Oral insulin immunotherapy in children at risk for type 1 diabetes in a randomised controlled trial

Robin Assfalg, Jan Knoop, Kristi L Hoffman, Markus Pfirrmann, Jose Maria Zapardiel-Gonzalo, Anna Hofelich, Anne Eugster, Marc Weigelt, Claudia Matzke, Julia Reinhardt, Yannick Fuchs, Melanie Bunk, Andreas Weiss, Markus Hippich, Kathrin Halfter, Stefanie M Hauck, Jörg Hasford, Joseph F Petrosino, Peter Achenbach, Ezio Bonifacio, Anette-Gabriele Ziegler, Robin Assfalg, Jan Knoop, Kristi L Hoffman, Markus Pfirrmann, Jose Maria Zapardiel-Gonzalo, Anna Hofelich, Anne Eugster, Marc Weigelt, Claudia Matzke, Julia Reinhardt, Yannick Fuchs, Melanie Bunk, Andreas Weiss, Markus Hippich, Kathrin Halfter, Stefanie M Hauck, Jörg Hasford, Joseph F Petrosino, Peter Achenbach, Ezio Bonifacio, Anette-Gabriele Ziegler

Abstract

Aims/hypothesis: Oral administration of antigen can induce immunological tolerance. Insulin is a key autoantigen in childhood type 1 diabetes. Here, oral insulin was given as antigen-specific immunotherapy before the onset of autoimmunity in children from age 6 months to assess its safety and immune response actions on immunity and the gut microbiome.

Methods: A phase I/II randomised controlled trial was performed in a single clinical study centre in Germany. Participants were 44 islet autoantibody-negative children aged 6 months to 2.99 years who had a first-degree relative with type 1 diabetes and a susceptible HLA DR4-DQ8-containing genotype. Children were randomised 1:1 to daily oral insulin (7.5 mg with dose escalation to 67.5 mg) or placebo for 12 months using a web-based computer system. The primary outcome was immune efficacy pre-specified as induction of antibody or T cell responses to insulin and measured in a central treatment-blinded laboratory.

Results: Randomisation was performed in 44 children. One child in the placebo group was withdrawn after the first study visit and data from 22 insulin-treated and 21 placebo-treated children were analysed. Oral insulin was well tolerated with no changes in metabolic variables. Immune responses to insulin were observed in children who received both insulin (54.5%) and placebo (66.7%), and the trial did not demonstrate an effect on its primary outcome (p = 0.54). In exploratory analyses, there was preliminary evidence that the immune response and gut microbiome were modified by the INS genotype Among children with the type 1 diabetes-susceptible INS genotype (n = 22), antibody responses to insulin were more frequent in insulin-treated (72.7%) as compared with placebo-treated children (18.2%; p = 0.03). T cell responses to insulin were modified by treatment-independent inflammatory episodes.

Conclusions/interpretation: The study demonstrated that oral insulin immunotherapy in young genetically at-risk children was safe, but was not associated with an immune response as predefined in the trial primary outcome. Exploratory analyses suggested that antibody responses to oral insulin may occur in children with a susceptible INS genotype, and that inflammatory episodes may promote the activation of insulin-responsive T cells.

Trial registration: Clinicaltrials.gov NCT02547519 FUNDING: The main funding source was the German Center for Diabetes Research (DZD e.V.).

Keywords: Autoimmunity; Insulin; Oral immunotherapy; Primary prevention; Type 1 diabetes.

