1 diabetes when given daily; ongoing trials are using less frequent administration of intranasal insulin
New onset intervention GAD vaccine GAD-alum Delay in loss of c-peptide in those with T1D < 6mo from baseline
New onset intervention HSP60 DiaPep277 Heat shock protein administered in several trials with limited efficacy; likely blocking innate immune system inflammation
Recent onset intervention Immunization with insulin B chain Insulin B chain + IFA Increased IL-10 producing T cells but no effect on c-peptide in a small pilot study
Intervention Proinsulin peptide immuno-therapy Intradermal proinsulin peptide Increased IL-10 production from peptide-specific T cells after 3 months of treatment

Table 2 .

Ongoing antigen therapy trials in type 1 diabetes

Study Title Agent Phase Patient Population Clinicaltrials.gov
*TrialNet Oral Insulin Oral Insulin 3 ≥2 islet autoantibodies (including insulin) for T1D prevention NCT00419562
*Intranasal insulin for the prevention of T1D Intranasal Insulin 2 Prevention for at risk individuals NCT00223613
NCT00336674
TrialNet GAD GAD-alum 2 New onset T1D NCT00529399
Diamyd GAD GAD-alum 3 New onset T1D NCT00723411
Study of BHT-3021 in T1D subjects Proinsulin DNA Vaccine 1/2 New onset T1D NCT00453375
*DiaPep277 in New Onset Adults DiaPep277 3 New onset T1D NCT01103284
*

indicates trials currently enrolling at the time of publication

In addition to insulin, immunization with the insulin B chain in IFA has been used in T1D patients, showing induction of IL-10 producing regulatory T cells, but no effect on C-peptide preservation (25). Peakman and coworkers are studying peptides of proinsulin, recognized by autoreactive T cells, to induce regulatory responses and ultimately tolerance to insulin, however no clinical benefit has been noted as of yet (22). Alternative routes of insulin dosing include intranasal insulin administration, where two initial trials did not yield positive results (44), likely because the nasal insulin had been administered too frequently (48). Bayhill Therapeutics developed an intramuscular proinsulin DNA vaccine, which is currently in a phase 2 trial to stop islet autoimmunity in new onset T1D patients with some encouraging initial benefits on C-peptide preservation (ADA disclosure last year).

GAD-Alum

The most extensively studied antigen specific therapy to date is the GAD-Alum vaccine. The initial study was done in individuals with latent autoimmune diabetes of adulthood (LADA) showing preservation of c-peptide with a single dose. Dose finding studies followed, showing that only a specific dose of 20ug was effective. Notable is the fact the both lower and higher doses were not efficacious, again indicating the importance of dose in antigen specific therapy (49). The underlying mechanistic reasons for this dose dependency are unknown, which is a major obstacle in defining key biomarkers that could predict the efficacy of therapy on an individual basis. In a double blind, randomized placebo controlled trial, GAD-Alum injection delayed the loss of c-peptide production in new onset T1D children and adolescents following a single course of therapy. Follow up at 30 months showed a significant preservation of insulin secretion only in those individuals that received the GAD-Alum vaccine within 6 months of T1D diagnosis (50). Similar to the immune suppressive therapies anti-CD3 monoclonal antibodies and rituximab (anti-CD20 monoclonal antibody targeting B cells), one year after therapy loss of c-peptide secretion resumed at a rate analogous to the control groups. In these trials, there was evidence of an immunologic effect with the induction of GAD65 specific regulatory T cells and with B lymphocytes (increased GAD autoantibodies) (51). Recently, four year data has been reported with GAD-Alum treatment demonstrating significantly better preserved fasting c-peptide in GAD treated subjects. The safety profile after four years is excellent revealing no treatment related adverse events (52) and larger phase 2/3 trials are underway (table 2).

