Primary prevention of beta-cell autoimmunity and type 1 diabetes - The Global Platform for the Prevention of Autoimmune Diabetes (GPPAD) perspectives

A G Ziegler, T Danne, D B Dunger, R Berner, R Puff, W Kiess, G Agiostratidou, J A Todd, E Bonifacio, A G Ziegler, T Danne, D B Dunger, R Berner, R Puff, W Kiess, G Agiostratidou, J A Todd, E Bonifacio

Abstract

Objective: Type 1 diabetes can be identified by the presence of beta-cell autoantibodies that often arise in the first few years of life. The purpose of this perspective is to present the case for primary prevention of beta-cell autoimmunity and to provide a study design for its implementation in Europe.

Methods: We examined and summarized recruitment strategies, enrollment rates, and outcomes in published TRIGR, FINDIA and BABYDIET primary prevention trials, and the TEDDY intensive observational study. A proposal for a recruitment and implementation strategy to perform a phase II/III primary prevention randomized controlled trial in infants with genetic risk for developing beta-cell autoimmunity is outlined.

Results: Infants with a family history of type 1 diabetes (TRIGR, BABYDIET, TEDDY) and infants younger than age 3 months from the general population (FINDIA, TEDDY) were enrolled into these studies. All studies used HLA genotyping as part of their eligibility criteria. Predicted beta-cell autoimmunity risk in the eligible infants ranged from 3% (FINDIA, TEDDY general population) up to 12% (TRIGR, BABYDIET). Amongst eligible infants, participation was between 38% (TEDDY general population) and 97% (FINDIA). Outcomes, defined as multiple beta-cell autoantibodies, were consistent with predicted risks. We subsequently modeled recruitment into a randomized controlled trial (RCT) that could assess the efficacy of oral insulin treatment as adapted from the Pre-POINT pilot trial. The RCT would recruit infants with and without a first-degree family history of type 1 diabetes and be based on general population genetic risk testing. HLA genotyping and, for the general population, genotyping at additional type 1 diabetes susceptibility SNPs would be used to identify children with around 10% risk of beta-cell autoimmunity. The proposed RCT would have 80% power to detect a 50% reduction in multiple beta-cell autoantibodies by age 4 years at a two-tailed alpha of 0.05, and would randomize around 1160 infants to oral insulin or placebo arms in order to fulfill this. It is estimated that recruitment would require testing of between 400,000 and 500,000 newborns or infants.

Conclusion: It is timely and feasible to establish a platform for primary prevention trials for type 1 diabetes in Europe. This multi-site European infrastructure would perform RCTs, supply data coordination and biorepository, provide cohorts for mechanistic and observational studies, and increase awareness for autoimmune diabetes.

Keywords: Antigen-based immunotherapy; Beta-cell autoimmunity; Prevention; Type 1 diabetes.

Figures

Figure 1
Figure 1
CD4+ T cell immune response profiles against insulin from the Pre-POINT trial . Multivariate gene expression analysis of insulin-responsive CD4+ T cells isolated from Pre-POINT participants with CD4+ T cell responses to insulin. The data show t-distributed Stochastic Neighbor Embedding (tSNE) analysis of pre-processed Ct values for all analyzed genes. A linear model was used to correct for confounding effects, which can mask relevant biological variability. Batch effects (dummy coding each plate/batch) were modeled jointly with dose effects to obtain a corrected gene expression data set. This resulted in two distinct clusters, with one cluster consisting of cells derived from children receiving 67.5 mg insulin (green symbols) and the second cluster consisting of cells derived from children receiving placebo (blue symbols), 2.5 mg insulin (red symbols), 7.5 mg insulin (yellow symbols), and 22.5 mg insulin (purple symbols). C1, C2, and C3 are component 1, component 2, and component 3 of the tSNE. Reproduced from reference #9 with permission from JAMA American Medical Association (License Number 3786541433624).
Figure 2
Figure 2
Design of GPPAD randomized controlled trial.

