Abstract
Objective
Review prediction of Type 1 diabetes in light of current trials for prevention and preclinical novel therapist.
Methods
We estimate from islet autoantibody testing of random cadaveric donors that approximately ½ million individuals in the United States express multiple islet autoantibodies and are developing Type 1A (immune mediated) diabetes. It is now possible to predict not only risk for Type 1A diabetes but also the approximate age of diabetes onset of children followed from birth.
Results
In animal models diabetes can be prevented and some of the immunologic therapies effective in animal models are able to delay loss of insulin secretion in man.
Conclusion
Unfortunately none of the therapies studied to date in man can completely arrest progressive loss of insulin secretion from destruction of islet beta cells. Nevertheless current knowledge of pathogenesis (targeting trimolecular recognition complex: MHC- peptide- T cell receptor) and natural history combined with newer diagnostic methods allows accurate diagnosis and has stimulated the search for novel safe and effective preventive therapies.
Keywords: Islet, autoantibody, diabetes, beta cells
STAGES IN THE DEVELOPMENT OF TYPE 1A DIABETES
Following the discovery of cytoplasmic islet cell antibodies by Botazzo and Doniach in 1974(1) and studies of monozygotic twins(2), in 1986 we proposed a model of the development of Type 1A diabetes as a chronic autoimmune disorder that could be divided into stages beginning with genetic susceptibility, followed by triggering of autoimmunity marked by the appearance of islet autoantibodies followed by progressive loss of islet beta cells with subsequent metabolic abnormalities and then overt hyperglycemia and essentially complete loss of islet beta cells(3) (Figure 1). The model has stood the test of time with enhancements related to the development of improved assays for islet autoantibodies(4-7) and C-peptide(8), defining genetic loci underlying diabetes risk(9), definition of multiple methods to define progressive loss of beta cell function(10), and better understanding of Type 1 diabetes pancreatic pathology(11). The slowly progressive course of autoimmune diabetes is almost certainly due to lobular non-synchronous destruction of islet beta cells creating over time more and more pseudoatrophic islets (islets lacking all beta cells (insulin producing islet cells) but retaining islet cells producing glucagon, somatostatin, and pancreatic polypeptide)(12). In the major NOD mouse model of autoimmune diabetes, insulin is the primary autoantigen, as evidenced by the ability to prevent essentially all diabetes by mutating a single amino acid of insulin(13;14). It is very likely that insulin is also the primary target in man given the specific beta cell destruction (non-beta islet cells express GAD, IA-2, and ZnT8). The levels of insulin autoantibodies correlate with the rate of progression to diabetes(15) and a polymorphism of the human insulin gene is the second most important genetic determinant of Type 1 diabetes(16;17). The protective variant of the insulin gene is associated with increased expression of insulin in the thymus, likely leading to deletion of thymic autoreactive T cells targeting insulin. Insulin autoantibodies are almost always the first autoantibody to appear in children followed from birth to Type 1 diabetes(18), but of note the levels of insulin autoantibodies vary over the years prior to diabetes, suggesting that the rate of islet beta cell destruction may well vary over time (since mean levels correlate with rate of progression to diabetes(15)). With sensitive assays for C-peptide (C-peptide is secreted by beta cells in a 1:1 ratio with insulin) approximately 10% of patients with long-term Type 1 diabetes retain some insulin secreting cells(8) (concordant with pathology)(19) while approximately 2% may retain physiologically significant insulin secretion. Rare individuals even after 50 years of Type 1 diabetes have areas of the pancreas that appear normal, with other areas with complete islet beta cell destruction(19). Dr. Atkinson in Gainesville heads the JDRF sponsored nPOD program (website JDRFnPod) with on-line slides of pancreas sections obtained from cadaveric donors with type 1 diabetes of varied duration as well as cadaveric donors without diabetes with expression of multiple autoantibodies. Pancreas of multiple islet autoantibody positive cadaveric donors can have both insulitis and areas of pseudoatrophic islets consistent with chronic progressive immune mediated beta cell destruction(20).
