IL-6 receptor blockade does not slow β cell loss in new-onset type 1 diabetes

Carla J Greenbaum, Elisavet Serti, Katharina Lambert, Lia J Weiner, Sai Kanaparthi, Sandra Lord, Stephen E Gitelman, Darrell M Wilson, Jason L Gaglia, Kurt J Griffin, William E Russell, Philip Raskin, Antoinette Moran, Steven M Willi, Eva Tsalikian, Linda A DiMeglio, Kevan C Herold, Wayne V Moore, Robin Goland, Mark Harris, Maria E Craig, Desmond A Schatz, David A Baidal, Henry Rodriguez, Kristina M Utzschneider, Hendrik J Nel, Carol L Soppe, Karen D Boyle, Karen Cerosaletti, Lynette Keyes-Elstein, S Alice Long, Ranjeny Thomas, James G McNamara, Jane H Buckner, Srinath Sanda, ITN058AI EXTEND Study Team, Carla J Greenbaum, Elisavet Serti, Katharina Lambert, Lia J Weiner, Sai Kanaparthi, Sandra Lord, Stephen E Gitelman, Darrell M Wilson, Jason L Gaglia, Kurt J Griffin, William E Russell, Philip Raskin, Antoinette Moran, Steven M Willi, Eva Tsalikian, Linda A DiMeglio, Kevan C Herold, Wayne V Moore, Robin Goland, Mark Harris, Maria E Craig, Desmond A Schatz, David A Baidal, Henry Rodriguez, Kristina M Utzschneider, Hendrik J Nel, Carol L Soppe, Karen D Boyle, Karen Cerosaletti, Lynette Keyes-Elstein, S Alice Long, Ranjeny Thomas, James G McNamara, Jane H Buckner, Srinath Sanda, ITN058AI EXTEND Study Team

Abstract

BackgroundIL-6 receptor (IL-6R) signaling drives development of T cell populations important to type 1 diabetes pathogenesis. We evaluated whether blockade of IL-6R with monoclonal antibody tocilizumab would slow loss of residual β cell function in newly diagnosed type 1 diabetes patients.MethodsWe conducted a multicenter, randomized, placebo-controlled, double-blind trial with tocilizumab in new-onset type 1 diabetes. Participants were screened within 100 days of diagnosis. Eligible participants were randomized 2:1 to receive 7 monthly doses of tocilizumab or placebo. The primary outcome was the change from screening in the mean AUC of C-peptide collected during the first 2 hours of a mixed meal tolerance test at week 52 in pediatric participants (ages 6-17 years).ResultsThere was no statistical difference in the primary outcome between tocilizumab and placebo. Immunophenotyping showed reductions in downstream signaling of the IL-6R in T cells but no changes in CD4 memory subsets, Th17 cells, Tregs, or CD4+ T effector cell resistance to Treg suppression. A DC subset decreased during therapy but regressed to baseline once therapy stopped. Tocilizumab was well tolerated.ConclusionTocilizumab reduced T cell IL-6R signaling but did not modulate CD4+ T cell phenotypes or slow loss of residual β cell function in newly diagnosed individuals with type 1 diabetes.Trial RegistrationClinicalTrials.gov NCT02293837.FundingNIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) and National Institute of Allergy and Infectious Diseases (NIAID) UM1AI109565, UL1TR000004 from NIH/National Center for Research Resources (NCRR) Clinical and Translational Science Award (CTSA), NIH/NIDDK P30DK036836, NIH/NIDDK U01DK103266, NIH/NIDDK U01DK103266, 1UL1TR000064 from NIH/NCRR CTSA, NIH/National Center for Advancing Translational Sciences (NCATS) UL1TR001878, UL1TR002537 from NIH/CTSA; National Health and Medical Research Council Practitioner Fellowship (APP1136735), NIH/NIDDK U01-DK085476, NIH/CTSA UL1-TR002494, Indiana Clinical and Translational Science Institute Award UL1TR002529, Vanderbilt Institute for Clinical and Translational Research UL1TR000445. NIH/NCATS UL1TR003142, NIH/CTSA program UL1-TR002494, Veteran Affairs Administration, and 1R01AI132774.

