337
Views
3
CrossRef citations to date
0
Altmetric
Review

Current immunosuppressive and antifibrotic therapies of systemic sclerosis and emerging therapeutic strategies

ORCID Icon &
Pages 1203-1218 | Received 23 Jul 2020, Accepted 01 Oct 2020, Published online: 19 Oct 2020

References

  • Fuschiotti P. Current perspectives on the immunopathogenesis of systemic sclerosis. Immunotargets Ther. 2016;5:21–35.
  • Distler JHW, Jüngel A, Pileckyte M, et al. Hypoxia-induced increase in the production of extracellular matrix proteins in systemic sclerosis. Arthritis Rheum. 2007;56:4203–4215.
  • LeRoy EC, Black C, Fleischmajer R, et al. Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol. 1988;15:202–205.
  • Nihtyanova SI, Schreiber BE, Ong VH, et al. Prediction of pulmonary complications and long-term survival in systemic sclerosis. Arthritis Rheumatol. 2014;66:1625–1635.
  • Tyndall AJ, Bannert B, Vonk M, et al. Causes and risk factors for death in systemic sclerosis: A study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann Rheum Dis. 2010;69:1809–1815.
  • Volkmann ER, Tashkin DP, Sim M, et al. Short-term progression of interstitial lung disease in systemic sclerosis predicts long-term survival in two independent clinical trial cohorts. Ann Rheum Dis. 2019;78:122–130.
  • Elhai M, Meune C, Avouac J, et al. Trends in mortality in patients with systemic sclerosis over 40 years: A systematic review and meta-analysis of cohort studies. Rheumatol (United Kingdom). 2012;51:1017–1026.
  • Steen VD, Medsger TA. Changes in causes of death in systemic sclerosis, 1972-2002. Ann Rheum Dis. 2007;66:940–944.
  • Van Den Hoogen FHJ, Boerbooms AMT, Swaak AJG, et al. Comparison of methotrexate with placebo in the treatment of systemic sclerosis: A 24 week randomized double-blind trial, followed by a 24 week observational trial. Br J Rheumatol. 1996;35:364–372.
  • Pope JE, Bellamy N, Seibold JR, et al. A randomized, controlled trial of methotrexate versus placebo in early diffuse scleroderma. Arthritis Rheum. 2001;44:1351–1358.
  • Hoyles RK, Ellis RW, Wellsbury J, et al. A multicenter, prospective, randomized, double-blind, placebo-controlled trial of corticosteroids and intravenous cyclophosphamide followed by oral azathioprine for the treatment of pulmonary fibrosis in scleroderma. Arthritis Rheum. 2006;54:3962–3970.
  • Tashkin DP, Elashoff R, Clements PJ, et al. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med. 2006;354:2655–2666.
  • Tashkin DP, Roth MD, Clements PJ, et al. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): a randomised controlled, double-blind, parallel group trial. Lancet Respir Med. 2016;4:708–719.
  • Chakravarty EF, Martyanov V, Fiorentino D, et al. Gene expression changes reflect clinical response in a placebo-controlled randomized trial of abatacept in patients with diffuse cutaneous systemic sclerosis. Arthritis Res Ther. 2015;17:159.
  • Khanna D, Spino C, Johnson S, et al. Abatacept in early diffuse cutaneous systemic sclerosis: results of a Phase II investigator-initiated, multicenter, double-blind, randomized, placebo-controlled trial. Arthritis Rheumatol. 2020;72:125–136.
  • Sircar G, Goswami RP, Sircar D, et al. Intravenous cyclophosphamide vs rituximab for the treatment of early diffuse scleroderma lung disease: open label, randomized, controlled trial. Rheumatol (United Kingdom). 2018;57:2106–2113.
  • Daoussis D, Liossis SNC, Tsamandas AC, et al. Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study. Rheumatology. 2010;49:271–280.
  • Boonstra M, Meijs J, Dorjée AL, et al. Rituximab in early systemic sclerosis. RMD Open. 2017;3:e000384.
  • 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;387:2630–2640.
  • Khanna D, Lin CJF, Furst DE, et al. Tocilizumab in systemic sclerosis: a randomized, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2020;28:S2213-2600(20)30318–0.