Figure.

Stages Model of development of Type 1A (autoimmune) diabetes as a chronic autoimmune disorder with variation in the rate of beta cells destruction during progression to hyperglycemia when approximately 80% of islet beta cells (insulin secreting cells of the pancreas) have been destroyed.
Given current knowledge of the natural history of Type 1A diabetes it is possible to design trials for prevention at each of the stages of the disorder. The major genetic determinant of Type 1A diabetes are HLA alleles(9;21). In particular the highest risk Type 1A diabetes genotype (consists of heterozygous individuals with DR3-DQ2 and DR4-DQ8) has a risk approaching 5%. Approximately 30% of patients with Type 1A diabetes have this genotype compared to 2% of the general U.S. population. In that DQ, DR, DP and HLA class I alleles can readily be measured and influence risk trials in genetically at risk individuals are underway (e.g. Bonifacio Prepoint study) in individuals lacking islet autoantibodies but predicted to have a 50% risk of activating islet autoimmunity(22-25). There are now four major biochemically defined islet autoantibodies (autoantibodies to insulin, glutamic acid decarboxylates (GAD), insulinoma associated antigen (IA-2), and the islet zinc transporter (ZnT8). Commercial assays are now available for each of these autoantibodies. Expression of >=2 of these four autoantibodies (with high specificity assays= i.e. <=1/100 controls positive for each autoantibody) in children followed from birth is associated with 90% development of diabetes by 10 years and with the diabetes trend suggesting that with more time the risk might rise to 100%(26;27). Thus trials post-autoantibody expression are feasible(28) and conceptually analogous to the treatment of chronic active hepatitis prior to cirrhosis.
In an individual with characteristic Type 1 diabetes (e.g. childhood onset, non-obese) a single autoantibody is usually sufficient to confirm diagnosis of Type 1A diabetes. There are two major caveats. Insulin autoantibodies can develop post-insulin injection in non-autoimmune diabetes patients. Islet autoantibodies are lost with time, though approximately 50% of patients with Type 1A diabetes can retain islet autoantibodies for decades. Expression of multiple (≥2) islet autoantibodies (of insulin, GAD65, IA-2, or ZnT8) in an individual with diabetes is essentially diagnostic of Type 1A or autoimmune diabetes. Relying on multiple autoantibodies for diagnosis rather than a single islet autoantibody is especially important for patients with a clinical diagnosis of Type 2 diabetes. In questionable patients, HLA DR and DQ typing can aid in diagnosis but such typing is not definitive. For example the allele DQB1*0602 is highly protective for type 1A diabetes as it is found in 20% of general U.S. population but only 1% of children with autoantibody positive Type 1A diabetes.
Lack of islet autoantibodies can also be informative. More than 90% of children with new onset diabetes express one or more of the four major islet autoantibodies. An onset child lacking all four of the islet autoantibodies has a 10% risk of monogenic forms of diabetes(29). The child's prognosis depends on the specific mutated gene with some disorders worse than Type 1A diabetes (e.g. Wolfram syndrome) and some allowing treatment with oral hypoglycemic agents (HNF-4) or no treatment at all (glucokinase heterozygous mutations). Approximately ½ of African American and Hispanic American children lack islet autoantibodies and a major subset of these have a non-autoimmune form of diabetes where beta cells per islet are lost (Pathology Pattern B) and there are no pseudoatrophic islets (Pathology Pattern A)(11).
STATUS OF TRIALS
A very large number of therapies have failed to influence loss of beta cell function despite ability to prevent diabetes in autoimmune animal models(30). This includes nicotinamide, low-dose subcutaneous insulin, immunization with the antigen GAD in alum(31), the immunostimulant BCG vaccine(32), and oral or nasal insulin (to induce mucosal tolerance)(33). A subset of subjects with high levels of insulin autoantibodies receiving oral insulin in the DPT NIH prevention study appeared to have significant protection, (for the highest level of insulin autoantibodies as much as a seven year delay). A Trialnet study is underway to determine if such protection can be confirmed(28).