Keywords: Beta cells; Diabetes; Endocrinology; Immunology; T cells.

Conflict of interest statement

Conflict of interest: CJG and SL have research support for their clinical trial site from Pfizer. DMW received support from Tolerion. SEG has served on advisory boards for Avotres, Biolojic, Caladrius Biosciences, ImmunoMolecular Therapeutics, Provention Bio, SAB Biotherapeutics, and Tolerion and participated in clinical trials with Caladrius Biosciences, Intrexon, Janssen, Provention Bio, and Tolerion. He led a Data and Safety Monitoring Board (DSMB) for Novo Nordisk (pediatric type 2 diabetes). JLG is a consultant to Vertex Pharmaceuticals and receives research support from Avotres. CLS’s husband works for FUJIFILM Irvine Scientific. JHB is a scientific cofounder and Scientific Advisory Board member of GentiBio, is a consultant for Bristol Myers Squibb and HotSpot Therapeutics, and has past and current research projects sponsored by Amgen, Bristol Myers Squibb, Janssen, Novo Nordisk, and Pfizer. She is a member of the Type 1 Diabetes TrialNet, a partner of the Allen Institute for Immunology, and a member of the scientific advisory boards for the La Jolla Institute for Immunology and Bristol Myers

Figures

Figure 1. CONSORT diagrams for both cohorts.
Figure 1. CONSORT diagrams for both cohorts.
(A) Pediatric participants and (B) adult participants. ANC, absolute neutrophil count; BILI, bilirubin; AST, aspartate aminotransferase.
Figure 2. Tocilizumab does not affect 2-hour…
Figure 2. Tocilizumab does not affect 2-hour C-peptide mAUC.
Markers represent the means, lines connect the medians, and error bars represent the 25th and 75th percentiles of C-peptide mAUC collected during the first 2 hours of the mixed meal tolerance test (MMTT) shown over the first year for (A) the pediatric cohort and (B) the adult cohort. ANCOVA models and mixed model analysis did not detect any statistically significant differences between the treatment groups at key time points.
Figure 3. Tocilizumab does not affect insulin…
Figure 3. Tocilizumab does not affect insulin usage and glucose control.
Markers represent the means, lines connect the medians, and error bars represent the 25th and 75th percentiles. HbA1c values for (A) the pediatric cohort and (B) the adult cohort. Average daily insulin usage expressed as total daily units/kg for (C) the pediatric cohort and (D) the adult cohort. ANCOVA models and mixed model analysis did not detect any statistically significant differences between the treatment groups at key time points.
Figure 4. Tocilizumab impairs IL-6R signaling in…
Figure 4. Tocilizumab impairs IL-6R signaling in T cells.
(A) Gating strategy of CD4+ Tregs and memory Teff cells and phosphorylated STAT3 (p-STAT3) expression after in vitro IL-6 stimulation, at baseline and week 12 of treatment of a representative treated patient. Longitudinal fold changes from baseline for (B) percentage of CD4+ Tem cells expressing p-STAT3, (C) MFI of p-STAT3 in CD4+ Tem cells, (D) percentage of Tregs expressing p-STAT3, (E) MFI of IL-6R in total CD4. Y axis scales are log transformed. Mean fold changes from baseline are presented at each visit. Error bars display SEM. P values were calculated using repeated measure 2-way ANOVA model. Statistically significant comparisons are shown with asterisks (***P < 0.0001; **P ≤ 0.001; *P ≤ 0.05). DN, double negative.
