Protocol of the adaptive study of IL-2 dose frequency on regulatory T cells in type 1 diabetes (DILfrequency): a mechanistic, non-randomised, repeat dose, open-label, response-adaptive study

Lucy A Truman, Marcin L Pekalski, Paula Kareclas, Marina Evangelou, Neil M Walker, James Howlett, Adrian P Mander, Jane Kennet, Linda S Wicker, Simon Bond, John A Todd, Frank Waldron-Lynch, Lucy A Truman, Marcin L Pekalski, Paula Kareclas, Marina Evangelou, Neil M Walker, James Howlett, Adrian P Mander, Jane Kennet, Linda S Wicker, Simon Bond, John A Todd, Frank Waldron-Lynch

Abstract

Introduction: Type 1 diabetes (T1D) is caused by autoimmune destruction of the insulin-producing β cells in the pancreatic islets, leading to insulinopenia and hyperglycaemia. Genetic analyses indicate that alterations of the interleukin-2 (IL-2) pathway mediating immune activation and tolerance predispose to T1D, specifically the polymorphic expression of the IL-2 receptor-α chain (CD25) on T lymphocytes. Replacement of physiological doses of IL-2 could restore self-tolerance and prevent further autoimmunity by enhancing the function of CD4(+) T regulatory cells (Tregs) to limit the activation of auto reactive T effector cells (Teffs). In this experimental medicine study, we use an adaptive trial design to determine the optimal dosing regimen for IL-2 to improve Treg function while limiting activation of Teffs in participants with T1D.

Methods and analysis: The Adaptive study of IL-2 dose frequency on Tregs in type 1 diabetes(DILfrequency) is a mechanistic, non-randomised, repeat dose open-label, response-adaptive study of 36 participants with T1D. The objective is to establish the optimal dose and frequency of ultra-low dose IL-2: to increase Treg frequency within the physiological range, to increase CD25 expression on Tregs, without increasing CD4(+) Teffs. DILfrequency has an initial learning phase where 12 participants are allocated to six different doses and frequencies followed by an interim statistical analysis. After analysis of the learning phase, the Dose and Frequency Committee will select the optimal targets for Treg frequency, Treg CD25 expression and Teff frequency. Three groups of eight participants will be treated consecutively in the confirming phase. Each dose and frequency selected will be based on statistical analysis of all data collected from the previous groups.

Ethics: Ethical approval for DILfrequency was granted on 12 August 2014.

Results: The results of this study will be reported, through peer-reviewed journals, conference presentations and an internal organisational report.

Trial registration numbers: NCT02265809, ISRCTN40319192, CRN17571.

Keywords: DIABETES & ENDOCRINOLOGY; GENETICS; IMMUNOLOGY.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/

Figures

Figure 1
Figure 1
Study design of the learning phase of DILfrequency. The study has 12 visits and starts with a screening visit followed by a treatment period of 10 visits and a follow-up visit that will be carried out approximately 4 weeks after the final dose of Proleukin. Twelve participants will be allocated in the learning phase to two different doses and four frequencies of administration of Proleukin to measure the change from baseline of CD4+ T regulatory cells (Tregs), CD4+ T effectors and CD25 expression on Tregs during treatment with ultra-low dose interleukin-2. At the first two dosing visits, 90 min time points are measured to access early immune activation and the effects of repeat dosing on these events.
Figure 2
Figure 2
Clinical fluorescence-activated cell sorting (FACS) assays on whole blood to measure absolute lymphocyte counts, proportions of lymphocytes and CD25 on T regulatory cells. Fluorescently labelled beads are added to whole blood and analysed to accurately count the absolute number of lymphocytes (A), beads (B), CD3+ T cells (C) CD4+ and CD8+ T cells (D) CD19+ B cells and CD19−, CD16+, CD56+ NK cells as a percentage of all lymphocytes (E). In a parallel whole blood FACS assay, a lymphocyte gate is drawn to include all events (F) and doublets are excluded (not shown). The CD3+, CD4+ T-cell gate excludes CD8+ T cells and B cells (G). CD127−, CD25+ T regulatory cells (Tregs) are separated from non-Tregs since the non-Tregs are heterogeneous for CD127 and CD25 and this percentage is used to calculate the absolute Treg count out of CD4+ (H), naïve, effector memory, central memory and total effector memory CD45 RA+(TEMRA) Tregs (I) and non-Tregs (J) are measured according to CD62L and CD45RA expression, as shown. A cocktail of six standardised beads labelled with different amounts of fluorescent allophycocyanin (APC) are measured by FACS daily to accurately measure CD25-APC on the surface of Tregs (K) and a standard curve plotted (L). The mean fluorescence intensity of CD25+ on Tregs can be accurately read from the curve, minimising interassay variation.