There are also several studies with documented delay of loss of beta cell function in trials at the onset of Type 1 diabetes. This includes studies utilizing cytoxan combined with anti-thymocyte globulin(34;35), anti-CD3(36) and anti-CD20(37) monoclonal antibodies, and CTLA-4IG(38). The most dramatic effects were seen in the Brazilian study of Cytoxan and anti-thymocyte globulin and GCSF, with the majority of the patients able to discontinue insulin and a subset off insulin for several years(34). This study was relatively small and lacked a placebo control group, with a concern that a subset of individuals might not have Type 1A diabetes. An obvious major concern is the utilization of relatively high dose cytoxan. Studies of anti-thymocyte globulin (ATG) and GCSF combined with ATG without cytoxan are underway. None of the other therapies delaying loss of C-peptide secretion had as dramatic an effect, potentially related to less effective immunosuppression. The chronic loss of beta cell function post-diabetes onset was typically delayed for 6 months to one year and then progressive loss resumed that paralleled loss of the placebo groups(39). It is noteworthy that the C-peptide curves were similar with multiple very different therapies. For instance the anti-CD3 antibodies are hypothesized to act by inducing regulatory tolerance(40) while the anti-CD20 antibodies dramatically deplete B-lymphocytes and both only delayed progressive loss of C-peptide secretion. To date the phase III trials of anti-CD3 antibodies have been disappointing in terms of not meeting clinical endpoints (e.g. HbA1c) despite multiple trials decreasing loss of C-peptide. This may relate to inadequate dosing and/or trial designs (e.g. HbA1c as endpoint with intensive insulin therapy in short-term diabetics, subjects from diverse countries mixed together). It may take combination therapy to impact progressive beta cell loss and such trials are planned(41).
CONCLUSION
I believe the status of current trials for the prevention of Type 1A diabetes (which includes trials preventing loss of beta cell function post-onset) suggest that altering the chronic progressive autoimmune killing of beta cells will not be “safely” achieved with broad targeting of the immune system. In addition, current insulin therapy of Type 1A diabetes is improving, particularly with development of continuous glucose monitoring devices that can turn off insulin pumps to limit severe hypoglycemia. This raises the bar for the safety of preventive immunologic therapies.
My epidemiology colleagues believe that the best prevention of Type 1A diabetes will come from elimination of triggering factors. For instance a large trial of removing cow's milk from the diet of infants is underway(42). Unfortunately triggering factors given the high risk for identical twins(43) are likely to be ubiquitous or potentially stochastic and have proven difficulty to identify. Nevertheless environmental factors influencing development of Type 1A diabetes certainly exist and are changing with the doubling of Type 1A diabetes incidence every 20 years(44).
For the prevention of Type 1A diabetes we are left with the need to better understand the specific molecular pathogenesis and to develop specifically targeted therapies. I believe therapies inducing tolerance to islet antigens or interrupting antigen presentation by specific HLA allele are most likely to lead to safe prevention.
The importance of regulatory T cells for the human immune system is undoubted(45). Children with mutations of the FoxP3 gene controlling development of a major subset of such T cells (IPEX syndrome) develop Type 1A diabetes in the first weeks of life (something essentially never seen in typical Type 1A diabetes).
Our studies have been pursuing small molecules that bind in the groove of specific MHC alleles. Blocking or enhancing presentation of target peptides (e.g. a fragment of insulin (B:9-23)) can prevent diabetes of NOD mice(46). Having molecular structures of relevant trimolecular complexes (target peptide, presenting MHC molecule and T cell receptors) recognizing the target peptide in specific MHC registers(47), can be utilized to design drugs to prevent diabetes. In rat models of type 1A diabetes a monoclonal antibody to germline encoded T cell receptor sequence (Vbeta 13) prevents diabetes(48).
At present for no autoimmune disorder is there an effective and safe antigen specific immunologic therapy. Thus it should be kept in mind that the field of the immunology of diabetes is pursuing what is currently impossible in man (though possible in animal models) and that success would likely have wide impact on multiple immunologic disorders.
