Biological and clinical insights from a randomized phase 2 study of an anti-oncostatin M monoclonal antibody in systemic sclerosis

Christopher P Denton, Francesco Del Galdo, Dinesh Khanna, Madelon C Vonk, Lorinda Chung, Sindhu R Johnson, John Varga, Daniel E Furst, Jane Temple, Chiara Zecchin, Eszter Csomor, Amy Lee, Nicolas Wisniacki, Shaun M Flint, Juliet Reid, Christopher P Denton, Francesco Del Galdo, Dinesh Khanna, Madelon C Vonk, Lorinda Chung, Sindhu R Johnson, John Varga, Daniel E Furst, Jane Temple, Chiara Zecchin, Eszter Csomor, Amy Lee, Nicolas Wisniacki, Shaun M Flint, Juliet Reid

Abstract

Objectives: The cytokine oncostatin M (OSM) is implicated in the pathology of SSc. Inhibiting OSM signalling using GSK2330811 (an anti-OSM monoclonal antibody) in patients with SSc has the potential to slow or stop the disease process.

Methods: This multicentre, randomized, double-blind, placebo-controlled study enrolled participants ≥18 years of age with active dcSSc. Participants were randomized 3:1 (GSK2330811:placebo) in one of two sequential cohorts to receive GSK2330811 (cohort 1: 100 mg; cohort 2: 300 mg) or placebo s.c. every other week for 12 weeks. The primary endpoint was safety; blood and skin biopsy samples were collected to explore mechanistic effects on inflammation and fibrosis. Clinical efficacy was an exploratory endpoint.

Results: Thirty-five participants were randomized to placebo (n = 8), GSK2330811 100 mg (n = 3) or GSK2330811 300 mg (n = 24). Proof of mechanism, measured by coordinate effects on biomarkers of inflammation or fibrosis, was not demonstrated following GSK2330811 treatment. There were no meaningful differences between GSK2330811 and placebo for any efficacy endpoints. The safety and tolerability of GSK2330811 were not favourable in the 300 mg group, with on-target, dose-dependent adverse events related to decreases in haemoglobin and platelet count that were not observed in the 100 mg or placebo groups.

Conclusion: Despite a robust and novel experimental medicine approach and evidence of target engagement, anticipated SSc-related biologic effects of GSK2330811 were not different from placebo and safety was unfavourable, suggesting OSM inhibition may not be a useful therapeutic strategy in SSc.

Trial registration number: ClinicalTrials.gov, NCT03041025; EudraCT, 2016-003417-95.

Keywords: GSK2330811; biomarkers; fibrosis; inflammation; monoclonal antibody; placebo-controlled trial; safety.

© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology.

Figures

Fig . 1
Fig. 1
(A–C) Ratio to baseline at day 85 and (D) including visits up to day 85a (A–C) Posterior medians are based on a Bayesian analysis at day 85 only. (D) Posterior medians are based on a Bayesian longitudinal analysis. aPer protocol population. αSMA: alpha smooth muscle actin; CCL2: C-C motif chemokine ligand 2; PIIINP: procollagen type III N-terminal peptide; 2GSSC: 2-gene skin score; BL: baseline; CrI: credible interval.
Fig . 2
Fig. 2
Mean (A) blood haemoglobin levels and (B) platelet counts (safety population) BL: baseline.

