Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function

S Alice Long, Mary Rieck, Srinath Sanda, Jennifer B Bollyky, Peter L Samuels, Robin Goland, Andrew Ahmann, Alex Rabinovitch, Sudeepta Aggarwal, Deborah Phippard, Laurence A Turka, Mario R Ehlers, Peter J Bianchine, Karen D Boyle, Steven A Adah, Jeffrey A Bluestone, Jane H Buckner, Carla J Greenbaum, Diabetes TrialNet and the Immune Tolerance Network, S Alice Long, Mary Rieck, Srinath Sanda, Jennifer B Bollyky, Peter L Samuels, Robin Goland, Andrew Ahmann, Alex Rabinovitch, Sudeepta Aggarwal, Deborah Phippard, Laurence A Turka, Mario R Ehlers, Peter J Bianchine, Karen D Boyle, Steven A Adah, Jeffrey A Bluestone, Jane H Buckner, Carla J Greenbaum, Diabetes TrialNet and the Immune Tolerance Network

Abstract

Rapamycin/interleukin-2 (IL-2) combination treatment of NOD mice effectively treats autoimmune diabetes. We performed a phase 1 clinical trial to test the safety and immunologic effects of rapamycin/IL-2 combination therapy in type 1 diabetic (T1D) patients. Nine T1D subjects were treated with 2-4 mg/day rapamycin orally for 3 months and 4.5 × 10(6) IU IL-2 s.c. three times per week for 1 month. β-Cell function was monitored by measuring C-peptide. Immunologic changes were monitored using flow cytometry and serum analyses. Regulatory T cells (Tregs) increased within the first month of therapy, yet clinical and metabolic data demonstrated a transient worsening in all subjects. The increase in Tregs was transient, paralleling IL-2 treatment, whereas the response of Tregs to IL-2, as measured by STAT5 phosphorylation, increased and persisted after treatment. No differences were observed in effector T-cell subset frequencies, but an increase in natural killer cells and eosinophils occurred with IL-2 therapy. Rapamycin/IL-2 therapy, as given in this phase 1 study, resulted in transient β-cell dysfunction despite an increase in Tregs. Such results highlight the difficulties in translating therapies to the clinic and emphasize the importance of broadly interrogating the immune system to evaluate the effects of therapy.

Trial registration: ClinicalTrials.gov NCT00525889.