Future Directions

Antigen specific therapies have many beneficial effects with regards to safety profiles and specificity in the mechanism of action. At the present time, more antigen specific therapies need to be tested in a rationale way as the GAD-Alum vaccine and the post-hoc analyzed subgroup of the oral insulin trial are the only approaches showing efficacy so far. Since the dose and route of administration appear to be crucial parameters, smaller proof-of-concept trials might be important to sort out such factors more rapidly and, at the same time, define key biomarkers. One such biomarker might be the existence of autoantibodies to a given islet antigen: As proposed by Harrison (53) and recently validated in a wet-lab based on valid in silico predictions by Entelos (Bresson et al. unpublished), individuals who generate autoantibodies to an islet antigen might be more prone to also make regulatory (for example TH2 like) responses to the same antigen. Thus, there is now technology available provided by Entelos to use in silicocomputer aided, methods to assess dose and immune response in animal models with the potential to expand this technology to human disease (54). It will then also be useful to identify new epitopes of insulin and the newly discovered zinc transporter 8 and used in individuals with those specific antibodies. Last, the use and selection of adjuvants is also important and those adjuvants that enhance protective immune responses need to be considered with antigen specific therapy (i.e. IFA, Alum etc).

To prevent and ultimately cure T1D, a combination therapy approach will most likely be needed. The understanding of the immunopathogenesis in type 1 diabetes has increased considerably over the last several years (1) but clinical immune intervention is in the initial stages. We are now at a point in the immunotherapy field reminiscent of oncology developing the first chemotherapeutic agents, which now provide excellent five year survival rates for numerous malignancies. At the current time, there are no FDA approved therapies to block the autoimmune process in type 1 diabetes. However, there are now several single agent therapies (anti-CD3 monoclonal antibodies, rituximab, and GAD-Alum) that can delay c-peptide loss in newly diagnosed individuals with type 1 diabetes but none that sustain long lasting tolerance. There are currently many more single agent therapies under investigation (55). However, it is unlikely that we will be able to administer systemically acting immune modulators repeatedly and for a long duration of time to consistently delete autoimmune memory as well as newly formed self-reactive effector cells, but we are now at the point of combining successful single agents into suitable combination therapies with islet antigens. Work in preclinical animal models has demonstrated synergy with such combined therapies. For example, anti-CD3 monoclonal antibodies paired with intranasal insulin were able to reverse diabetes better than either single agent alone in a murine model of type 1 diabetes (56). Recently the Immune Tolerance Network (ITN) and Juvenile Diabetes Research Foundation (JDRF) partnered to make recommendations for developing combination immunotherapies in type 1 diabetes. In all, over 40 possible combinations of therapies could be considered (57). Combined therapies provide the benefits of synergy with the potential to lower efficacious doses which will lessen the side effects from long-term immune suppression. We favor providing an antigen specific therapy under the umbrella of an immune suppressive therapy such as anti-CD3 or an anti-inflammatory agent (IL-1 monoclonal antibody or receptor antagonist) currently under investigation (58; 59).

Conclusions

The field of immune intervention in type 1 diabetes is at an exciting time with the potential to prevent and induce tolerance in diabetes. Antigen specific therapies act by inducing adaptive regulatory T cells that then act locally at the site of insulitis in the pancreas or the pancreatic lymph nodes. Clinically, the GAD-Alum vaccine has shown efficacy in delaying the loss of endogenous insulin production and oral insulin in an antibody positive subset of patients as well as a pro-insulin expressing DNA vaccine in a small number of individuals have provided encouraging data while all having an excellent safety profile. In the future, antigen-specific therapies have the potential to become alone-standing therapies when given early enough to prevent T1D in those at risk. In addition, combination therapies with systemically acting immune modulators (either anti-inflammatory or anti-T cell) targeting different pathways in the disease pathogenesis, raise the level of optimism over the next decade.

Key points.

  • Antigen specific therapies selectively eliminate islet-reactive T cells and induce regulatory T cells that can act locally within pancreatic lymph nodes or islets without affecting general immune competence.