  • Burt RK, Shah SJ, Dill K, et al. Autologous non-myeloablative haemopoietic stem-cell transplantation compared with pulse cyclophosphamide once per month for systemic sclerosis (ASSIST): an open-label, randomised phase 2 trial. Lancet. 2011;378:498–506.
  • 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;311:2490–2498. .
  • 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.
  • Distler O, Highland KB, Gahlemann M, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N Engl J Med. 2019;380:2518–2528.
  • Wells AU, Flaherty KR, Brown KK, et al. Nintedanib in patients with progressive fibrosing interstitial lung diseases—subgroup analyses by interstitial lung disease diagnosis in the INBUILD trial: a randomised, double-blind, placebo-controlled, parallel-group trial. Lancet Respir Med. 2020;8:453–460.
  • Humbert M, Coghlan JG, Ghofrani HA, et al. Riociguat for the treatment of pulmonary arterial hypertension associated with connective tissue disease: results from PATENT-1 and PATENT-2. Ann Rheum Dis. 2017;76:422–426.
  • Nagaraja V, Spino C, Bush E, et al. A multicenter randomized, double-blind, placebo-controlled pilot study to assess the efficacy and safety of riociguat in systemic sclerosis-associated digital ulcers. Arthritis Res Ther. 2019;21:202.
  • Distler O, Pope J, Denton C, et al. RISE-SSc: riociguat in diffuse cutaneous systemic sclerosis. Respir Med. 2017;122:S14–7.
  • Khanna D, Albera C, Fischer A, et al. An open-label, phase II study of the safety and tolerability of pirfenidone in patients with scleroderma-associated interstitial lung disease: the LOTUSS trial. J Rheumatol. 2016;43:1672–1679.
  • Acharya N, Sharma SK, Mishra D, et al. Efficacy and safety of pirfenidone in systemic sclerosis-related interstitial lung disease—a randomised controlled trial. Rheumatol Int. 2020;40:703–710.
  • Allanore Y, Denton C, Khanna D et al. Efficacy and Safety of Romilkimab in Diffuse Cutaneous Systemic Sclerosis (dcSSc): A Randomized, Double-Blind, Placebo-Controlled, 24-week, Proof of Concept Study [abstract]. Arthritis Rheumatol [Internet]. 2019;71:(suppl 10). [cited 2020 Jul 22]. Available from: https://acrabstracts.org/abstract/efficacy-and-safety-of-romilkimab-in-diffuse-cutaneous-systemic-sclerosis-dcssc-a-randomized-double-blind-placebo-controlled-24-week-proof-of-concept-study/
  • Lafyatis R, Mantero JC, Gordon J, et al. Inhibition of β-catenin signaling in the skin rescues cutaneous adipogenesis in systemic sclerosis: a randomized, double-blind, placebo-controlled trial of C-82. J Invest Dermatol. 2017;137:2473–2483.
  • Johnson SR, Feldman BM, Pope JE, et al. Shifting our thinking about uncommon disease trials: the case of methotrexate in scleroderma. J Rheumatol. 2009;36:323–329.
  • Kowal-Bielecka O, Fransen J, Avouac J, et al. Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76:1327–1339.
  • Åkesson A, Scheja A, Lundin A, et al. Improved pulmonary function in systemic sclerosis after treatment with cyclophosphamide. Arthritis Rheum. 1994;37:729–735.
  • White B, Moore WC, Wigley FM, et al. Cyclophosphamide is associated with pulmonary function and survival benefit in patients with scleroderma and alveolitis. Ann Intern Med. 2000;132:947–954.
  • Tashkin DP, Elashoff R, Clements PJ, et al. Effects of 1-year treatment with cyclophosphamide on outcomes at 2 years in scleroderma lung disease. Am J Respir Crit Care Med. 2007;176:1026–1034.
  • Namas R, Tashkin DP, Furst DE, et al. Efficacy of mycophenolate mofetil and oral cyclophosphamide on skin thickness: post hoc analyses from two randomized placebo-controlled trials. Arthritis Care Res. 2018;70:439–444.
  • Omair MA, Alahmadi A, Johnson SR. Safety and effectiveness of mycophenolate in systemic sclerosis. A systematic review. PLoS One. 2015;10:e0124205.