Figures

FIG. 1.
FIG. 1.
Transient decrease in β-cell function with rapamycin/IL-2 combination therapy. Trial design is shown in A for which T1D patients were treated with 4.5 × 106 IU IL-2 s.c. three times per week for 1 month and rapamycin 2–4 mg/day orally for 3 months with target blood rapamycin trough levels of 5–10 ng/mL (see Supplementary Fig. 1). β-Cell function was monitored using an MMTT, and PBMCs were collected for mechanistic studies on the indicated days. B: Peak C-peptide values from the MMTT were assessed for all subjects over time. Statistical significance was determined using a paired Student t test comparing screening and day 84 and days 84–168.
FIG. 2.
FIG. 2.
Rapamycin/IL-2 combination therapy transiently increases nTregs in the peripheral blood. Thawed PBMCs were stained for viability, CD4, CD25, CD127, and FOXP3. A and B: The frequency of CD4+ cells that are CD25+CD127lo and the number of cells that are CD3+CD4+FOXP3+ within PBMCs are shown for each patient. C: nTregs quantified using real-time PCR analysis of the Treg-specific demethylated region of the FOXP3 gene relative to the methylated region are expressed as percentage of CD3 cells (www.epiontis.com). D: PBMCs were stimulated with PMA/ionomysin in the presence of brefeldin A/myosin prior to staining for FOXP3 and IFN-γ. Means ± SD are shown in D. Statistical significance was determined by a one-way ANOVA and is shown in each graph. Significant differences from time 0 to each time point were determined using a paired Student t test and are indicated with an asterisk. *P ≤ 0.05 and **P ≤ 0.01.
FIG. 3.
FIG. 3.
Rapamycin/IL-2 combination therapy results in a persistent enhancement of IL-2 responsiveness in CD4+CD25+ T cells. Thawed PBMCs were stimulated with media alone or 100 IU/mL IL-2 prior to fixation, permeabilization, and staining for CD4, CD25, and pSTAT5. Representative dot plots of CD4+ cells are shown in A. B: The frequency of pSTAT5+ cells of CD4+CD25+ cells (A, top) is shown for control (n = 5) and T1D (n = 5) subjects matched to subjects in the trial (noted by ITN R + 2 Trial) by HLA, age, sex, and genotype (PTPN221858 or PTPN2rs1893217). Day 0 and 28 analyses are shown for patients in the trial. Statistical significance was determined between control and T1D subjects (matched + trial) using a two-sample Student t test and between day 0 and 28 in the trial using a paired Student t test. C: The frequency and fold increase (FI) of pSTAT5 in CD4+CD25+ cells are shown over time for patients in the trial. Statistical significance was determined by a one-way ANOVA and is shown in both graphs. Significance from time 0 to each time point was determined using a paired Student t test and is indicated with an asterisk. *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001.
FIG. 4.
FIG. 4.
Rapamycin/IL-2 combination therapy modulates pAkt, but not phosphorylated (p)Erk or pS6, in CD4+ T cells. Thawed PBMCs were stimulated with media alone or cross-linking of anti-CD3/anti-CD28–bound antibodies with Fab x-linker for 20, 30, or 10 min for Erk, S6, and Akt analysis, respectively. Cells were then fixed, permeabilized, and stained for CD4, CD8, pAkt, pErk, and pS6. Representative histograms are shown for pErk (A), pS6 (B), and pAkt (C) with media (gray line) and anti-CD3/anti-CD28 (black line) stimulation. Fold increase (FI) was determined by the equation (MFI with CD3/CD28 stim) ÷ (MFI with media) for each sample and is compared between day 0 and 84 for pErk (A), pS6 (B), and pAkt (C). Data from all subjects receiving full dosing of rapamycin are included except for pAkt for subjects 1 and 2, for which pAkt data were not acquired. Statistical significance was determined by a paired Student t test.
FIG. 5.
FIG. 5.
Rapamycin/IL-2 combination therapy did not alter CD4 helper subset differentiation or CD8 memory composition. Thawed PBMCs were stained for viability, CD4, CD8, CD45RO, CXCR3, and CRTH2. A: The frequency of memory CD45RO+ (solid square) and naive CD45RO− (open circles) in the CD4+ (left) and CD8+ (right) T-cell populations is shown. B: The frequency of CXCR3 and CRTH2 of CD4+CD45RO+ cells is shown as approximate markers of Th1 and Th2 cells, respectively. C: PBMCs were stimulated for 6 h with PMA/ionomysin in the presence of brefeldin A/myosin prior to staining for CD4, CD45RO, IL-17, and IFN-γ. Frequencies of IFN-γ+ (open squares, left axis) and IL-17+ (solid circles, right axis) cells of CD4+CD45RO+-gated cells are shown. Means ± SD are shown for all subjects assayed. Statistical significance was determined by a one-way ANOVA and is shown in each plot.
FIG. 6.
FIG. 6.
Rapamycin/IL-2 combination therapy increased eosinophils and sIL-2RA while decreasing serum TGF-β. A: The frequency of eosinophils in whole blood was determined in a standard hematologic assay and is shown for each subject over time. B: Serum analytes were measured using a multiplex Searchlight chemiluminiscent assay. Averages of duplicates for all subjects are shown for day 0 and 84, and subjects 1, 2, 3, 5, and 7 are shown for time points between days 14 and 70. Of the 22 cytokines and cytokine receptors tested, data are shown where statistical differences were observed. Measures are shown for each subject over time, and statistical significance was determined by a one-way ANOVA, noted in the graph. Significance from time 0 to each time point was determined using a paired Student t test and is indicated with an asterisk. *P ≤ 0.05, **P ≤ 0.01, and ***P ≤ 0.001.
FIG. 7.
FIG. 7.
Rapamycin/IL-2 combination therapy transiently augments NK cell frequency. Thawed PBMCs were stained for viability, CD56, CD3, and CD16. A: The number of CD56+CD3− NK cells of PBMCs are shown for individual subjects. B: The average ± SD frequencies for CD56hiCD16+ (open squares), CD56hiCD16− (closed triangles), and CD56loCD16+ (closed circles) subsets of CD56+ NK cells within the live lymphocyte gate are shown. Statistical significance was determined by a one-way ANOVA, noted in the graph. Significance from time 0 to each time point was determined using a paired Student t test and is indicated with an asterisk. *P ≤ 0.05, **P ≤ 0.01.