  • Oral insulin may delay the onset of type 1 diabetes up to seven years in first degree relatives of type 1 diabetic individuals with high titer insulin autoantibodies.

  • Immunization with GAD-Alum preserves insulin secretion (measured by c-peptide) after 30 months in individuals that received the vaccine within 6 months of type 1 diabetes diagnosis.

  • In the future, antigen specific therapies have the potential to become stand alone therapies when given early enough to prevent type 1 diabetes in those at risk.

Acknowledgements

This work was supported by grants from the National Institutes of Diabetes & Digestive & Kidney Diseases U01 Prevention Center Award for MGVH, the Juvenile Diabetes Foundation (17-2010-744) for AWM, and the Brehm Coalition, and the Children’s Diabetes Foundation.

Footnotes

The authors declare no conflicts of interest.

Contributor Information

Aaron W. Michels, Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Court, MS B140, PO BOX 6511, Aurora, CO 80045, Aaron.Michels@ucdenver.edu, Phone: 303-724-6812, Fax: 303-724-6839.

Matthias von Herrath, Center for Type 1 Diabetes, La Jolla Institute for Allergy and Immunology, La Jolla, California, matthias@lial.org.

Reference List

  • 1. Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010 Apr 29;464(7293):1293–1300. doi: 10.1038/nature08933. ** Excellent review on the current understanding of the immunopathogenesis of type 1 diabetes and future directions in the field to prevent and cure diabetes.
  • 2.Maahs DM, Rewers M. Editorial: Mortality and renal disease in type 1 diabetes mellitus--progress made, more to be done. J Clin Endocrinol Metab. 2006 Oct;91(10):3757–3759. doi: 10.1210/jc.2006-1730. [DOI] [PubMed] [Google Scholar]
  • 3.Steffes MW, Sibley S, Jackson M, Thomas W. Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diab care. 2003 Mar;26(3):832–836. doi: 10.2337/diacare.26.3.832. [DOI] [PubMed] [Google Scholar]
  • 4.Harjutsalo V, Sjoberg L, Tuomilehto J. Time trends in the incidence of type 1 diabetes in Finnish children: a cohort study. Lancet. 2008 May 24;371(9626):1777–1782. doi: 10.1016/S0140-6736(08)60765-5. [DOI] [PubMed] [Google Scholar]
  • 5. Patterson CC, Dahlquist GG, Gyurus E, Green A, Soltesz G. Incidence trends for childhood type 1 diabetes in Europe during 1989–2003 and predicted new cases 2005–20: a multicentre prospective registration study. Lancet. 2009 Jun 13;373(9680):2027–2033. doi: 10.1016/S0140-6736(09)60568-7. ** This study showed the increasing incidence of type 1 diabetes in a Finnish population, especially in young children.
  • 6.Effect of intensive therapy on residual beta-cell function in patients with type 1 diabetes in the diabetes control and complications trial. A randomized, controlled trial. The Diabetes Control and Complications Trial Research Group. Ann Intern Med. 1998 Apr 1;128(7):517–523. doi: 10.7326/0003-4819-128-7-199804010-00001. [DOI] [PubMed] [Google Scholar]
  • 7.Grajwer LA, Pildes RS, Horwitz DL, Rubenstein AH. Control of juvenile diabetes mellitus and its relationship to endogenous insulin secretion as measured by C-peptide immunoreactivity. J Pediatr. 1977 Jan;90(1):42–48. doi: 10.1016/s0022-3476(77)80762-2. [DOI] [PubMed] [Google Scholar]