  • Naidu GSRSNK, Sharma SK, Adarsh MB, et al. Effect of mycophenolate mofetil (MMF) on systemic sclerosis-related interstitial lung disease with mildly impaired lung function: a double-blind, placebo-controlled, randomized trial. Rheumatol Int. 2020;40:207–216.
  • Fernández-Codina A, Walker KM, Pope JE. Treatment algorithms for systemic sclerosis according to experts. Arthritis Rheumatol. 2018;70:1820–1828.
  • Sewell WAC, Jolles S. Immunomodulatory action of intravenous immunoglobulin. Immunology. 2002;107:387–393.
  • Cherin P, Herson S, Wechsler B, et al. Efficacy of intravenous gammaglobulin therapy in chronic refractory polymyositis and dermatomyositis: an open study with 20 adult patients. Am J Med. 1991;91:162–168.
  • Cherin P, Piette JC, Wechsler B, et al. Intravenous gamma globulin as first line therapy in polymyositis and dermatomyositis: an open study in 11 adult patients. J Rheumatol. 1994;21:1092–1097.
  • Kivity S, Katz U, Daniel N, et al. Evidence for the use of intravenous immunoglobulins - A review of the literature. Clin Rev Allergy Immunol. 2010;38:201–269.
  • Kajii M, Suzuki C, Kashihara J, et al. Prevention of excessive collagen accumulation by human intravenous immunoglobulin treatment in a murine model of bleomycin-induced scleroderma. Clin Exp Immunol. 2011;163:235–241.
  • Levy Y, Amital H, Langevitz P, et al. Intravenous immunoglobulin modulates cutaneous involvement and reduces skin fibrosis in systemic sclerosis: an open-label study. Arthritis Rheum. 2004;50:1005–1007.
  • Poelman CL, Hummers LK, Wigley FM, et al. Intravenous immunoglobulin may be an effective therapy for refractory, active diffuse cutaneous systemic sclerosis. J Rheumatol. 2015;42:236–242.
  • Sanges S, Rivière S, Mekinian A, et al. Intravenous immunoglobulins in systemic sclerosis: data from a French nationwide cohort of 46 patients and review of the literature. Autoimmun Rev. 2017;16:377–384.
  • Chaigne B, Rodeia S, Benmostefa N, et al. Corticosteroid-sparing benefit of intravenous immunoglobulin in systemic sclerosis-associated myopathy: A comparative study in 52 patients. Autoimmun Rev. 2020. DOI:https://doi.org/10.1016/j.autrev.2019.102431
  • Takehara K, Ihn H, Sato S. A randomised, double-blind, placebo-controlled trial: intravenous immunoglobulin treatment in patients with diffuse cutaneous systemic sclerosis. Clin Exp Rheumatol. 2013;31:151–156.
  • Kalogerou A, Gelou E, Mountantonakis S, et al. Early T cell activation in the skin from patients with systemic sclerosis. Ann Rheum Dis. 2005;64:1233–1235.
  • Truchetet ME, Brembilla NC, Montanari E, et al. Increased frequency of circulating Th22 in addition to Th17 and Th2 lymphocytes in systemic sclerosis: association with interstitial lung disease. Arthritis Res Ther. 2011;13:R166.
  • Postlethwaite AE, Holness MA, Katai H, et al. Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4. J Clin Invest. 1992;90:1479–1485.
  • Khan K, Xu S, Nihtyanova S, et al. Clinical and pathological significance of interleukin 6 overexpression in systemic sclerosis. Ann Rheum Dis. 2012;71:1235–1242.
  • O’Reilly S, Hügle T, Van Laar JM. T cells in systemic sclerosis: A reappraisal. Rheumatol (United Kingdom). 2012;51:1540–1549.
  • Ponsoye M, Frantz C, Ruzehaji N, et al. Treatment with abatacept prevents experimental dermal fibrosis and induces regression of established inflammation-driven fibrosis. Ann Rheum Dis. 2016;75:2142–2149.
  • De Paoli FV, Nielsen BD, Rasmussen F, et al. Abatacept induces clinical improvement in patients with severe systemic sclerosis. Scand J Rheumatol. 2014;43:342–345.