Acknowledgements
This work was supported by grants from:
National Institute of Health: R01 DK 032083, U19AI050864, P30 DK 057516, NO1 AI 15416
The Juvenile Diabetes Research Foundation: International Autoimmunity Center
The Brehm Coalition
The Helmsley Foundation
The Children's Diabetes Foundation
Dr. Eisenbarth is on 2 University Provisional patents for treating autoimmunity with small molecules
There is also a research grant from Novartis in the same area
Reference List
- 1.Bottazzo GF, Florin-Christensen A, Doniach D. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet. 1974;2(7892):1279–83. doi: 10.1016/s0140-6736(74)90140-8. [DOI] [PubMed] [Google Scholar]
- 2.Srikanta S, Ganda OP, Eisenbarth GS, Soeldner JS. Islet cell antibodies and beta cell function in monozygotic triplets and twins initially discordant for Type I diabetes mellitus. N Engl J Med. 1983;308(6):322–5. doi: 10.1056/NEJM198302103080607. [DOI] [PubMed] [Google Scholar]
- 3.Eisenbarth GS. Type I diabetes mellitus. A chronic autoimmune disease. N Engl J Med. 1986;314:1360–8. doi: 10.1056/NEJM198605223142106. [DOI] [PubMed] [Google Scholar]
- 4.Palmer JP, Asplin CM, Clemons P, Lyen K, Tatpati O, Raghu PK, Paquette TL. Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science. 1983;222(4630):1337–9. doi: 10.1126/science.6362005. [DOI] [PubMed] [Google Scholar]
- 5.Baekkeskov S, Aanstoot H-J, Christgau S, Reetz A, Solimena M, Cascalho M, Folli F, Richter-Olesen H, DeCamilli P. Identification of the 64K autoantigen in insulin-dependent diabetes as the GABA-synthesizing enzyme glutamic acid decarboxylase. Nature. 1990;347(6289):151–6. doi: 10.1038/347151a0. [published erratum appears in Nature 1990 Oct 25;347(6295):782]
- 6.Wenzlau JM, Moua O, Sarkar SA, Yu L, Rewers M, Eisenbarth GS, Davidson HW, Hutton JC. SlC30A8 is a major target of humoral autoimmunity in type 1 diabetes and a predictive marker in prediabetes. Ann N Y Acad Sci. 2008 Dec;1150:256–9. doi: 10.1196/annals.1447.029. [DOI] [PubMed] [Google Scholar]
- 7.Lan MS, LU J, Goto Y, Notkins AL. Molecular cloning and identification of a receptor-type protein tyrosine phosphatase, IA-2, from human insulinoma. DNA Cell Biol. 1994;13(5):505–14. doi: 10.1089/dna.1994.13.505. [DOI] [PubMed] [Google Scholar]
- 8.Wang L, Lovejoy NF, Faustman DL. Persistence of Prolonged C-peptide Production in Type 1 Diabetes as Measured With an Ultrasensitive C-peptide Assay. Diab care. 2012 Mar;35(3):465–70. doi: 10.2337/dc11-1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Concannon P, Rich SS, Nepom GT. Genetics of type 1A diabetes. N Engl J Med. 2009 Apr 16;360(16):1646–54. doi: 10.1056/NEJMra0808284. [DOI] [PubMed] [Google Scholar]
- 10.Sosenko JM, Skyler JS, Krischer JP, Greenbaum CJ, Mahon J, Rafkin LE, Cuthbertson D, Cowie C, Herold K, Eisenbarth G, Palmer JP. Glucose excursions between states of glycemia with progression to type 1 diabetes in the diabetes prevention trial-type 1 (DPT-1). diab. 2010 Oct;59(10):2386–9. doi: 10.2337/db10-0534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gianani R, Campbell-Thompson M, Sarkar SA, Wasserfall C, Pugliese A, Solis JM, Kent SC, Hering BJ, West E, Steck A, Bonner-Weir S, Atkinson MA, Coppieters K, von HM, Eisenbarth GS. Dimorphic histopathology of long-standing childhood-onset diabetes. diabetol. 2010 Jan 9; doi: 10.1007/s00125-009-1642-y. [DOI] [PubMed] [Google Scholar]