Figures

Fig. 1
Fig. 1
Schematics of participant disposition, design and treatment groups. (a) Disposition of the participants. All children had a first-degree family history of type 1 diabetes. Study endpoint was the development of persistent antibodies to GAD, IA-2 or ZnT8. (b) Study design and treatment groups
Fig. 2
Fig. 2
Blood glucose concentration and insulin/C-peptide ratio over time. (ad) Blood glucose concentrations were measured in children before and after intake of oral insulin at a dose of 7.5 mg at baseline visit 1 (a; n = 22), 22.5 mg at 3 months visit 2 (b; n = 22), and 67.5 mg at 6 months visit 3 (c; n =22), or placebo at visits 1–3 (d; n = 63). The concentrations for individual children are connected by lines. The dashed line indicates the threshold for hypoglycaemia at 2.78 mmol/l (ad). (eg) The insulin/C-peptide ratio is plotted for each time point at visit 1 (e), visit 2 (f) and visit 3 (g) for children receiving oral insulin (red triangles) or placebo (blue circles)
Fig. 3
Fig. 3
Responses to treatment and analysis of responses. (a, b) Immune response to oral insulin or placebo. Kaplan–Meier analysis of a positive antibody response to insulin (a) and CD4+ T cell response to insulin (b) as defined by the primary outcome criteria in children who received placebo (blue line; n = 21) or oral insulin (red line; n = 22). The follow-up time is calculated from the first day of treatment (a, b). (c) CD4+ T cell response to insulin calculated as the SI relative to medium control at baseline (visit 1) and at 12 months (visit 5) in children who received placebo (blue circles; n = 21 at baseline, n = 18 at 12 months) or oral insulin (red circles; n = 22 at baseline, n = 18 at 12 months). (d) Kaplan–Meier analysis of a positive antibody response to insulin as defined by the primary outcome criteria in children with the INS AA genotype who received placebo (blue line; n = 11) or oral insulin (red line; n = 11; p = 0.0085)
Fig. 4
Fig. 4
Microbiome alterations in relation to age, INS genotype and treatment. (a, b) Alpha diversity in relation to age over the time of study participation in samples from baseline (n = 40), 6 months (n = 40) and 12 months (n = 35); shown are richness (observed OTU) (a) and evenness (Shannon) (b). (c, d) Beta diversity in relation to age over time of study participation (baseline, 6 months, 12 months); shown are Jaccard distance (c; p < 0.0001) and Bray–Curtis distance (d; p < 0.0001). Each dot represents the distance between two samples within the age range (c, d). (eg) Beta diversity differences by PCoA (baseline, 6 months, 12 months); shown are Jaccard distance in children with the INS AA genotype (purple dots and lines) or the INS AT or TT genotype (bright-blue dots and lines) (e; p = 0.0258), Bray–Curtis distance in children with the INS AA genotype (purple dots and lines) or the INS AT or TT genotype (bright-blue dots and lines) (f; p = 0.0422), and Jaccard distance in children with the INS AA genotype who received placebo (dark-blue dots and lines) or oral insulin (red dots and lines), and in children with the INS AT or TT genotype who received placebo (bright-orange dots and lines) or oral insulin (fuchsia dots and lines) (g; p = 0.0069). (h) Alpha diversity (Shannon) in children with the INS AA genotype who had a negative (n = 22 samples) or positive (n = 4 samples) antibody response to insulin (6 months, 12 months) (p = 0.0395). Unless indicated, the plots include both placebo- and oral insulin-treated children. For treatment-related analyses, only post-baseline samples at 6 and 12 months were included. Ab, antibody; PC1, principal component 1; PC2, principal component 2; PCoA, principal coordinates analysis
Fig. 5
Fig. 5
T cell responses to insulin in relation to INS genotype and monocyte CD169 expression. (ad) Monocyte CD169 expression and CD4+ T cell responses to insulin over the time of study participation (baseline, 3, 6, 9, 12 months) in all study participants. (a) Representative flow cytometry histograms of CD169 staining intensity on monocytes. Shown are three samples with low (blue; 0.6% positive cells), moderate (light blue, 21.2% positive cells) and high monocyte CD169 expression (red, 99.7% positive cells). A threshold of >5% positive monocytes was used as the threshold for defining monocyte CD169+ samples. (b) Percentage of CD169+ cells out of CD14+ monocytes in relation to age over the time of study participation in children who received placebo (blue lines; n = 21) or oral insulin (red lines; n = 22); plotted on a log scale. (c) Correlation between the frequency of CD169+ monocytes and the frequency of intermediate monocytes in peripheral blood from children who received placebo (blue circles; n = 102 samples) or oral insulin (red circles; n = 104 samples; r = 0.52, p < 0.0001). (d) CD4+ T cell responses to insulin (SI) in samples from 20 children with the INS AT or TT genotype and 22 children with the INS AA genotype and stratified by monocyte CD169 expression as negative (CD169neg, grey circles; n = 148 samples) or positive (CD169pos, green circles; n = 58 samples) in all study visits; plotted on a log scale. (e) The frequencies of insulin-responsive CD4+ T cells (n = 1036 cells from 22 samples) in the Th1/Th21-like cell clusters 2 and 3 (left; white bars) and the Treg-like clusters 9, 10 and 11 (right; grey bars) according to whether cells were from children with the INS AT/TT (n = 550 cells) or INS AA (n = 486 cells) genotype and samples that were monocyte CD169 negative (n = 559 cells) or positive (n = 477 cells). **p < 0.01, ***p < 0.001