References

    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. 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. 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. Buckner JH. Mechanisms of impaired regulation by CD4(+)CD25(+)FOXP3(+) regulatory T cells in human autoimmune diseases. Nat Rev Immunol 2010;10:849–859
    1. Knip M, Siljander H. Autoimmune mechanisms in type 1 diabetes. Autoimmun Rev 2008;7:550–557
    1. Thomson AW, Turnquist HR, Raimondi G. Immunoregulatory functions of mTOR inhibition. Nat Rev Immunol 2009;9:324–337
    1. Delgoffe GM, Kole TP, Zheng Y, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 2009;30:832–844
    1. Delgoffe GM, Pollizzi KN, Waickman AT, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol 2011;12:295–303
    1. Powell JD, Delgoffe GM. The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism. Immunity 2010;33:301–311
    1. Monti P, Scirpoli M, Maffi P, et al. Rapamycin monotherapy in patients with type 1 diabetes modifies CD4+CD25+FOXP3+ regulatory T-cells. Diabetes 2008;57:2341–2347
    1. Malek TR. The biology of interleukin-2. Annu Rev Immunol 2008;26:453–479
    1. Turka LA, Walsh PT. IL-2 signaling and CD4+ CD25+ Foxp3+ regulatory T cells. Front Biosci 2008;13:1440–1446
    1. Hulme MA, Wasserfall CH, Atkinson MA, Brusko TM. Central role for interleukin-2 in type 1 diabetes. Diabetes 2012;61:14–22
    1. Grinberg-Bleyer Y, Baeyens A, You S, et al. IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J Exp Med 2010;207:1871–1878
    1. Koulmanda M, Budo E, Bonner-Weir S, et al. Modification of adverse inflammation is required to cure new-onset type 1 diabetic hosts. Proc Natl Acad Sci USA 2007;104:13074–13079
    1. Tang Q, Adams JY, Penaranda C, et al. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 2008;28:687–697
    1. Koreth J, Matsuoka K, Kim HT, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med 2011;365:2055–2066
    1. Saadoun D, Rosenzwajg M, Joly F, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 2011;365:2067–2077
    1. Rabinovitch A, Suarez-Pinzon WL, Shapiro AM, Rajotte RV, Power R. Combination therapy with sirolimus and interleukin-2 prevents spontaneous and recurrent autoimmune diabetes in NOD mice. Diabetes 2002;51:638–645
    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. Verge CF, Gianani R, Kawasaki E, et al. Prediction of type I diabetes in first-degree relatives using a combination of insulin, GAD, and ICA512bdc/IA-2 autoantibodies. Diabetes 1996;45:926–933
    1. Long SA, Cerosaletti K, Bollyky PL, et al. Defects in IL-2R signaling contribute to diminished maintenance of FOXP3 expression in CD4(+)CD25(+) regulatory T-cells of type 1 diabetic subjects. Diabetes 2010;59:407–415
    1. Wieczorek G, Asemissen A, Model F, et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue. Cancer Res 2009;69:599–608
    1. Sehouli J, Loddenkemper C, Cornu T, et al. Epigenetic quantification of tumor-infiltrating T-lymphocytes. Epigenetics 2011;6:236–246
    1. Gottlieb PA, Quinlan S, Krause-Steinrauf H, et al. Type 1 Diabetes TrialNet MMF/DZB Study Group Failure to preserve beta-cell function with mycophenolate mofetil and daclizumab combined therapy in patients with new-onset type 1 diabetes. Diabetes Care 2010;33:826–832
    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. 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. Huehn J, Polansky JK, Hamann A. Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Nat Rev Immunol 2009;9:83–89
    1. Tanemura M, Saga A, Kawamoto K, et al. Rapamycin induces autophagy in islets: relevance in islet transplantation. Transplant Proc 2009;41:334–338
    1. Zhang N, Su D, Qu S, et al. Sirolimus is associated with reduced islet engraftment and impaired beta-cell function. Diabetes 2006;55:2429–2436
    1. Piemonti L, Maffi P, Monti L, et al. Beta cell function during rapamycin monotherapy in long-term type 1 diabetes. Diabetologia 2011;54:433–439
    1. Di Paolo S, Teutonico A, Leogrande D, Capobianco C, Schena PF. Chronic inhibition of mammalian target of rapamycin signaling downregulates insulin receptor substrates 1 and 2 and AKT activation: a crossroad between cancer and diabetes? J Am Soc Nephrol 2006;17:2236–2244
    1. Brauner H, Elemans M, Lemos S, et al. Distinct phenotype and function of NK cells in the pancreas of nonobese diabetic mice. J Immunol 2010;184:2272–2280
    1. Schleinitz N, Vély F, Harlé JR, Vivier E. Natural killer cells in human autoimmune diseases. Immunology 2010;131:451–458
    1. Bielekova B, Catalfamo M, Reichert-Scrivner S, et al. Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. Proc Natl Acad Sci USA 2006;103:5941–5946
    1. Bernstein ZP, Porter MM, Gould M, et al. Prolonged administration of low-dose interleukin-2 in human immunodeficiency virus-associated malignancy results in selective expansion of innate immune effectors without significant clinical toxicity. Blood 1995;86:3287–3294
    1. Hoenstein R, Admon D, Solomon A, Norris A, Moqbel R, Levi-Schaffer F. Interleukin-2 activates human peripheral blood eosinophils. Cell Immunol 2001;210:116–124
    1. Varker KA, Kondadasula SV, Go MR, et al. Multiparametric flow cytometric analysis of signal transducer and activator of transcription 5 phosphorylation in immune cell subsets in vitro and following interleukin-2 immunotherapy. Clin Cancer Res 2006;12:5850–5858
    1. Orange JS, Roy-Ghanta S, Mace EM, et al. IL-2 induces a WAVE2-dependent pathway for actin reorganization that enables WASp-independent human NK cell function. J Clin Invest 2011;121:1535–1548
    1. Turnquist HR, Cardinal J, Macedo C, et al. mTOR and GSK-3 shape the CD4+ T-cell stimulatory and differentiation capacity of myeloid DCs after exposure to LPS. Blood 2010;115:4758–4769
    1. Robb RJ, Kutny RM. Structure-function relationships for the IL 2-receptor system. IV. Analysis of the sequence and ligand-binding properties of soluble Tac protein. J Immunol 1987;139:855–862
    1. Maier LM, Anderson DE, Severson CA, et al. Soluble IL-2RA levels in multiple sclerosis subjects and the effect of soluble IL-2RA on immune responses. J Immunol 2009;182:1541–1547

Source: PubMed

3
Abonner