References

    1. Daneman D. Type 1 diabetes. Lancet 2006;367:847–58. 10.1016/S0140-6736(06)68341-4
    1. Yang JH, Cutler AJ, Ferreira RC et al. . Natural variation in interleukin-2 sensitivity influences regulatory T-cell frequency and function in individuals with long-standing type 1 diabetes. Diabetes 2015;64:3891–902. 10.2337/db15-0516
    1. Oram RA, McDonald TJ, Shields BM et al. . Most people with long-duration type 1 diabetes in a large population-based study are insulin microsecretors. Diabetes Care 2015;38:323–8. 10.2337/dc14-0871
    1. The Diabetes Control and Complications Trial Research Group. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 1993;329:977–86. 10.1056/NEJM199309303291401
    1. The Diabetes Control and Complications Trial Research Group Hypoglycaemia in the Diabetes Control and Complications Trial. Diabetes 1997;46:271–86. 10.2337/diab.46.2.271
    1. Steffes MW, Sibley S, Jackson M et al. . Beta-cell function and the development of diabetes-related complications in the diabetes control and complications trial. Diabetes Care 2003;26:832–6. 10.2337/diacare.26.3.832
    1. Kuhtreiber WM, Washer SL, Hsu E et al. . Low levels of C-peptide have clinical significance for established Type 1 diabetes. Diabet Med 2015;32:1346–53.
    1. Todd JA. Etiology of type 1 diabetes. Immunity 2010;32:457–67. 10.1016/j.immuni.2010.04.001
    1. Sakaguchi S, Miyara M, Costantino CM et al. . FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 2010;10:490–500. 10.1038/nri2785
    1. Todd JA, Walker NM, Cooper JD et al. . Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 2007;39:857–64. 10.1038/ng2068
    1. Rubio-Cabezas O, Minton JA, Caswell R et al. . Clinical heterogeneity in patients with FOXP3 mutations presenting with permanent neonatal diabetes. Diabetes Care 2009;32:111–16. 10.2337/dc08-1188
    1. Amy A, Caudy Sreelatha T, Reddy Talal Chatila et al. . CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked—like syndrome, and defective IL-10 expression from CD4 lymphocytes J Allergy Clin Immunol 2007;119:482–7.
    1. Garg G, Tyler JR, Yang JH et al. . Type 1 diabetes-associated IL2RA variation lowers IL-2 signaling and contributes to diminished CD4+CD25+ regulatory T cell function. J Immunol 2012;188:4644–53. 10.4049/jimmunol.1100272
    1. Dendrou CA, Wicker LS. The IL-2/CD25 pathway determines susceptibility to T1D in humans and NOD mice. J Clin Immunol 2008;28:685–96. 10.1007/s10875-008-9237-9
    1. Dendrou CA, Plagnol V, Fung E et al. . Cell-specific protein phenotypes for the autoimmune locus IL2RA using a genotype-selectable human bioresource. Nat Genet 2009;41:1011–15. 10.1038/ng.434
    1. Burchill MA, Yang J, Vogtenhuber C et al. . IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+regulatory T cells. J Immunology 2007;178:280–90. 10.4049/jimmunol.178.1.280
    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;59:407–15. 10.2337/db09-0694
    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: 2055–66. 10.1056/NEJMoa1108188
    1. Matsuoka K, Koreth J, Kim HT et al. . Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med 2013;5:179ra43 10.1126/scitranslmed.3005265
    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;365:2067–77. 10.1056/NEJMoa1105143