References

    1. Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet. 2014;383:69–82. doi: 10.1016/S0140-6736(13)60591-7.
    1. Bonifacio E, Achenbach P. Birth and coming of age of islet autoantibodies. Clin Exp Immunol. 2019;198:294–305. doi: 10.1111/cei.13360.
    1. Ziegler AG, 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. doi: 10.2337/diabetes.48.3.460.
    1. Ilonen J, Hammais A, Laine AP, et al. Patterns of beta-cell autoantibody appearance and genetic associations during the first years of life. Diabetes. 2013;62:3636–3640. doi: 10.2337/db13-0300.
    1. Krischer JP, Lynch KF, Schatz DA, et al. The 6 year incidence of diabetes-associated autoantibodies in genetically at-risk children: The TEDDY study. Diabetologia. 2015;58:980–987. doi: 10.1007/s00125-015-3514-y.
    1. Parikka V, Nanto-Salonen K, Saarinen M, et al. Early seroconversion and rapidly increasing autoantibody concentrations predict prepubertal manifestation of type 1 diabetes in children at genetic risk. Diabetologia. 2012;55:1926–1936. doi: 10.1007/s00125-012-2523-3.
    1. Ziegler AG, Bonifacio E, Group B-BS Age-related islet autoantibody incidence in offspring of patients with type 1 diabetes. Diabetologia. 2012;55:1937–1943. doi: 10.1007/s00125-012-2472-x.
    1. 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;114:589–597. doi: 10.1172/JCI200421307.
    1. Ziegler AG, Rewers M, Simell O et al (2013) Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 309:2473–2479. 10.1001/jama.2013.6285
    1. Bennett ST, Lucassen AM, Gough SC, et al. Susceptibility to human type 1 diabetes at IDDM2 is determined by tandem repeat variation at the insulin gene minisatellite locus. Nat Genet. 1995;9:284–292. doi: 10.1038/ng0395-284.
    1. Barratt BJ, Payne F, Lowe CE, et al. Remapping the insulin gene/IDDM2 locus in type 1 diabetes. Diabetes. 2004;53:1884–1889. doi: 10.2337/diabetes.53.7.1884.
    1. Walter M, Albert E, Conrad M, et al. IDDM2/insulin VNTR modifies risk conferred by IDDM1/HLA for development of type 1 diabetes and associated autoimmunity. Diabetologia. 2003;46:712–720. doi: 10.1007/s00125-003-1082-z.
    1. Vafiadis P, Bennett ST, Todd JA, et al. Insulin expression in human thymus is modulated by INS VNTR alleles at the IDDM2 locus. Nat Genet. 1997;15:289–292. doi: 10.1038/ng0397-289.
    1. Pugliese A, Zeller M, Fernandez A, Jr, et al. The insulin gene is transcribed in the human thymus and transcription levels correlated with allelic variation at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Nat Genet. 1997;15:293–297. doi: 10.1038/ng0397-293.
    1. Du Toit G, Roberts G, Sayre PH, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. N Engl J Med. 2015;372:803–813. doi: 10.1056/NEJMoa1414850.
    1. Miller SD, Turley DM, Podojil JR. Antigen-specific tolerance strategies for the prevention and treatment of autoimmune disease. Nat Rev Immunol. 2007;7:665–677. doi: 10.1038/nri2153.