References

    1. Denton CP, Khanna D.. Systemic sclerosis. Lancet 2017;390:1685–99.
    1. Stern EP, Denton CP.. The pathogenesis of systemic sclerosis. Rheum Dis Clin North Am 2015;41:367–82.
    1. Electronic Medicines Compendium. Ofev 100 mg soft capsules. 2021. (14 July 2021, date last accessed).
    1. US Food and Drug Administration. ACTEMRA prescribing information. 2021. (27 July 2021, date last accessed).
    1. Sullivan KM, Goldmuntz EA, Keyes-Elstein L. et al. Myeloablative autologous stem-cell transplantation for severe scleroderma. N Engl J Med 2018;378:35–47.
    1. van Laar JM, Farge D, Sont JK. et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA 2014;311:2490–8.
    1. Johnson SR, Furst DE.. Current therapeutic approaches in scleroderma: clinical models of effective antifibrotic therapies. Curr Treatm Opt Rheumatol 2020;6:382–93.
    1. Feeney M, Syed F, Khan K. et al. Oncostatin M as a potential molecular target in systemic sclerosis. Arthritis Rheumatol 2015;67: 2297–8.
    1. Stifano G, Sornasse T, Rice LM. et al. Skin gene expression is prognostic for the trajectory of skin disease in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheumatol 2018;70:912–9.
    1. Marden G, Wan Q, Wilks J. et al. The role of the oncostatin M/OSM receptor beta axis in activating dermal microvascular endothelial cells in systemic sclerosis. Arthritis Res Ther 2020;22:179.
    1. Ayaub EA, Dubey A, Imani J. et al. Overexpression of OSM and IL-6 impacts the polarization of pro-fibrotic macrophages and the development of bleomycin-induced lung fibrosis. Sci Rep 2017;7:13281.
    1. Botelho FM, Rodrigues R, Guerette J. et al. Extracellular matrix and fibrocyte accumulation in BALB/c mouse lung upon transient overexpression of oncostatin M. Cells 2019;8:126.
    1. Mozaffarian A, Brewer AW, Trueblood ES. et al. Mechanisms of oncostatin M-induced pulmonary inflammation and fibrosis. J Immunol 2008;181:7243–53.
    1. Matsuda M, Tsurusaki S, Miyata N. et al. Oncostatin M causes liver fibrosis by regulating cooperation between hepatic stellate cells and macrophages in mice. Hepatology 2018;67:296–312.
    1. Reid J, Zamuner S, Edwards K. et al. In vivo affinity and target engagement in skin and blood in a first-time-in-human study of an anti-oncostatin M monoclonal antibody. Br J Clin Pharmacol 2018;84:2280–91.
    1. Allanore Y, Distler O, Jagerschmidt A. et al. Lysophosphatidic acid receptor 1 antagonist SAR100842 for patients with diffuse cutaneous systemic sclerosis: a double-blind, randomized, eight-week placebo-controlled study followed by a sixteen-week open-label extension study. Arthritis Rheumatol 2018;70:1634–43.
    1. Denton CP, Ong VH, Xu S. et al. Therapeutic interleukin-6 blockade reverses transforming growth factor-beta pathway activation in dermal fibroblasts: insights from the faSScinate clinical trial in systemic sclerosis. Ann Rheum Dis 2018;77:1362–71.
    1. Nagahama KY, Togo S, Holz O. et al. Oncostatin M modulates fibroblast function via signal transducers and activators of transcription proteins-3. Am J Respir Cell Mol Biol 2013;49:582–91.
    1. Rice LM, Padilla CM, McLaughlin SR. et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J Clin Invest 2015;125:2795–807.
    1. Khanna D, Denton CP, Jahreis A. et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 2016;387:2630–40.
    1. Rice LM, Ziemek J, Stratton EA. et al. A longitudinal biomarker for the extent of skin disease in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheumatol 2015;67:3004–15.
    1. Abignano G, Blagojevic J, Bissell LA. et al. European multicentre study validates enhanced liver fibrosis test as biomarker of fibrosis in systemic sclerosis. Rheumatology 2019;58:254–9.
    1. Abignano G, Cuomo G, Buch MH. et al. The enhanced liver fibrosis test: a clinical grade, validated serum test, biomarker of overall fibrosis in systemic sclerosis. Ann Rheum Dis 2014;73:420–7.
    1. Akesson A, Scheja A, Lundin A, Wollheim FA.. Improved pulmonary function in systemic sclerosis after treatment with cyclophosphamide. Arthritis Rheum 1994;37:729–35.
    1. Duncan MR, Hasan A, Berman B.. Oncostatin M stimulates collagen and glycosaminoglycan production by cultured normal dermal fibroblasts: insensitivity of sclerodermal and keloidal fibroblasts. J Invest Dermatol 1995;104:128–33.
    1. Guo X, Higgs BW, Bay-Jensen AC. et al. Suppression of T cell activation and collagen accumulation by an anti-IFNAR1 mAb, anifrolumab, in adult patients with systemic sclerosis. J Invest Dermatol 2015;135:2402–9.
    1. Lee YJ, Shin KC, Kang SW. et al. Type III procollagen N-terminal propeptide, soluble interleukin-2 receptor, and von Willebrand factor in systemic sclerosis. Clin Exp Rheumatol 2001;19:69–74.
    1. Quillinan NP, McIntosh D, Vernes J, Haq S, Denton CP.. Treatment of diffuse systemic sclerosis with hyperimmune caprine serum (AIMSPRO): a phase II double-blind placebo-controlled trial. Ann Rheum Dis 2014;73:56–61.
    1. Assassi S, Wu M, Tan FK. et al. Skin gene expression correlates of severity of interstitial lung disease in systemic sclerosis. Arthritis Rheum 2013;65:2917–27.
    1. Johnson SR, Hinchcliff M, Asano Y.. Controversies: molecular vs. clinical systemic sclerosis classification. J Scleroderma Relat Disord 2016;1:277–85.
    1. Johnson SR, Khanna D, Allanore Y, Matucci-Cerinic M, Furst DE.. Systemic sclerosis trial design moving forward. J Scleroderma Relat Disord 2016;1:177–80.
    1. Johnson SR, Tomlinson GA, Granton JT, Hawker GA, Feldman BM.. Applied Bayesian methods in the rheumatic diseases. Rheum Dis Clin North Am 2018;44:361–70.
    1. Akarsu M, Hurşitoğlu M, Toprak Z. et al. Relationships among oncostatin M, insulin resistance, and chronic inflammation: a pilot study. Arch Endocrinol Metab 2020;64:38–44.
    1. Stawski L, Trojanowska M.. Oncostatin M and its role in fibrosis. Connect Tissue Res 2019;60:40–9.
    1. Gearing DP, Bruce AG.. Oncostatin M binds the high-affinity leukemia inhibitory factor receptor. New Biol 1992;4:61–5.
    1. Miyajima A, Kinoshita T, Tanaka M. et al. Role of oncostatin M in hematopoiesis and liver development. Cytokine Growth Factor Rev 2000;11:177–83.
    1. Tanaka M, Hirabayashi Y, Sekiguchi T. et al. Targeted disruption of oncostatin M receptor results in altered hematopoiesis. Blood 2003;102:3154–62.
    1. Wallace PM, Macmaster JF, Rillema JR. et al. In vivo properties of oncostatin M. Ann N Y Acad Sci 1995;762:42–54.

Source: PubMed

3
Abonnieren