- 12.Eisenbarth GS. Banting Lecture 2009: An Unfinished Journey: Molecular Pathogenesis to Prevention of Type 1A Diabetes. diab. 2010 Apr;59(4):759–74. doi: 10.2337/db09-1855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.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):220–3. doi: 10.1038/nature03523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vafiadis P, Bennett ST, Todd JA, Nadeau J, Grabs R, Goodyer CG, Wickramasinghe S, Colle E, Polychronakos C. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet. 1997;15:289–92. doi: 10.1038/ng0397-289. [DOI] [PubMed] [Google Scholar]
- 15.Steck AK, Johnson K, Barriga KJ, Miao D, Yu L, Hutton JC, Eisenbarth GS, Rewers MJ. Age of Islet Autoantibody Appearance and Mean Levels of Insulin, But Not GAD or IA-2 Autoantibodies, Predict Age of Diagnosis of Type 1 Diabetes: Diabetes Autoimmunity Study in the Young. Diab care. 2011 May 11; doi: 10.2337/dc10-2088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bell GI, Horita S, Karam JH. A polymorphic locus near the human insulin gene is associated with insulin-dependent diabetes mellitus. diab. 1984;33:176–83. doi: 10.2337/diab.33.2.176. [DOI] [PubMed] [Google Scholar]
- 17.Pugliese A, Zeller M, Fernandez A, Zalcberg LJ, Bartlett RJ, Ricordi C, Pietropaolo M, Eisenbarth GS, Bennett ST, Patel DD. The insulin gene is transcribed in the human thymus and transcription levels correlate with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type I diabetes. Nat Genet. 1997;15(3):293–7. doi: 10.1038/ng0397-293. [DOI] [PubMed] [Google Scholar]
- 18.Achenbach P, Koczwara K, Knopff A, Naserke H, Ziegler AG, Bonifacio E. Mature high-affinity immune responses to (pro)insulin anticipate the autoimmune cascade that leads to type 1 diabetes. J Clin Invest. 2004 Aug;114(4):589–97. doi: 10.1172/JCI21307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G, Bonner-Weir S, King GL. Residual insulin production and pancreatic ss-cell turnover after 50 years of diabetes: Joslin Medalist Study. diab. 2010 Nov;59(11):2846–53. doi: 10.2337/db10-0676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.In't VP, Lievens D, De Grijse J, Ling Z, van der AB, Pipeleers-Marichal M, Gorus F, Pipeleers D. Screening for insulitis in adult autoantibody-positive organ donors. Diabetes. 2007 Sep;56(9):2400–4. doi: 10.2337/db07-0416. [DOI] [PubMed] [Google Scholar]
- 21.Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, Mychaleckyj JC, Todd JA, Bonella P, Fear AL, Lavant E, Louey A, Moonsamy P. HLA DR-DQ Haplotypes and Genotypes and Type 1 Diabetes Risk: Analysis of the Type 1 Diabetes Genetics Consortium Families. Diabetes. 2008 Feb 5;57:1084–92. doi: 10.2337/db07-1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Baschal EE, Aly TA, Babu SR, Fernando MS, Yu L, Miao D, Barriga KJ, Norris JM, Noble JA, Erlich HA, Rewers MJ, Eisenbarth GS. HLA-DPB1*0402 Protects Against Type 1A Diabetic Autoimmunity in the Highest Risk DR3-DQB1*0201/DR4-DQB1*0302 DAISY Population. diab. 2007 May 18;56(9):2405–9. doi: 10.2337/db07-0029. [DOI] [PubMed] [Google Scholar]