Figure 5. Tocilizumab does not alter frequency…
Figure 5. Tocilizumab does not alter frequency of CD4+ T cell subsets.
(A) Percentage of Tregs in total CD4. Percentage of (B) memory, (C) central memory, (D) effector memory, (E) Th17 (IL-17a+), (F) Tfh (IL-21+) in CD4+ Teff cells. Y axis scales are log transformed. Mean values are presented at each visit. Error bars display SEM. Repeated measure 2-way ANOVA did not detect any statistically significant differences between the treatment groups at key time points.
Figure 6. Tocilizumab does not alter Teff…
Figure 6. Tocilizumab does not alter Teff cell response to Treg suppression.
Percentage suppression of CD25 expression on Teff cells by Tregs. For all time points, Teff cells were cocultured with EF670-labeled Tregs and anti-CD3/anti-CD28 Dynabeads (1:28 beads/Teff) for 48 hours. Four Treg/Teff ratios were tested: 1:4 (top left), 1:8 (top right), 1:16 (bottom left), and 1:32 (bottom right). For flow cytometry, Teff cells were in the EF670– gate and stained with anti-CD25 PE-Cy7. Percentage suppression was calculated as follows: s = ([ab]/a) × 100, where a is the percentage CD25+ in the absence of Tregs and b is the percentage of CD25+ in the presence of Tregs. Data are represented as mean ± SEM. There were no statistical differences between placebo and tocilizumab at any Treg/Teff ratio or any time point. Repeated measures 2-way ANOVA did not detect any statistically significant differences between the treatment groups at key time points.
Figure 7. Tocilizumab changes frequencies of DC…
Figure 7. Tocilizumab changes frequencies of DC populations.
(A) Gating strategy for DC subsets (plasmacytoid DCs, pDCs; conventional type 1 DCs, cDC1s; and conventional type 2 DCs, cDC2s) and classical, nonclassical, and intermediate monocytes. (B) Percentage of classical monocytes in HLA-DR+ cells. (C) Percentage of pDCs (CD123+HLA-DR+) in CD14loCD1c– cells. (D) Percentage of cDC1s (CD141hiCD1c–) and (E) cDC2s (CD141loCD1c+) in CD14–CD16–cells. Y axis scales are log transformed and presented in actual scale. Mean values are presented at each visit. Error bars display SEM. P values were calculated using repeated measure 2-way ANOVA model. Statistically significant comparisons are shown with asterisks (***P < 0.0001; *P ≤ 0.05).
Figure 8. Increased serum IL-6 and IL-6R…
Figure 8. Increased serum IL-6 and IL-6R with tocilizumab therapy.
(A) C-reactive protein, (B) IL-6, and (C) soluble IL-6R. Y axis scales are log transformed and presented in actual scale for IL-6 and IL-6R analytes. C-reactive protein is plotted in actual scale. Mean values are presented at each visit. Error bars display SEM. P values were calculated using repeated measure 2-way ANOVA model. (***P < 0.0001.)