  • 8.The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. New Engl J Med. 1993;329:977–986. doi: 10.1056/NEJM199309303291401. [DOI] [PubMed] [Google Scholar]
  • 9.Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, Gitelman SE, Harlan DM, Xu D, Zivin RA, Bluestone JA. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med. 2002 May 30;346(22):1692–1698. doi: 10.1056/NEJMoa012864. [DOI] [PubMed] [Google Scholar]
  • 10.Herold KC, Gitelman SE, Masharani U, Hagopian W, Bisikirska B, Donaldson D, Rother K, Diamond B, Harlan DM, Bluestone JA. A Single Course of Anti-CD3 Monoclonal Antibody hOKT3{gamma}1(Ala-Ala) Results in Improvement in C-Peptide Responses and Clinical Parameters for at Least 2 Years after Onset of Type 1 Diabetes. Diabetes. 2005 Jun;54(6):1763–1769. doi: 10.2337/diabetes.54.6.1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Keymeulen B, Vandemeulebroucke E, Ziegler AG, Mathieu C, Kaufman L, Hale G, Gorus F, Goldman M, Walter M, Candon S, Schandene L, Crenier L, De Block C, Seigneurin JM, De Pauw P, Pierard D, Weets I, Rebello P, Bird P, Berrie E, Frewin M, Waldmann H, Bach JF, Pipeleers D, Chatenoud L. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med. 2005 Jun 23;352(25):2598–2608. doi: 10.1056/NEJMoa043980. [DOI] [PubMed] [Google Scholar]
  • 12. Keymeulen B, Walter M, Mathieu C, Kaufman L, Gorus F, Hilbrands R, Vandemeulebroucke E, Van d, V, Crenier L, De BC, Candon S, Waldmann H, Ziegler AG, Chatenoud L, Pipeleers D. Four-year metabolic outcome of a randomised controlled CD3-antibody trial in recent-onset type 1 diabetic patients depends on their age and baseline residual beta cell mass. diabetol. 2010 Jan 14; doi: 10.1007/s00125-009-1644-9. * This study provides 4 year data with one of the anti-CD3 monoclonal antibodies used in a recent onset type 1 diabetes immune intervention study.
  • 13. Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, Becker DJ, Gitelman SE, Goland R, Gottlieb PA, Marks JB, McGee PF, Moran AM, Raskin P, Rodriguez H, Schatz DA, Wherrett D, Wilson DM, Lachin JM, Skyler JS. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med. 2009 Nov 26;361(22):2143–2152. doi: 10.1056/NEJMoa0904452. ** The first study using an anti-CD20 monoclonal antibody to treat new onset type 1 diabetics, delays the loss of endogenous c-peptide.
  • 14. Herold KC, Gitelman S, Greenbaum C, Puck J, Hagopian W, Gottlieb P, Sayre P, Bianchine P, Wong E, Seyfert-Margolis V, Bourcier K, Bluestone JA. Treatment of patients with new onset Type 1 diabetes with a single course of anti-CD3 mAb Teplizumab preserves insulin production for up to 5 years. Clin Immunol. 2009 Aug;132(2):166–173. doi: 10.1016/j.clim.2009.04.007. * This study provides 5 year data with one of the anti-CD3 monoclonal antibodies used in a new onset type 1 diabetes immune intervention study.
  • 15.Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb P, Rewers M, Eisenbarth GS, Jensen J, Davidson HW, Hutton JC. The cation efflux transporter ZnT8 (Slc30A8) is a major autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A. 2007 Oct 23;104(43):17040–17045. doi: 10.1073/pnas.0705894104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Verge CF, Gianani R, Kawasaki E, Yu L, Pietropaolo M, Jackson RA, Chase HP, Eisenbarth GS. Prediction of type I diabetes in first-degree relatives using a combination of insulin, GAD, ICA512bdc/IA-2 autoantibodies. Diabetes. 1996;45(7):926–933. doi: 10.2337/diab.45.7.926. [DOI] [PubMed] [Google Scholar]