- 23.Baschal EE, Eisenbarth GS. Extreme genetic risk for type 1A diabetes in the post-genome era. J Autoimmun. 2008 Aug;31(1):1–6. doi: 10.1016/j.jaut.2008.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Baschal EE, Baker PR, Eyring KR, Siebert JC, Jasinski JM, Eisenbarth GS. The HLAB*3906 allele imparts a high risk of diabetes only on specific HLA-DR/DQ haplotypes. diabetol. 2011 May 1; doi: 10.1007/s00125-011-2161-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Howson JM, Walker NM, Clayton D, Todd JA. Confirmation of HLA class II independent type 1 diabetes associations in the major histocompatibility complex including HLA-B and HLA-A. Diabetes Obes Metab. 2009 Feb;11(Suppl 1):31–45. doi: 10.1111/j.1463-1326.2008.01001.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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, and ICA512bdc/IA-2 autoantibodies. diab. 1996;45(7):926–33. doi: 10.2337/diab.45.7.926. [DOI] [PubMed] [Google Scholar]
- 27.Bingley PJ, Christie MR, Bonifacio E, Bonfanti R, Shattock M, Fonte M-T, Bottazzo G-F, Gale EAM. Combined analysis of autoantibodies improves prediction of IDDM in islet cell antibody-positive relatives. diab. 1994;43(11):1304–10. doi: 10.2337/diab.43.11.1304. [DOI] [PubMed] [Google Scholar]
- 28.Skyler JS. Type 1 diabetes mellitus: immune intervention. Int J Clin Pract Suppl. 2012 Feb;(175):56–61. doi: 10.1111/j.1742-1241.2011.02855.x. [DOI] [PubMed] [Google Scholar]
- 29.Shields BM, McDonald TJ, Ellard S, Campbell MJ, Hyde C, Hattersley AT. The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. diabetol. 2012 Jan 5; doi: 10.1007/s00125-011-2418-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Roep BO, Atkinson M, von HM. Satisfaction (not) guaranteed: re-evaluating the use of animal models of type 1 diabetes. Nat Rev Immunol. 2004 Dec;4(12):989–97. doi: 10.1038/nri1502. [DOI] [PubMed] [Google Scholar]
- 31.Wherrett DK, Bundy B, Becker DJ, DiMeglio LA, Gitelman SE, Goland R, Gottlieb PA, Greenbaum CJ, Herold KC, Marks JB, Monzavi R, Moran A, Orban T, Palmer JP, Raskin P, Rodriguez H, Schatz D, Wilson DM, Krischer JP, Skyler JS. Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet. 2011 Jul 23;378(9788):319–27. doi: 10.1016/S0140-6736(11)60895-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Klingensmith G, Allen H, Hayward A, Stohry L, Humber C, Jensen P, Simoes E, Chase HP. Vaccination with BCG at diagnosis does not alter the course of IDDM. Diabetes. 1997;46(Suppl 1):193A. Ref Type: Abstract. [Google Scholar]
- 33.Michels AW, Eisenbarth GS. Immune intervention in type 1 diabetes. Semin Immunol. 2011 Jun;23(3):214–9. doi: 10.1016/j.smim.2011.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Couri CE, Oliveira MC, Stracieri AB, Moraes DA, Pieroni F, Barros GM, Madeira MI, Malmegrim KC, Foss-Freitas MC, Simoes BP, Martinez EZ, Foss MC, Burt RK, Voltarelli JC. C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2009 Apr 15;301(15):1573–9. doi: 10.1001/jama.2009.470. [DOI] [PubMed] [Google Scholar]
- 35.Couri CE, Voltarelli JC. Stem cell-based therapies and immunomodulatory approaches in newly diagnosed type 1 diabetes. Curr Stem Cell Res Ther. 2011 Mar;6(1):10–5. doi: 10.2174/157488811794480690. [DOI] [PubMed] [Google Scholar]
- 36.Herold KC, Bluestone JA. Type 1 diabetes immunotherapy: is the glass half empty or half full? Sci Transl Med. 2011 Aug 10;3(95):95fs1. doi: 10.1126/scitranslmed.3002981. [DOI] [PubMed] [Google Scholar]