References

    1. Ziegler A.G., Rewers M., Simell O., Simell T., Lempainen J., Steck A. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. The Journal of the American Medical Association. 2013;309:2473–2479.
    1. Ziegler A.G., Nepom G.T. Prediction and pathogenesis in type 1 diabetes. Immunity. 2010;32:468–478.
    1. Harrison L.C. Vaccination against self to prevent autoimmune disease: the type 1 diabetes model. Immunology & Cell Biology. 2008;89:139–145.
    1. Harrison L.C., Hafler D.A. Antigen-specific therapy for autoimmune disease. Current Opinion in Immunology. 2000;12:704–711.
    1. Takiishi T., Korf H., Van Belle T.L., Robert S., Grieco F.A., Caluwaerts S. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. Journal of Clinical Investigation. 2012;122:1717–1725.
    1. Streeter H.B., Rigden R., Martin K.F., Scolding N.J., Wraith D.C. Preclinical development and first-in-human study of ATX-MS-1467 for immunotherapy of MS. Neurol Neuroimmunol Neuroinflamm. 2015;2:e93.
    1. Du Toit G., Roberts G., Sayre P.H., Bahnson H.T., Radulovic S., Santos A.F., LEAP Study Team Randomized trial of peanut consumption in infants at risk for peanut allergy. The New England Journal of Medicine. 2015;372:803–813.
    1. Lutterotti A., Yousef S., Sputtek A., Stürner K.H., Stellmann J.P., Breiden P. Antigen-specific tolerance by autologous myelin peptide-coupled cells: a phase 1 trial in multiple sclerosis. Science Translational Medicine. 2013;5 188ra75.
    1. Bonifacio E., Ziegler A.G., Klingensmith G., Schober E., Bingley P.J., Rottenkolber M., The Pre-POINT study group Effects of high dose oral insulin on immune responses in children at high risk for type 1 diabetes: the Pre-POINT randomized clinical trial. The Journal of the American Medical Association. 2015;313:1541–1549.
    1. Knip M., Åkerblom H.K., Becker D., Dosch H.M., Dupre J., Fraser W., TRIGR Study Group Hydrolyzed infant formula and early β-cell autoimmunity: a randomized clinical trial. The Journal of the American Medical Association. 2014;311:2279–2287.
    1. Hummel S., Pflüger M., Hummel M., Bonifacio E., Ziegler A.G. Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study. Diabetes Care. 2011;34:1301–1305.
    1. Norris J.M., Barriga K., Klingensmith G., Hoffman M., Eisenbarth G.S., Erlich H.A. Timing of initial cereal exposure in infancy and risk of islet autoimmunity. The Journal of the American Medical Association. 2003;290:1713–1720.
    1. Ziegler A.G., Schmid S., Huber D., Hummel M., Bonifacio E. Early infant feeding and risk of developing type 1 diabetes-associated autoantibodies. The Journal of the American Medical Association. 2003;290:1721–1728.
    1. Vaarala O., Ilonen J., Ruohtula T., Pesola J., Virtanen S.M., Härkönen T. Removal of bovine insulin from cow's milk formula and early initiation of beta-cell autoimmunity in the FINDIA pilot study. Archives of Pediatrics and Adolescent Medicine. 2012;166:608–614.
    1. The TEDDY Study Group The environmental determinants of diabetes in the young (TEDDY) study: study design. Pediatric Diabetes. 2007;8:286–298.
    1. Zhang Z.J., Davidson L., Eisenbarth G., Weiner H.L. Suppression of diabetes in nonobese diabetic mice by oral administration of porcine insulin. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:10252–10256.
    1. Skyler J.S., Krischer J.P., Wolfsdorf J., Cowie C., Palmer J.P., Greenbaum C. Effects of oral insulin in relatives of patients with type 1diabetes: the diabetes prevention trial-type 1. Diabetes Care. 2005;28:1068–1076.
    1. Ziegler A.G., Hummel M., Schenker M., Bonifacio E. Autoantibody appearance and risk for development of childhood diabetes in offspring of parents with type 1 diabetes: the 2-year analysis of the German BABYDIAB Study. Diabetes. 1999;48:460–468.