  • 17.Sherr J, Sosenko J, Skyler JS, Herold KC. Prevention of type 1 diabetes: the time has come. Nat Clin Pract Endocrinol Metab. 2008 Apr 29; doi: 10.1038/ncpendmet0832. [DOI] [PubMed] [Google Scholar]
  • 18.Liu E, Yu L, Moriyama H, Eisenbarth GS. Animal models of insulin-dependent diabetes. Methods Mol Med. 2004;102:195–212. doi: 10.1385/1-59259-805-6:195. [DOI] [PubMed] [Google Scholar]
  • 19.Yang Y, Santamaria P. Lessons on autoimmune diabetes from animal models. Clin Sci (Lond) 2006 Jun;110(6):627–639. doi: 10.1042/CS20050330. [DOI] [PubMed] [Google Scholar]
  • 20.Sanda S, Bollyky J, Standifer N, Nepom G, Hamerman JA, Greenbaum C. Short-term IL-1beta blockade reduces monocyte CD11b integrin expression in an IL-8 dependent fashion in patients with type 1 diabetes. Clin Immunol. 2010 Aug;136(2):170–173. doi: 10.1016/j.clim.2010.04.009. [DOI] [PubMed] [Google Scholar]
  • 21.Wachlin G, Augstein P, Schroder D, Kuttler B, Kloting I, Heinke P, Schmidt S. IL-1 beta, IFN-gamma and TNF-alpha increase vulnerability of pancreatic beta cells to autoimmune destruction. Journal of Autoimmunity. 2003;20(4):303–312. doi: 10.1016/s0896-8411(03)00039-8. [DOI] [PubMed] [Google Scholar]
  • 22. Thrower SL, James L, Hall W, Green KM, Arif S, Allen JS, Van-Krinks C, Lozanoska-Ochser B, Marquesini L, Brown S, Wong FS, Dayan CM, Peakman M. Proinsulin peptide immunotherapy in type 1 diabetes: report of a first-in-man Phase I safety study. Clin Exp Immunol. 2009 Feb;155(2):156–165. doi: 10.1111/j.1365-2249.2008.03814.x. ** This study reports the use of intradermal proinsulin to induce peptide specific T cells producing IL-10 in patients with established type 1 diabetes.
  • 23.Campbell JD, Buckland KF, McMillan SJ, Kearley J, Oldfield WL, Stern LJ, Gronlund H, van HM, Reynolds CJ, Boyton RJ, Cobbold SP, Kay AB, Altmann DM, Lloyd CM, Larche M. Peptide immunotherapy in allergic asthma generates IL-10-dependent immunological tolerance associated with linked epitope suppression. J Exp Med. 2009 Jul 6;206(7):1535–1547. doi: 10.1084/jem.20082901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Verhoef A, Alexander C, Kay AB, Larche M. T cell epitope immunotherapy induces a CD4+ T cell population with regulatory activity. PLoS Med. 2005 Mar;2(3):e78. doi: 10.1371/journal.pmed.0020078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Orban T, Farkas K, Jalahej H, Kis J, Treszl A, Falk B, Reijonen H, Wolfsdorf J, Ricker A, Matthews JB, Tchao N, Sayre P, Bianchine P. Autoantigen-specific regulatory T cells induced in patients with type 1 diabetes mellitus by insulin B-chain immunotherapy. J Autoimmun. 2009 Nov 18; doi: 10.1016/j.jaut.2009.10.005. * Study with the insulin B chain administered in IFA to recent onset individuals with type 1 diabetes. IL-10 producing T cells resulted but no preservation of c-peptide in a small pilot study.
  • 26.Zhu H, Qiu W, Lei P, Zhou W, Wen X, He F, Li L, Dai H, Shen G, Gong F. IL-10 gene modified dendritic cells inhibit T helper type 1-mediated alloimmune responses and promote immunological tolerance in diabetes. Cell Mol Immunol. 2008 Feb;5(1):41–46. doi: 10.1038/cmi.2008.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Battaglia M, Gregori S, Bacchetta R, Roncarolo MG. Tr1 cells: from discovery to their clinical application. Semin Immunol. 2006 Apr;18(2):120–127. doi: 10.1016/j.smim.2006.01.007. [DOI] [PubMed] [Google Scholar]