    1. Skyler JS, Krischer JP, Wolfsdorf J, et al. Effects of oral insulin in relatives of patients with type 1 diabetes: The diabetes prevention trial--type 1. Diabetes Care. 2005;28:1068–1076. doi: 10.2337/diacare.28.5.1068.
    1. Writing Committee for the Type 1 Diabetes TrialNet Oral Insulin Study G, Krischer JP, Schatz DA, Bundy B, Skyler JS, Greenbaum CJ (2017) Effect of Oral insulin on prevention of diabetes in relatives of patients with type 1 diabetes: A randomized clinical trial. JAMA 318:1891–1902. 10.1001/jama.2017.17070
    1. Nanto-Salonen K, Kupila A, Simell S, et al. 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;372:1746–1755. doi: 10.1016/S0140-6736(08)61309-4.
    1. Harrison LC, Honeyman MC, Steele CE, et al. 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. doi: 10.2337/diacare.27.10.2348.
    1. Diabetes Prevention Trial--Type 1 Diabetes Study G Effects of insulin in relatives of patients with type 1 diabetes mellitus. N Engl J Med. 2002;346:1685–1691. doi: 10.1056/NEJMoa012350.
    1. Elding Larsson H, Lundgren M, Jonsdottir B, Cuthbertson D, Krischer J, Di A-ITSG Safety and efficacy of autoantigen-specific therapy with 2 doses of alum-formulated glutamate decarboxylase in children with multiple islet autoantibodies and risk for type 1 diabetes: A randomized clinical trial. Pediatr Diabetes. 2018;19:410–419. doi: 10.1111/pedi.12611.
    1. Bonifacio E, Ziegler A, Achenbach P, Barker J, Eisenbarth G. Translating mucosal antigen based prevention of autoimmune diabetes to human. Novartis Found Symp. 2008;292:187–199. doi: 10.1002/9780470697405.ch17.
    1. Bonifacio E, Ziegler AG, Klingensmith G et al (2015) Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes: The pre-POINT randomized clinical trial. JAMA 313:1541–1549. 10.1001/jama.2015.2928
    1. Heninger AK, Eugster A, Kuehn D, et al. A divergent population of autoantigen-responsive CD4(+) T cells in infants prior to beta cell autoimmunity. Sci Transl Med. 2017;9:eaaf8848. doi: 10.1126/scitranslmed.aaf8848.
    1. Stewart CJ, Ajami NJ, O'Brien JL, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562:583–588. doi: 10.1038/s41586-018-0617-x.
    1. Bonifacio E, Scirpoli M, Kredel K, Fuchtenbusch M, Ziegler AG. Early autoantibody responses in prediabetes are IgG1 dominated and suggest antigen-specific regulation. J Immunol. 1999;163:525–532.
    1. Bonifacio E, Yu L, Williams AK, et al. Harmonization of glutamic acid decarboxylase and islet antigen-2 autoantibody assays for national institute of diabetes and digestive and kidney diseases consortia. J Clin Endocrinol Metab. 2010;95:3360–3367. doi: 10.1210/jc.2010-0293.
    1. Achenbach P, Lampasona V, Landherr U, et al. Autoantibodies to zinc transporter 8 and SLC30A8 genotype stratify type 1 diabetes risk. Diabetologia. 2009;52:1881–1888. doi: 10.1007/s00125-009-1438-0.
    1. Naserke HE, Dozio N, Ziegler AG, Bonifacio E. Comparison of a novel micro-assay for insulin autoantibodies with the conventional radiobinding assay. Diabetologia. 1998;41:681–683. doi: 10.1007/s001250050968.