    1. Ilonen J., Hammais A., Laine A.P., Lempainen J., Vaarala O., Veijola R. Patterns of β-cell autoantibody appearance and genetic associations during the first years of life. Diabetes. 2013;62:3636–3640.
    1. Nakayama M., Abiru N., Moriyama H., Babaya N., Liu E., Miao D. Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice. Nature. 2005;435:220–223.
    1. Barratt B.J., Payne F., Lowe C.E., Hermann R., Healy B.C., Harold D. Remapping the insulin gene/IDDM2 locus in type 1 diabetes. Diabetes. 2004;53:1884–1889.
    1. Walter M., Albert E., Conrad M., Keller E., Hummel M., Ferber K. IDDM2/insulin VNTR modifies risk conferred by IDDM1/HLA for development of type 1 diabetes and associated autoimmunity. Diabetologia. 2003;46:712–720.
    1. Vafiadis P., Bennett S.T., Todd J.A., Nadeau J., Grabs R., Goodyer C.G. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nature Genetics. 1997;15:289–292.
    1. Badenhoop K., Kahles H., Penna-Martinez M. Vitamin D, immune tolerance, and prevention of type 1 diabetes. Current Diabetes Reports. 2012;12:635–642.
    1. Gibson V.B., Nikolic T., Pearce V.Q., Demengeot J., Roep B.O., Peakman M. Proinsulin multi-peptide immunotherapy induces antigen-specific regulatory T cells and limits autoimmunity in a humanized model. Clinical & Experimental Immunology. 2015;182:251–260.
    1. Serr I., Fürst R.W., Achenbach P., Scherm M.G., Gökmen F., Haupt F. Type 1 diabetes vaccine candidates promote human Foxp3+Treg induction in humanized mice. Nature Communications. 2016 (in press)
    1. Harrison L.C., Honeyman M.C., Steele C.E., Stone N.L., Sarugeri E., Bonifacio E. Pancreatic beta-cell function and immune responses to insulin after administration of intranasal insulin to humans at risk for type 1 diabetes. Diabetes Care. 2004;27:2348–2355.
    1. Uusitalo U., Liu X., Yang J., Aronsson C.A., Hummel S., Butterworth M., TEDDY Study Group, 2016 Association of early exposure of probiotics and islet autoimmunity in the TEDDY study. The Journal of the American Medical Association Pediatrics. 2016 Jan 1;170(1):20–28.
    1. Dopico X.C., Evangelou M., Ferreira R., Guo H., Pekalski M., Smyth D. Widespread seasonal gene expression reveals annual differences in human immunity and physiology. Nature Communications. 2015;6:7000.
    1. Ziegler A.G., Bonifacio E., the BABYDIAB-BABYDIET Study Group Age-related islet autoantibody incidence in offspring of patients with type 1 diabetes. Diabetologia. 2012;55:1937–1943.
    1. Parikka V., Näntö-Salonen K., Saarinen M., Simell T., Ilonen J., Hyöty H. Early seroconversion and rapidly increasing autoantibody concentrations predict prepubertal manifestation of type 1 diabetes in children at genetic risk. Diabetologia. 2012;55:1926–1936.
    1. Krischer J.P., Lynch K.F., Schatz D.A., Ilonen J., Lernmark Å., Hagopian W.A., the TEDDY Study Group The 6 year incidence of diabetes-associated autoantibodies in genetically at-risk children: the TEDDY study. Diabetologia. 2015;58:980–987.
    1. Bonifacio E., Hummel M., Walter M., Schmid S., Ziegler A.G. IDDM1 and multiple family history of type 1 diabetes combine to identify neonates at high risk for type 1 diabetes. Diabetes Care. 2004;27:2695–2699.
    1. Winkler C., Krumsiek J., Buettner F., Angermüller C., Giannopoulou E.Z., Theis F.J. Feature ranking of type 1 diabetes susceptibility genes improves prediction of type 1 diabetes. Diabetologia. 2014;57:2521–2529.
    1. Lambert A.P., Gillespie K.M., Thomson G., Cordell H.J., Todd J.A., Gale E.A. Absolute risk of childhood-onset type 1 diabetes defined by human leukocyte antigen class II genotype: a population-based study in the United Kingdom. Journal of Clinical Endocrinology and Metabolism. 2004;89:4037–4043.
    1. Oram R.A., Patel K., Hill A., Shields B., McDonald T.J., Jones A. A type 1 diabetes genetic risk score can aid discrimination between type 1 and type 2 diabetes in young adults. Diabetes Care. 2016;39(3):337–344.

Source: PubMed

3
Abonneren