Intralymphatic Glutamic Acid Decarboxylase With Vitamin D Supplementation in Recent-Onset Type 1 Diabetes: A Double-Blind, Randomized, Placebo-Controlled Phase IIb Trial

Johnny Ludvigsson, Zdenek Sumnik, Terezie Pelikanova, Lia Nattero Chavez, Elena Lundberg, Itxaso Rica, Maria A Martínez-Brocca, Marisol Ruiz de Adana, Jeanette Wahlberg, Anastasia Katsarou, Ragnar Hanas, Cristina Hernandez, Maria Clemente León, Ana Gómez-Gila, Marcus Lind, Marta Ferrer Lozano, Theo Sas, Ulf Samuelsson, Stepanka Pruhova, Fabricia Dietrich, Sara Puente Marin, Anders Nordlund, Ulf Hannelius, Rosaura Casas, Johnny Ludvigsson, Zdenek Sumnik, Terezie Pelikanova, Lia Nattero Chavez, Elena Lundberg, Itxaso Rica, Maria A Martínez-Brocca, Marisol Ruiz de Adana, Jeanette Wahlberg, Anastasia Katsarou, Ragnar Hanas, Cristina Hernandez, Maria Clemente León, Ana Gómez-Gila, Marcus Lind, Marta Ferrer Lozano, Theo Sas, Ulf Samuelsson, Stepanka Pruhova, Fabricia Dietrich, Sara Puente Marin, Anders Nordlund, Ulf Hannelius, Rosaura Casas

Abstract

Objective: To evaluate the efficacy of aluminum-formulated intralymphatic glutamic acid decarboxylase (GAD-alum) therapy combined with vitamin D supplementation in preserving endogenous insulin secretion in all patients with type 1 diabetes (T1D) or in a genetically prespecified subgroup.

Research design and methods: In a multicenter, randomized, placebo-controlled, double-blind trial, 109 patients aged 12-24 years (mean ± SD 16.4 ± 4.1) with a diabetes duration of 7-193 days (88.8 ± 51.4), elevated serum GAD65 autoantibodies, and a fasting serum C-peptide >0.12 nmol/L were recruited. Participants were randomized to receive either three intralymphatic injections (1 month apart) with 4 μg GAD-alum and oral vitamin D (2,000 IE daily for 120 days) or placebo. The primary outcome was the change in stimulated serum C-peptide (mean area under the curve [AUC] after a mixed-meal tolerance test) between baseline and 15 months.

Results: Primary end point was not met in the full analysis set (treatment effect ratio 1.091 [CI 0.845-1.408]; P = 0.5009). However, GAD-alum-treated patients carrying HLA DR3-DQ2 (n = 29; defined as DRB1*03, DQB1*02:01) showed greater preservation of C-peptide AUC (treatment effect ratio 1.557 [CI 1.126-2.153]; P = 0.0078) after 15 months compared with individuals receiving placebo with the same genotype (n = 17). Several secondary end points showed supporting trends, and a positive effect was seen in partial remission (insulin dose-adjusted HbA1c ≤9; P = 0.0310). Minor transient injection site reactions were reported.

Conclusion: Intralymphatic administration of GAD-alum is a simple, well-tolerated treatment that together with vitamin D supplementation seems to preserve C-peptide in patients with recent-onset T1D carrying HLA DR3-DQ2. This constitutes a disease-modifying treatment for T1D with a precision medicine approach.

Trial registration: ClinicalTrials.gov NCT03345004.

© 2021 by the American Diabetes Association.

Figures

Figure 1
Figure 1
Trial profile. Between December 2017 and April 2019, a total of 140 patients were screened for a fasting C-peptide ≥0.12 nmol/L and positivity for GAD65 antibodies (1Information on HLA genotype was missing for one patient in the placebo group.
Figure 2
Figure 2
Primary and key secondary study outcomes. Relative change from baseline (back transformed from log scale model) in C-peptide AUCmean 0–120 min during an MMTT for the two treatment groups (GAD-alum and placebo) in the FAS (A) and the prespecified subgroup HLA DR3-DQ2 (B). Error bars indicate SD. C: Forest plot depicting estimated treatment difference between GAD-alum and placebo groups for key secondary end points (IDAA1c and HbA1c in nmol/mol and daily exogenous insulin dose in units/kg/24 h) observed in the FAS and in the the prespecified subgroup HLA DR3-DQ2. Error bars indicate 95% CI. P values are indicated. Primary and key secondary efficacy end point variables were analyzed using a restricted maximum likelihood–based repeated measures approach (mixed-model repeated measures), as described in Research Design and Methods.
Figure 3
Figure 3
Pharmacological outcomes. Median changes from baseline of GADA and proliferation (Pr) of PMBC (stimulation index [SI]) (A) and GAD-stimulated secretion by PBMC of IL-10 and IL-13 levels (B) for GAD-alum–treated patients with and without the DR3-DQ2 haplotype as well as placebo-treated patients. P values by Wilcoxon test are indicated.

References

    1. Lind M, Pivodic A, Svensson AM, Ólafsdóttir AF, Wedel H, Ludvigsson J. HbA1c level as a risk factor for retinopathy and nephropathy in children and adults with type 1 diabetes: Swedish population based cohort study. BMJ 2019;366:l4894.
    1. Lind M, Svensson AM, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2015;372:880–881
    1. Rawshani A, Sattar N, Franzén S, et al. . Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018;392:477–486
    1. Ludvigsson J. The clinical potential of low-level C-peptide secretion. Expert Rev Mol Diagn 2016;16:933–940
    1. Herold KC, Hagopian W, Auger JA, et al. . Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med 2002;346:1692–1698
    1. Sherry N, Hagopian W, Ludvigsson J, et al. .; Protégé Trial Investigators . Teplizumab for treatment of type 1 diabetes (Protégé study): 1-year results from a randomised, placebo-controlled trial. Lancet 2011;378:487–497
    1. Quattrin T, Haller MJ, Steck AK, et al. .; T1GER Study Investigators . Golimumab and beta-cell function in youth with new-onset type 1 diabetes. N Engl J Med 2020;383:2007–2017
    1. Haller MJ, Schatz DA, Skyler JS, et al. .; Type 1 Diabetes TrialNet ATG-GCSF Study Group . Low-dose anti-thymocyte globulin (ATG) preserves β-cell function and improves HbA1c in new-onset type 1 diabetes. Diabetes Care 2018;41:1917–1925
    1. Rigby MR, Harris KM, Pinckney A, et al. . Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J Clin Invest 2015;125:3285–3296
    1. Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, et al. .; Type 1 Diabetes TrialNet Anti-CD20 Study Group . Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med 2009;361:2143–2152
    1. Näntö-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
    1. Ludvigsson J, Faresjö M, Hjorth M, et al. . GAD treatment and insulin secretion in recent-onset type 1 diabetes. N Engl J Med 2008;359:1909–1920
    1. Wherrett DK, Bundy B, Becker DJ, et al. .; Type 1 Diabetes TrialNet GAD Study Group . Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial. Lancet 2011;378:319–327
    1. Ludvigsson J, Krisky D, Casas R, et al. . GAD65 antigen therapy in recently diagnosed type 1 diabetes mellitus. N Engl J Med 2012;366:433–442
    1. Krischer JP, Lynch KF, Lernmark Å, et al. .; TEDDY Study Group . Genetic and environmental interactions modify the risk of diabetes-related autoimmunity by 6 years of age: the TEDDY study. Diabetes Care 2017;40:1194–1202
    1. Battaglia M, Ahmed S, Anderson MS, et al. . Introducing the endotype concept to address the challenge of disease heterogeneity in type 1 diabetes. Diabetes Care 2020;43:5–12
    1. Hannelius U, Beam CA, Ludvigsson J. Efficacy of GAD-alum immunotherapy associated with HLA-DR3-DQ2 in recently diagnosed type 1 diabetes. Diabetologia 2020;63:2177–2181
    1. Senti G, Freiburghaus AU, Larenas-Linnemann D, et al. . Intralymphatic immunotherapy: update and unmet needs. Int Arch Allergy Immunol 2019;178:141–149
    1. Casas R, Dietrich F, Barcenilla H, et al. . Glutamic acid decarboxylase injection into lymph-nodes: beta cell-function and immune responses in recent onset type 1 diabetes patients. Front Immunol 2020;11:564921.
    1. Greenbaum CJ, Mandrup-Poulsen T, McGee PF, et al. .; Type 1 Diabetes Trial Net Research Group; European C-Peptide Trial Study Group . Mixed-meal tolerance test versus glucagon stimulation test for the assessment of beta-cell function in therapeutic trials in type 1 diabetes. Diabetes Care 2008;31:1966–1971
    1. Axelsson S, Chéramy M, Åkerman L, Pihl M, Ludvigsson J, Casas R. Cellular and humoral immune responses in type 1 diabetic patients participating in a phase III GAD-alum intervention trial. Diabetes Care 2013;36:3418–3424
    1. Mortensen HB, Hougaard P, Swift P, et al. .; Hvidoere Study Group on Childhood Diabetes . New definition for the partial remission period in children and adolescents with type 1 diabetes. Diabetes Care 2009;32:1384–1390
    1. Lachin JM, McGee PL, Greenbaum CJ, et al. .; Type 1 Diabetes Trial Network . Sample size requirements for studies of treatment effects on beta-cell function in newly diagnosed type 1 diabetes. PLoS One 2011;6:e26471.
    1. Beam CA, MacCallum C, Herold KC, Wherrett DK, Palmer J, Ludvigsson J; Type 1 Diabetes TrialNet Study Group . GAD vaccine reduces insulin loss in recently diagnosed type 1 diabetes: findings from a Bayesian meta-analysis. Diabetologia 2017;60:43–49
    1. Maloy KJ, Erdmann I, Basch V, et al. . Intralymphatic immunization enhances DNA vaccination. Proc Natl Acad Sci USA 2001;98:3299–3303
    1. Johansen P, Häffner AC, Koch F, et al. . Direct intralymphatic injection of peptide vaccines enhances immunogenicity. Eur J Immunol 2005;35:568–574
    1. Senti G, Prinz Vavricka BM, Erdmann I, et al. . Intralymphatic allergen administration renders specific immunotherapy faster and safer: a randomized controlled trial. Proc Natl Acad Sci USA 2008;105:17908–17912
    1. Ludvigsson J, Wahlberg J, Casas R. Intralymphatic injection of autoantigen in type 1 diabetes. N Engl J Med 2017;376:697–699
    1. Ludvigsson J, Tavira B, Casas R. More on intralymphatic injection of autoantigen in type 1 diabetes. N Engl J Med 2017;377:403–405
    1. Piemonti L, Monti P, Sironi M, et al. . Vitamin D3 affects differentiation, maturation, and function of human monocyte-derived dendritic cells. J Immunol 2000;164:4443–4451
    1. Bizzarri C, Pitocco D, Napoli N, et al. .; IMDIAB Group . No protective effect of calcitriol on beta-cell function in recent-onset type 1 diabetes: the IMDIAB XIII trial. Diabetes Care 2010;33:1962–1963
    1. Ludvigsson J. Autoantigen treatment in type 1 diabetes: unsolved questions on how to select autoantigen and administration route. Int J Mol Sci 2020;21:1598
    1. Alhadj Ali M, Liu Y-F, 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.
    1. Ostrov DA, Alkanani A, McDaniel KA, et al. . Methyldopa blocks MHC class II binding to disease-specific antigens in autoimmune diabetes. J Clin Invest 2018;128:1888–1902
    1. Chung WK, Erion K, Florez JC, et al. . Precision medicine in diabetes: a consensus report from the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 2020;43:1617–1635

Source: PubMed

3
Abonner