- 37.Pescovitz MD, Torgerson TR, Ochs HD, Ocheltree E, McGee P, Krause-Steinrauf H, Lachin JM, Canniff J, Greenbaum C, Herold KC, Skyler JS, Weinberg A. Effect of rituximab on human in vivo antibody immune responses. J Allergy Clin Immunol. 2011 Sep 9; doi: 10.1016/j.jaci.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Orban T, Bundy B, Becker DJ, DiMeglio LA, Gitelman SE, Goland R, Gottlieb PA, Greenbaum CJ, Marks JB, Monzavi R, Moran A, Raskin P, Rodriguez H, Russell WE, Schatz D, Wherrett D, Wilson DM, Krischer JP, Skyler JS. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet. 2011 Jul 30;378(9789):412–9. doi: 10.1016/S0140-6736(11)60886-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.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):166–73. doi: 10.1016/j.clim.2009.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bisikirska B, Colgan J, Luban J, Bluestone JA, Herold KC. TCR stimulation with modified anti-CD3 mAb expands CD8 T cell population and induces CD8CD25 Tregs. J Clin Invest. 2005 Oct 1;115(10):2904–13. doi: 10.1172/JCI23961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bresson D, von Herrath M. Moving towards efficient therapies in type 1 diabetes: to combine or not to combine? Autoimmun Rev. 2007 Apr;6(5):315–22. doi: 10.1016/j.autrev.2006.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Knip M, Virtanen SM, Seppa K, Ilonen J, Savilahti E, Vaarala O, Reunanen A, Teramo K, Hamalainen AM, Paronen J, Dosch HM, Hakulinen T, Akerblom HK. Dietary intervention in infancy and later signs of beta-cell autoimmunity. N Engl J Med. 2010 Nov 11;363(20):1900–8. doi: 10.1056/NEJMoa1004809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Redondo MJ, Jeffrey J, Fain PR, Eisenbarth GS, Orban T. Concordance for islet autoimmunity among monozygotic twins. N Engl J Med. 2008 Dec 25;359(26):2849–50. doi: 10.1056/NEJMc0805398. [DOI] [PubMed] [Google Scholar]
- 44.Dabelea D, Bell RA, D'Agostino RB, Jr., Imperatore G, Johansen JM, Linder B, Liu LL, Loots B, Marcovina S, Mayer-Davis EJ, Pettitt DJ, Waitzfelder B. Incidence of diabetes in youth in the United States. JAMA. 2007 Jun 27;297(24):2716–24. doi: 10.1001/jama.297.24.2716. [DOI] [PubMed] [Google Scholar]
- 45.Tang Q, Bluestone JA. The Foxp3+ regulatory T cell: a jack of all trades, master of regulation. Nat Immunol. 2008 Mar;9(3):239–44. doi: 10.1038/ni1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Michels AW, Ostrov DA, Zhang L, Nakayama M, Fuse M, McDaniel K, Roep BO, Gottlieb PA, Atkinson MA, Eisenbarth GS. Structure-Based Selection of Small Molecules to Alter Allele-Specific MHC Class II Antigen Presentation. J Immunol. 2011 Oct 31; doi: 10.4049/jimmunol.1100746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Stadinski B, Kappler J, Eisenbarth GS. Molecular targeting of islet autoantigens. Immunity. 2010 Apr 23;32(4):446–56. doi: 10.1016/j.immuni.2010.04.008. [DOI] [PubMed] [Google Scholar]
- 48.Liu Z, Cort L, Eberwine R, Herrmann T, Leif JH, Greiner DL, Yahalom B, Blankenhorn EP, Mordes JP. Prevention of Type 1 Diabetes in the Rat With an Allele-Specific Anti-T-Cell Receptor Antibody: Vbeta13 as a Therapeutic Target and Biomarker. diab. 2012 Feb 24; doi: 10.2337/db11-0867. [DOI] [PMC free article] [PubMed] [Google Scholar]