  • 28.Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 2006 Aug;212:28–50. doi: 10.1111/j.0105-2896.2006.00420.x. [DOI] [PubMed] [Google Scholar]
  • 29.Arif S, Tree TI, Astill TP, Tremble JM, Bishop AJ, Dayan CM, Roep BO, Peakman M. Autoreactive T cell responses show proinflammatory polarization in diabetes but a regulatory phenotype in health. J Clin Invest. 2004 Feb;113(3):451–463. doi: 10.1172/JCI19585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.von Herrath MG, Dyrberg T, Oldstone MB. Oral insulin treatment suppresses virus-induced antigen-specific destruction of beta cells and prevents autoimmune diabetes in transgenic mice. J Clin Invest. 1996 Sep 15;98(6):1324–1331. doi: 10.1172/JCI118919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Weiner HL. Oral tolerance, an active immunologic process mediated by multiple mechanisms. J Clin Invest. 2000 Oct;106(8):935–937. doi: 10.1172/JCI11348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Peakman M, von HM. Antigen-specific immunotherapy for type 1 diabetes: maximizing the potential. diab. 2010 Sep;59(9):2087–2093. doi: 10.2337/db10-0630. ** Well written review on antigen specific therapies in type 1 diabetes and future directions in the field.
  • 33.Tang Q, Bluestone JA. The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol. 2008 Mar;9(3):239–244. doi: 10.1038/ni1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Homann D, Holz A, Bot A, Coon B, Wolfe T, Petersen J, Dyrberg TP, Grusby MJ, von Herrath MG. Autoreactive CD4+ T cells protect from autoimmune diabetes via bystander suppression using the IL-4/Stat6 pathway. Immunity. 1999 Oct;11(4):463–472. doi: 10.1016/s1074-7613(00)80121-1. [DOI] [PubMed] [Google Scholar]
  • 35.Michels AW, Nakayama M. 2The anti-insulin trimolecular complex in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes. 2010 May 19; doi: 10.1097/MED.0b013e32833aba41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Nakayama M, Abiru N, Moriyama H, Babaya N, Liu E, Miao D, Yu L, Wegmann DR, Hutton JC, Elliott JF, Eisenbarth GS. Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature. 2005 May 12;435(7039):220–223. doi: 10.1038/nature03523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Concannon P, Rich SS, Nepom GT. Genetics of type 1A diabetes. N Engl J Med. 2009 Apr 16;360(16):1646–1654. doi: 10.1056/NEJMra0808284. [DOI] [PubMed] [Google Scholar]
  • 38.Pugliese A, Fernandez A, Zeller M, Zalcberg LJ, Ricordi C, Pietropaolo M, Eisenbarth GS, Bennett ST, Patel DD. Ectopic transcription of IDDM-associated islet cell autoantigen genes and monoallelic expression of the insulin gene in human thymus. 1996 submitted. [Google Scholar]
  • 39.Wicklow BA, Polychronakos C. Insulin auto-immunity: implications for the prevention of Type 1 diabetes mellitus. Expert Rev Clin Immunol. 2009 Jan;5(1):55–62. doi: 10.1586/1744666X.5.1.55. [DOI] [PubMed] [Google Scholar]