    1. Virgin HW, Todd JA. Metagenomics and personalized medicine. Cell 2011;147:44–56. 10.1016/j.cell.2011.09.009
    1. Waldron-Lynch F, Kareclas P, Irons K et al. . Rationale and study design of the adaptive study of IL-2 dose on regulatory T cells in type 1 diabetes (DILT1D): a non-randomised, open label, adaptive dose finding trial. BMJ Open 2014;4:e005559 10.1136/bmjopen-2014-005559
    1. Bond S, Mander A, Todd J et al. . Adaptive dose-finding designs to identify multiple doses that achieve multiple response targets. Trials 2013;14:O78 10.1186/1745-6215-14-S1-O78
    1. Hartemann A, Bensimon G, Payan CA et al. . Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2013;1:295–305. 10.1016/S2213-8587(13)70113-X
    1. Rosenzwajg M, Churlaud G, Mallone R et al. . Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun 2015;58:48–58. 10.1016/j.jaut.2015.01.001
    1. Yu A, Snowhite I, Vendrame F et al. . Selective IL-2 responsiveness of regulatory T cells through multiple intrinsic mechanisms supports the use of low-dose IL-2 therapy in type 1 diabetes. Diabetes 2015;64:2172–83. 10.2337/db14-1322
    1. Churlaud G, Jimenez V, Ruberte J et al. . Sustained stimulation and expansion of Tregs by IL2 control autoimmunity without impairing immune responses to infection, vaccination and cancer. Clin Immunol 2014;151:114–26. 10.1016/j.clim.2014.02.003
    1. Long SA, Buckner JH, Greenbaum CJ. IL-2 therapy in type 1 diabetes: ‘Trials’ and tribulations. Clin Immunol 2013;149:324–31. 10.1016/j.clim.2013.02.005
    1. Fyfe G, Fisher RI, Rosenberg SA et al. . Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 1995;13:688–96.
    1. Abrams D, Lévy Y, Losso MH et al. . Interleukin-2 therapy in patients with HIV infection. N Engl J Med 2009;361:1548–59. 10.1056/NEJMoa0903175
    1. Long SA, Rieck M, Sanda S et al. . Rapamycin/IL-2 Combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function. Diabetes 2012;61:2340–8. 10.2337/db12-0049
    1. Heywood J, Evangelou M, Goymer D et al. . Effective recruitment of participants to a phase I study using the internet and publicity releases through charities and patient organisations: analysis of the adaptive study of IL-2 dose on regulatory T cells in type 1 diabetes (DILT1D). Trials 2015;16:86 10.1186/s13063-015-0583-7
    1. ADDRESS2 After Diabetes Diagnosis REsearch Support System (ADDRESS). 2014.
    1. DGAP. Diabetes: genes, autoimmunity and prevention. 2014.
    1. Poli A, Michel T, Thérésine M et al. . CD56bright natural killer (NK) cells: an important NK cell subset. Immunology 2009;126: 458–65. 10.1111/j.1365-2567.2008.03027.x
    1. Ito S, Bollard CM, Carlsten M et al. . Ultra-low dose interleukin-2 promotes immune-modulating function of regulatory T cells and natural killer cells in healthy volunteers. Mol Ther 2014;22:1388–95. 10.1038/mt.2014.50
    1. Sim GC, Martin-Orozco N, Jin L et al. . IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J Clin Invest 2014;124:99–110. 10.1172/JCI46266
    1. Rosenzwajg M, Churlaud G, Hartemann A et al. . Interleukin-2 in the pathogenesis and therapy of type 1 diabetes. Curr Diab Rep 2014;14:553 10.1007/s11892-014-0553-6

Source: PubMed

3
Sottoscrivi