References

    1. Rawshani A, 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(10146):477–486. doi: 10.1016/S0140-6736(18)31506-X.
    1. Lind M, et al. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371(21):1972–1982. doi: 10.1056/NEJMoa1408214.
    1. Livingstone SJ, et al. Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008-2010. JAMA. 2015;313(1):37–44. doi: 10.1001/jama.2014.16425.
    1. Orban T, et al. Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial. Lancet. 2011;378(9789):412–419. doi: 10.1016/S0140-6736(11)60886-6.
    1. Pescovitz MD, et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N Engl J Med. 2009;361(22):2143–2152. doi: 10.1056/NEJMoa0904452.
    1. Herold KC, et al. 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. Diabetes. 2013;62(11):3766–3774.
    1. Rigby MR, et al. Targeting of memory T cells with alefacept in new-onset type 1 diabetes (T1DAL study): 12 month results of a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Diabetes Endocrinol. 2013;1(4):284–294. doi: 10.1016/S2213-8587(13)70111-6.
    1. Haller MJ, et al. Low-dose anti-thymocyte globulin preserves C-peptide, reduces HbA1c, and increases regulatory to conventional T-cell ratios in new-onset type 1 diabetes: two-year clinical trial data. Diabetes. 2019;68(6):1267–1276. doi: 10.2337/db19-0057.
    1. Von Herrath MG, et al. 278-OR: efficacy and safety of anti-interleukin (IL)-21 in combination with liraglutide in adults recently diagnosed with type 1 diabetes. Diabetes. 2021;9(4):212–224.
    1. Quattrin T, et al. Golimumab and beta-cell function in youth with new-onset type 1 diabetes. N Engl J Med. 2020;383(21):2007–2017. doi: 10.1056/NEJMoa2006136.
    1. Rose-John S. Interleukin-6 family cytokines. Cold Spring Harb Perspect Biol. 2018;10(2):a028415. doi: 10.1101/cshperspect.a028415.
    1. Heinrich PC, et al. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. Biochem J. 2003;374(pt 1):1–20.
    1. Samson M, et al. Brief report: inhibition of interleukin-6 function corrects Th17/Treg cell imbalance in patients with rheumatoid arthritis. Arthritis Rheum. 2012;64(8):2499–2503. doi: 10.1002/art.34477.
    1. Arif S, et al. Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated β-cell death. Diabetes. 2011;60(8):2112–2119. doi: 10.2337/db10-1643.
    1. Spence A, Tang Q. Restoring regulatory T cells in type 1 diabetes. Curr Diab Rep. 2016;16(11):110. doi: 10.1007/s11892-016-0807-6.
    1. Schneider A, et al. The effector T cells of diabetic subjects are resistant to regulation via CD4+ FOXP3+ regulatory T cells. J Immunol. 2008;181(10):7350–7355. doi: 10.4049/jimmunol.181.10.7350.
    1. Ferreira RC, et al. Functional IL6R 358Ala allele impairs classical IL-6 receptor signaling and influences risk of diverse inflammatory diseases. PLoS Genet. 2013;9(4):e1003444. doi: 10.1371/journal.pgen.1003444.
    1. Brunner HI, et al. Efficacy and safety of tocilizumab in patients with polyarticular-course juvenile idiopathic arthritis: results from a phase 3, randomised, double-blind withdrawal trial. Ann Rheum Dis. 2015;74(6):1110–1117. doi: 10.1136/annrheumdis-2014-205351.
    1. Smolen JS, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371(9617):987–997. doi: 10.1016/S0140-6736(08)60453-5.
    1. Goodman WA, et al. IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J Immunol. 2009;183(5):3170–3176. doi: 10.4049/jimmunol.0803721.
    1. Schneider A, et al. In active relapsing-remitting multiple sclerosis, effector T cell resistance to adaptive T(regs) involves IL-6-mediated signaling. Sci Transl Med. 2013;5(170):170ra15.
    1. Bettelli E, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–238. doi: 10.1038/nature04753.
    1. Bluestone JA, et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med. 2015;7(315):315ra189.
    1. Orabona C, et al. Deficiency of immunoregulatory indoleamine 2,3-dioxygenase 1in juvenile diabetes. JCI Insight. 2018;3(6):e96244. doi: 10.1172/jci.insight.96244.
    1. Bradshaw EM, et al. Monocytes from patients with type 1 diabetes spontaneously secrete proinflammatory cytokines inducing Th17 cells. J Immunol. 2009;183(7):4432–4439. doi: 10.4049/jimmunol.0900576.
    1. Hundhausen C, et al. Enhanced T cell responses to IL-6 in type 1 diabetes are associated with early clinical disease and increased IL-6 receptor expression. Sci Transl Med. 2016;8(356):356ra119.