  • 40.Barratt BJ, Payne F, Lowe CE, Hermann R, Healy BC, Harold D, Concannon P, Gharani N, McCarthy MI, Olavesen MG, McCormack R, Guja C, Ionescu-Tirgoviste C, Undlien DE, Ronningen KS, Gillespie KM, Tuomilehto-Wolf E, Tuomilehto J, Bennett ST, Clayton DG, Cordell HJ, Todd JA. Remapping the Insulin Gene/IDDM2 Locus in Type 1 Diabetes. diab. 2004 Jul;53(7):1884–1889. doi: 10.2337/diabetes.53.7.1884. [DOI] [PubMed] [Google Scholar]
  • 41.Fan Y, Rudert WA, Grupillo M, He J, Sisino G, Trucco M. Thymus-specific deletion of insulin induces autoimmune diabetes. EMBO J. 2009 Sep 16;28(18):2812–2824. doi: 10.1038/emboj.2009.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mahon JL, Sosenko JM, Rafkin-Mervis L, Krause-Steinrauf H, Lachin JM, Thompson C, Bingley PJ, Bonifacio E, Palmer JP, Eisenbarth GS, Wolfsdorf J, Skyler JS. The TrialNet Natural History Study of the Development of Type 1 Diabetes: objectives, design, and initial results. Pediatr Diabetes. 2009 Apr;10(2):97–104. doi: 10.1111/j.1399-5448.2008.00464.x. [DOI] [PubMed] [Google Scholar]
  • 43.Skyler JS, Brown D, Chase HP, Collier E, Cowie C, Eisenbarth GS, Fradkin J, Grave G, Greenbaum C, Jackson RA, Kaufman FR, Krischer JP, Marks JB, Palmer JP, Ricker A, Schatz DA, Wilson D, Winter WE, Wolfsdorf J, Zeidler A, Dickler H, Eastman RC, Maclaren NK, Malone JI, Robertson PR et al. Effects of insulin in relatives of patients with type 1 diabetes mellitus. New Engl J Med. 2002;346(22):1685–1691B. doi: 10.1056/NEJMoa012350. [DOI] [PubMed] [Google Scholar]
  • 44.Nanto-Salonen K, Kupila A, Simell S, Siljander H, Salonsaari T, Hekkala A, Korhonen S, Erkkola R, Sipila JI, Haavisto L, Siltala M, Tuominen J, Hakalax J, Hyoty H, Ilonen J, Veijola R, Simell T, Knip M, Simell O. Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial. Lancet. 2008 Nov 15;372(9651):1746–1755. doi: 10.1016/S0140-6736(08)61309-4. [DOI] [PubMed] [Google Scholar]
  • 45.Walter M, Philotheou A, Bonnici F, Ziegler AG, Jimenez R. No effect of the altered peptide ligand NBI-6024 on beta-cell residual function and insulin needs in new-onset type 1 diabetes. Diab care. 2009 Nov;32(11):2036–2040. doi: 10.2337/dc09-0449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Skyler JS. Update on worldwide efforts to prevent type 1 diabetes. Ann N Y Acad Sci. 2008 Dec;1150:190–196. doi: 10.1196/annals.1447.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Skyler JS, Krischer JP, Wolfsdorf J, Cowie C, Palmer JP, Greenbaum C, Cuthbertson D, Rafkin-Mervis LE, Chase HP, Leschek E. Effects of oral insulin in relatives of patients with type 1 diabetes: The Diabetes Prevention Trial--Type 1. Diab care. 2005 May;28(5):1068–1076. doi: 10.2337/diacare.28.5.1068. [DOI] [PubMed] [Google Scholar]
  • 48. Fousteri G, Chan JR, Zheng Y, Whiting C, Dave A, Bresson D, Croft M, von HM. Virtual optimization of nasal insulin therapy predicts immunization frequency to be crucial for diabetes protection. diab. 2010 Dec;59(12):3148–3158. doi: 10.2337/db10-0561. ** This study demonstrates the use of computer aided (in silico) systems to predict the frequency of intranasal insulin administration is crucial for the prevention type 1 diabetes.
  • 49.Agardh CD, Cilio CM, Lethagen A, Lynch K, Leslie RD, Palmer M, Harris RA, Robertson JA, Lernmark A. Clinical evidence for the safety of GAD65 immunomodulation in adult-onset autoimmune diabetes. J Diabetes Complications. 2005 Jul;19(4):238–246. doi: 10.1016/j.jdiacomp.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 50.Ludvigsson J, Faresjo M, Hjorth M, Axelsson S, Cheramy M, Pihl M, Vaarala O, Forsander G, Ivarsson S, Johansson C, Lindh A, Nilsson NO, Aman J, Ortqvist E, Zerhouni P, Casas R. GAD Treatment and Insulin Secretion in Recent-Onset Type 1 Diabetes. N Engl J Med. 2008 Oct;Aug;359:1909–1920. doi: 10.1056/NEJMoa0804328. [DOI] [PubMed] [Google Scholar]