    1. Koczwara K, Muller D, Achenbach P, Ziegler AG, Bonifacio E. Identification of insulin autoantibodies of IgA isotype that preferentially target non-human insulin. Clin Immunol. 2007;124:77–82. doi: 10.1016/j.clim.2007.03.545.
    1. Dupont WD, Plummer WD., Jr Power and sample size calculations for studies involving linear regression. Control Clin Trials. 1998;19:589–601. doi: 10.1016/S0197-2456(98)00037-3.
    1. Oksanen J, Blanchet GF et al (2019) Vegan: Community Ecology Package. R package version 2.0–2. Available from
    1. Wickham H, et al. ggplot2: Elegant graphics for data analysis (use R) Heidelberg: Springer; 2009.
    1. Sharma A, Liu X, Hadley D, et al. Identification of non-HLA genes associated with development of islet autoimmunity and type 1 diabetes in the prospective TEDDY cohort. J Autoimmun. 2018;89:90–100. doi: 10.1016/j.jaut.2017.12.008.
    1. Ferreira RC, Guo H, Coulson RM, et al. A type I interferon transcriptional signature precedes autoimmunity in children genetically at risk for type 1 diabetes. Diabetes. 2014;63:2538–2550. doi: 10.2337/db13-1777.
    1. Ziegler AG, Achenbach P, Berner R, et al. Oral insulin therapy for primary prevention of type 1 diabetes in infants with high genetic risk: The GPPAD-POInT (global platform for the prevention of autoimmune diabetes primary oral insulin trial) study protocol. BMJ Open. 2019;9:e028578. doi: 10.1136/bmjopen-2018-028578.
    1. Davis-Richardson AG, Ardissone AN, Dias R, et al. Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front Microbiol. 2014;5:678. doi: 10.3389/fmicb.2014.00678.
    1. Rasmussen T, Witso E, Tapia G, Stene LC, Ronningen KS. Self-reported lower respiratory tract infections and development of islet autoimmunity in children with the type 1 diabetes high-risk HLA genotype: The MIDIA study. Diabetes Metab Res Rev. 2011;27:834–837. doi: 10.1002/dmrr.1258.
    1. Beyerlein A, Wehweck F, Ziegler AG, Pflueger M. Respiratory infections in early life and the development of islet autoimmunity in children at increased type 1 diabetes risk: Evidence from the BABYDIET study. JAMA Pediatr. 2013;167:800–807. doi: 10.1001/jamapediatrics.2013.158.
    1. Lonnrot M, Lynch KF, Elding Larsson H, et al. Respiratory infections are temporally associated with initiation of type 1 diabetes autoimmunity: The TEDDY study. Diabetologia. 2017;60:1931–1940. doi: 10.1007/s00125-017-4365-5.
    1. Beyerlein A, Donnachie E, Jergens S, Ziegler AG (2016) Infections in early life and development of type 1 diabetes. JAMA 315:1899–1901. 10.1001/jama.2016.2181
    1. Alhadj Ali M, Liu YF, Arif S, et al. Metabolic and immune effects of immunotherapy with proinsulin peptide in human new-onset type 1 diabetes. Sci Transl Med. 2017;9:eaaf7779. doi: 10.1126/scitranslmed.aaf7779.
    1. Wilson DS, Damo M, Hirosue S, et al. Synthetically glycosylated antigens induce antigen-specific tolerance and prevent the onset of diabetes. Nat Biomed Eng. 2019;3:817–829. doi: 10.1038/s41551-019-0424-1.

Source: PubMed

3
S'abonner