    1. Garbers C, et al. The interleukin-6 receptor Asp358Ala single nucleotide polymorphism rs2228145 confers increased proteolytic conversion rates by ADAM proteases. Biochim Biophys Acta. 2014;1842(9):1485–1494.
    1. Pesce B, et al. Effect of interleukin-6 receptor blockade on the balance between regulatory T cells and T helper type 17 cells in rheumatoid arthritis patients. Clin Exp Immunol. 2013;171(3):237–242. doi: 10.1111/cei.12017.
    1. Thiolat A, et al. Interleukin-6 receptor blockade enhances CD39+ regulatory T cell development in rheumatoid arthritis and in experimental arthritis. Arthritis Rheumatol. 2014;66(2):273–283. doi: 10.1002/art.38246.
    1. Chavele KM, et al. Cutting edge: circulating plasmablasts induce the differentiation of human T follicular helper cells via IL-6 production. J Immunol. 2015;194(6):2482–2485. doi: 10.4049/jimmunol.1401190.
    1. Moran A, et al. Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials. Lancet. 2013;381(9881):1905–1915. doi: 10.1016/S0140-6736(13)60023-9.
    1. Yousif AS, et al. The persistence of interleukin-6 is regulated by a blood buffer systemderived from dendritic cells. Immunity. 2021;54(2):235–246. doi: 10.1016/j.immuni.2020.12.001.
    1. Nishimoto N, et al. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112(10):3959–3964. doi: 10.1182/blood-2008-05-155846.
    1. Ferreira RC, et al. Human IL-6R(hi)TIGIT(-) CD4(+)CD127(low)CD25(+) T cells display potent in vitro suppressive capacity and a distinct Th17 profile. Clin Immunol. 2017;179:25–39. doi: 10.1016/j.clim.2017.03.002.
    1. Richez C, et al. Tocilizumab treatment decreases circulating myeloid dendritic cells and monocytes, 2 components of the myeloid lineage. J Rheumatol. 2012;39(6):1192–1197. doi: 10.3899/jrheum.111439.
    1. Binnewies M, et al. Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell. 2019;177(3):556–571. doi: 10.1016/j.cell.2019.02.005.
    1. Price JD, Tarbell KV. The role of dendritic cell subsets and innate immunity in the pathogenesis of type 1 diabetes and other autoimmune diseases. Front Immunol. 2015;6:288.
    1. Kim JH, et al. Regulation of interleukin-6-induced hepatic insulin resistance by mammalian target of rapamycin through the STAT3-SOCS3 pathway. J Biol Chem. 2008;283(2):708–715. doi: 10.1074/jbc.M708568200.
    1. Senn JJ, et al. Suppressor of cytokine signaling-3 (SOCS-3), a potential mediator of interleukin-6-dependent insulin resistance in hepatocytes. J Biol Chem. 2003;278(16):13740–13746. doi: 10.1074/jbc.M210689200.
    1. Marasco MR, et al. Interleukin-6 reduces β-cell oxidative stress by linking autophagy with the antioxidant response. Diabetes. 2018;67(8):1576–1588. doi: 10.2337/db17-1280.
    1. Greenbaum CJ, et al. 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(10):1966–1971. doi: 10.2337/dc07-2451.
    1. Welch S, et al. Minimal model analysis of intravenous glucose tolerance test-derived insulin sensitivity in diabetic subjects. J Clin Endocrinol Metab. 1990;71(6):1508–1518. doi: 10.1210/jcem-71-6-1508.
    1. Boston RC, et al. MINMOD Millennium: a computer program to calculate glucose effectiveness and insulin sensitivity from the frequently sampled intravenous glucose tolerance test. Diabetes Technol Ther. 2003;5(6):1003–1015. doi: 10.1089/152091503322641060.
    1. American Diabetes Association. 6. Glycemic targets: standards of medical care in diabetes-2020. Diabetes Care. 2020;43(suppl 1):S66–S76.
    1. Rigby MR, et al. Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J Clin Invest. 2015;125(8):3285–3296. doi: 10.1172/JCI81722.
    1. Speake C, et al. A composite immune signature parallels disease progression across T1D subjects. JCI Insight. 2019;4(23):e126917. doi: 10.1172/jci.insight.126917.
    1. Herold KC, et al. An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. N Engl J Med. 2019;381(7):603–613. doi: 10.1056/NEJMoa1902226.
    1. Long AE, et al. A novel and rapid method to quantify Treg mediated suppression of CD4 T cells. J Immunol Methods. 2017;449:15–22. doi: 10.1016/j.jim.2017.06.009.
    1. Wherrett DK, et al. 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(9788):319–327. doi: 10.1016/S0140-6736(11)60895-7.
    1. Gottlieb PA, et al. 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(4):826–832. doi: 10.2337/dc09-1349.

Source: PubMed

3
Abonnieren