  • 51.Hjorth M, Axelsson S, Ryden A, Faresjo M, Ludvigsson J, Casas R. GAD-alum treatment induces GAD(65)-specific CD4(+)CD25(high)FOXP3(+) cells in type 1 diabetic patients. Clin Immunol. 2010 Oct 31; doi: 10.1016/j.clim.2010.10.004. [DOI] [PubMed] [Google Scholar]
  • 52. Ludvigsson J, Hjorth M, Cheramy M, Axelsson S, Pihl M, Forsander G, Nilsson NO, Samuelsson BO, Wood T, Aman J, Ortqvist E, Casas R. Extended evaluation of the safety and efficacy of GAD treatment of children and adolescents with recent-onset type 1 diabetes: a randomised controlled trial. diabetol. 2011 Mar;54(3):634–640. doi: 10.1007/s00125-010-1988-1. ** Five year results of the GAD-alum intervention in adolescents shows an excellent safety profile and individuals retaining the ability to produce c-peptide when treated within 6 months of diagnosis.
  • 53.Wentworth JM, Fourlanos S, Harrison LC. Reappraising the stereotypes of diabetes in the modern diabetogenic environment. Nat Rev Endocrinol. 2009 Sep;5(9):483–489. doi: 10.1038/nrendo.2009.149. [DOI] [PubMed] [Google Scholar]
  • 54. Shoda L, Kreuwel H, Gadkar K, Zheng Y, Whiting C, Atkinson M, Bluestone J, Mathis D, Young D, Ramanujan S. The Type 1 Diabetes PhysioLab Platform: a validated physiologically based mathematical model of pathogenesis in the non-obese diabetic mouse. Clin Exp Immunol. 2010 Aug;161(2):250–267. doi: 10.1111/j.1365-2249.2010.04166.x. ** This study reports the creation of an in silico program to virtually test immune intervention strategies in the non-obese diabetic (NOD) mouse model of diabetes.
  • 55. Skyler JS. Immunomodulation for type 1 diabetes mellitus. Int J Clin Pract Suppl. 2010 Feb;(166):59–63. doi: 10.1111/j.1742-1241.2009.02280.x. ** Immune modulation trials in type 1 diabetes are briefly reviewed from 1/2008 to 6/2009.
  • 56.Bresson D, Togher L, Rodrigo E, Chen YL, Bluestone JA, Herold KC, von Herrath M. Anti-CD3 and nasal proinsulin combination therapy enhances remission from recent-onset autoimmune diabetes by inducing Tregs. Journal of Clinical Investigation. 2006;116(5):1371–1381. doi: 10.1172/JCI27191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Matthews JB, Staeva TP, Bernstein PL, Peakman M, von HM. Developing combination immunotherapies for type 1 diabetes: recommendations from the ITN-JDRF Type 1 Diabetes Combination Therapy Assessment Group. Clin Exp Immunol. 2010 May;160(2):176–184. doi: 10.1111/j.1365-2249.2010.04153.x. ** Recommendations from the JDRF and ITN regarding the use of combination immune therapies in type 1 diabetes. This should be read by those with an interest in the field of immunotherapy.
  • 58.Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007 Apr 12;356(15):1517–1526. doi: 10.1056/NEJMoa065213. [DOI] [PubMed] [Google Scholar]
  • 59.Strom TB, Koulmanda M. Cytokine related therapies for autoimmune disease. Curr Opin Immunol. 2008 Dec;20(6):676–681. doi: 10.1016/j.coi.2008.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES