Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders

Kevan C Herold, Stephen E Gitelman, Mario R Ehlers, Peter A Gottlieb, Carla J Greenbaum, William Hagopian, Karen D Boyle, Lynette Keyes-Elstein, Sudeepta Aggarwal, Deborah Phippard, Peter H Sayre, James McNamara, Jeffrey A Bluestone, AbATE Study Team, Kevan C Herold, Stephen E Gitelman, Mario R Ehlers, Peter A Gottlieb, Carla J Greenbaum, William Hagopian, Karen D Boyle, Lynette Keyes-Elstein, Sudeepta Aggarwal, Deborah Phippard, Peter H Sayre, James McNamara, Jeffrey A Bluestone, AbATE Study Team

Abstract

Trials of immune therapies in new-onset type 1 diabetes (T1D) have shown success, but not all subjects respond, and the duration of response is limited. Our aim was to determine whether two courses of teplizumab, an Fc receptor-nonbinding anti-CD3 monoclonal antibody, reduces the decline in C-peptide levels in patients with T1D 2 years after disease onset. We also set out to identify characteristics of responders. We treated 52 subjects with new-onset T1D with teplizumab for 2 weeks at diagnosis and after 1 year in an open-label, randomized, controlled trial. In the intent to treat analysis of the primary end point, patients treated with teplizumab had a reduced decline in C-peptide at 2 years (mean -0.28 nmol/L [95% CI -0.36 to -0.20]) versus control (mean -0.46 nmol/L [95% CI -0.57 to -0.35]; P = 0.002), a 75% improvement. The most common adverse events were rash, transient upper respiratory infections, headache, and nausea. In a post hoc analysis we characterized clinical responders and found that metabolic (HbA1c and insulin use) and immunologic features distinguished this group from those who did not respond to teplizumab. We conclude that teplizumab treatment preserves insulin production and reduces the use of exogenous insulin in some patients with new-onset T1D. Metabolic and immunologic features at baseline can identify a subgroup with robust responses to immune therapy.

Trial registration: ClinicalTrials.gov NCT00129259.

Figures

FIG. 1.
FIG. 1.
Enrollment, randomization, and participation. Subjects (n = 125) were screened and 83 were eligible for enrollment. The majority of subjects who were excluded failed to meet entry criteria. Of 83 subjects randomized, 25 in the control group and 52 in the teplizumab group underwent an MMTT at baseline, received the first dose of study drug (teplizumab group), and are included in the ITT analysis. Of the subjects randomized to the teplizumab group, 12 did not receive the second course of drug because of AEs leading to discontinuation during cycle 1 (n = 6), had predefined laboratory abnormalities that precluded readministration (n = 4), or were withdrawn (n = 2). In addition, 3 of 40 subjects who started cycle 2 discontinued treatment because of AEs. The reasons for drug discontinuation are listed in Supplementary Table 1.
FIG. 2.
FIG. 2.
C-peptide (C-pep) responses in drug-treated and control subjects. A: The mean ± 25th and 75th percentiles of the C-pep AUC (nanomoles per liter) are shown for the drug and control groups (***P = 0.002; **P < 0.02, ANCOVA for each time point). B: Estimates and 95% CIs from a mixed effects model, with fixed effects for treatment group and linear and quadratic trends over time and random subject-level effects for intercepts and linear trends over time. Drug-treated subjects are estimated to have a delay of decline in C-pep by 15.9 months (i.e., would reach control 6-month values 21.9 months after study entry). C: Proportion of subjects with detectable C-pep (i.e., >0.05 nmol/L). The actual study dates when the MMTTs were performed are shown. There was a significantly greater loss of detectable C-pep secretion in the control group at month 24 (P = 0.002, χ2 test).
FIG. 3.
FIG. 3.
Insulin use and HbA1c in drug and control groups. A: HbA1c levels in the drug and control groups (means ± 25th and 75th percentiles are shown; *P < 0.05). B: Average insulin use in the 3 days before the visit was calculated (group means ± 25th and 75th percentiles; *P < 0.05, ***P < 0.005; P < 0.001 for overall trend at month 12, and P = 0.022 at month 18 using ANCOVA).
FIG. 4.
FIG. 4.
Identification of clinical responders to teplizumab. A: The cumulative frequency of subjects and distribution of percentage decrease from baseline C-peptide AUC at month 24. The arrow shows the smallest percentage loss of C-peptide AUC in the control group. B: The C-peptide AUC at each time point (means ± 25th and 75th percentiles) is shown for the responders (red line) and nonresponders (green line) in the drug-treated group and for the control subjects (blue line). ***P < 0.001 between responders and nonresponders at each time point based on ANCOVAs.
FIG. 5.
FIG. 5.
Baseline clinical characteristics in responders versus nonresponders. A: Ladder plot of covariates and clinical response. The effects of the indicated covariates on the ratio of log odds of responder status and 95% CIs are shown. The baseline HbA1c (P = 0.011) and insulin use (P = 0.004) were inversely associated with response. Baseline insulin use (B) and HbA1c (C) in the drug-treated responders (red) and nonresponders (green) (**P = 0.004, *P = 0.011).

References

    1. Herold KC, Gitelman SE, Masharani U, et al. A single course of anti-CD3 monoclonal antibody hOKT3gamma1(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;54:1763–1769
    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. Orban T, Bundy B, Becker DJ, et al. Type 1 Diabetes TrialNet Abatacept Study Group Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet 2011;378:412–419
    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. Keymeulen B, Vandemeulebroucke E, Ziegler AG, et al. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med 2005;352:2598–2608
    1. Herold KC, Gitelman S, Greenbaum C, et al. Immune Tolerance Network ITN007AI Study Group 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;132:166–173
    1. Keymeulen B, Walter M, Mathieu C, et al. 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. Diabetologia 2010;53:614–623
    1. Press RR, de Fijter JW, Guchelaar HJ. Individualizing calcineurin inhibitor therapy in renal transplantation—current limitations and perspectives. Curr Pharm Des 2010;16:176–186
    1. Bougnères PF, Landais P, Boisson C, et al. Limited duration of remission of insulin dependency in children with recent overt type I diabetes treated with low-dose cyclosporin. Diabetes 1990;39:1264–1272
    1. Bertin-Maghit S, Pang D, O’Sullivan B, et al. Interleukin-1β produced in response to islet autoantigen presentation differentiates T-helper 17 cells at the expense of regulatory T-cells: Implications for the timing of tolerizing immunotherapy. Diabetes 2011;60:248–257
    1. Yang XO, Nurieva R, Martinez GJ, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 2008;29:44–56
    1. Larsen CM, Faulenbach M, Vaag A, et al. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med 2007;356:1517–1526
    1. 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;128:517–523
    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. Herold KC, Gitelman SE, Willi SM, et al. Teplizumab treatment may improve C-peptide responses in participants with type 1 diabetes after the new-onset period: a randomised controlled trial. Diabetologia 2013;56:391–400
    1. Greenbaum CJ, Beam CA, Boulware D, et al. Type 1 Diabetes TrialNet Study Group Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data. Diabetes 2012;61:2066–2073
    1. Herold KC, Pescovitz MD, McGee P, et al. Type 1 Diabetes TrialNet Anti-CD20 Study Group Increased T cell proliferative responses to islet antigens identify clinical responders to anti-CD20 monoclonal antibody (rituximab) therapy in type 1 diabetes. J Immunol 2011;187:1998–2005
    1. Gerriets VA, Rathmell JC. Metabolic pathways in T cell fate and function. Trends Immunol 2012;33:168–173
    1. Cham CM, Gajewski TF. Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J Immunol 2005;174:4670–4677
    1. Mandrup-Poulsen T, Spinas GA, Prowse SJ, et al. Islet cytotoxicity of interleukin 1. Influence of culture conditions and islet donor characteristics. Diabetes 1987;36:641–647
    1. Maedler K, Sergeev P, Ris F, et al. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 2002;110:851–860
    1. Robertson RP, Harmon J, Tran PO, Tanaka Y, Takahashi H. Glucose toxicity in beta-cells: type 2 diabetes, good radicals gone bad, and the glutathione connection. Diabetes 2003;52:581–587
    1. Antonelli A, Fallahi P, Ferrari SM, et al. Serum Th1 (CXCL10) and Th2 (CCL2) chemokine levels in children with newly diagnosed type 1 diabetes: a longitudinal study. Diabet Med 2008;25:1349–1353
    1. Coppieters KT, Dotta F, Amirian N, et al. Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients. J Exp Med 2012;209:51–60
    1. Itoh N, Hanafusa T, Miyazaki A, et al. Mononuclear cell infiltration and its relation to the expression of major histocompatibility complex antigens and adhesion molecules in pancreas biopsy specimens from newly diagnosed insulin-dependent diabetes mellitus patients. J Clin Invest 1993;92:2313–2322
    1. Ablamunits V, Bisikirska B, Herold KC. Acquisition of regulatory function by human CD8(+) T cells treated with anti-CD3 antibody requires TNF. Eur J Immunol 2010;40:2891–2901
    1. Ablamunits V, Henegariu O, Preston-Hurlburt P, Herold KC. NKG2A is a marker for acquisition of regulatory function by human CD8+ T cells activated with anti-CD3 antibody. Eur J Immunol 2011;41:1832–1842
    1. Bisikirska B, Colgan J, Luban J, Bluestone JA, Herold KC. TCR stimulation with modified anti-CD3 mAb expands CD8+ T cell population and induces CD8+CD25+ Tregs. J Clin Invest 2005;115:2904–2913
    1. Belghith M, Bluestone JA, Barriot S, Mégret J, Bach JF, Chatenoud L. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med 2003;9:1202–1208
    1. You S, Leforban B, Garcia C, Bach JF, Bluestone JA, Chatenoud L. Adaptive TGF-beta-dependent regulatory T cells control autoimmune diabetes and are a privileged target of anti-CD3 antibody treatment. Proc Natl Acad Sci U S A 2007;104:6335–6340
    1. Waldron-Lynch F, Henegariu O, Deng S, Preston-Hurlburt P, Tooley J, Flavell R, Herold KC. Teplizumab induces human gut-tropic regulatory cells in humanized mice and patients. Sci Transl Med 2012;4:118ra112

Source: PubMed

3
Se inscrever