  • Castellví I, Elhai M, Bruni C, et al. Safety and effectiveness of abatacept in systemic sclerosis: the EUSTAR experience. Semin Arthritis Rheum. 2020;S0049-0172(20)30014-7. DOI:https://doi.org/10.1016/j.semarthrit.2019.12.004.
  • Okano Y. Antinuclear antibody in systemic sclerosis (scleroderma). Rheum Dis Clin North Am. 1996;22:709–735.
  • Sato S, Fujimoto M, Hasegawa M, et al. Altered blood B lymphocyte homeostasis in systemic sclerosis: expanded naive B cells and diminished but activated memory B cells. Arthritis Rheum. 2004;50:1918–1927.
  • Famularo G, Giacomelli R, Alesse E, et al. Polyclonal B lymphocyte activation in progressive systemic sclerosis. J Clin Lab Immunol. 1989;29:59–63.
  • Hasegawa M, Fujimoto M, Takehara K, et al. Pathogenesis of systemic sclerosis: altered B cell function is the key linking systemic autoimmunity and tissue fibrosis. J Dermatol Sci. 2005;39:1–7.
  • Lafyatis R, O’Hara C, Feghali-Bostwick CA, et al. B cell infiltration in systemic sclerosis-associated interstitial lung disease. Arthritis Rheum. 2007;56:3167–3168.
  • Ebata S, Yoshizaki A, Fukasawa T, et al. Rituximab therapy is more effective than cyclophosphamide therapy for Japanese patients with anti-topoisomerase I-positive systemic sclerosis-associated interstitial lung disease. J Dermatol. 2019;46:1006–1013.
  • Daoussis D, Melissaropoulos K, Sakellaropoulos G, et al. A multicenter, open-label, comparative study of B-cell depletion therapy with Rituximab for systemic sclerosis-associated interstitial lung disease. Semin Arthritis Rheum. 2017;46:625–631.
  • Daoussis D, Liossis SNC, Tsamandas AC, et al. Effect of long-term treatment with rituximab on pulmonary function and skin fibrosis in patients with diffuse systemic sclerosis. Clin Exp Rheumatol. 2012;30:S17–22.
  • Bosello SL, De Luca G, Rucco M, et al. Long-term efficacy of B cell depletion therapy on lung and skin involvement in diffuse systemic sclerosis. Semin Arthritis Rheum. 2015;44:428–436.
  • Smith V, Van Praet JT, Vandooren B, et al. Rituximab in diffuse cutaneous systemic sclerosis: an open-label clinical and histopathological study. Ann Rheum Dis. 2010;69:193–197.
  • Smith V, Piette Y, Van Praet JT, et al. Two-year results of an open pilot study of a 2-treatment course with rituximab in patients with early systemic sclerosis with diffuse skin involvement. J Rheumatol. 2013;40:52–57.
  • Jordan S, Distler JHW, Maurer B, et al. Effects and safety of rituximab in systemic sclerosis: an analysis from the European Scleroderma Trial and Research (EUSTAR) group. Ann Rheum Dis. 2015;74:1188–1194.
  • Elhai M, Boubaya M, Distler O, et al. Outcomes of patients with systemic sclerosis treated with rituximab in contemporary practice: A prospective cohort study. Ann Rheum Dis. 2019;78:979–987.
  • Stohl W, Hilbert DM. The discovery and development of belimumab: the anti-BLyS-lupus connection. Nat Biotechnol. 2012;30:69–77.
  • Gordon JK, Martyanov V, Franks JM, et al. Belimumab for the treatment of early diffuse systemic sclerosis: results of a randomized, double-blind, placebo-controlled, pilot trial. Arthritis Rheumatol. 2018;70:308–316.
  • Kadono T, Kikuchi K, Ihn H, et al. Increased production of interleukin 6 and interleukin 8 in scleroderma fibroblasts. J Rheumatol. 1998;25:296–301.
  • Hasegawa M, Sato S, Ihn H, et al. Enhanced production of interleukin-6 (IL-6), oncostatin M and soluble IL-6 receptor by cultured peripheral blood mononuclear cells from patients with systemic sclerosis. Rheumatology. 1999;38:612–617.
  • Sato S, Hasegawa M, Takehara K. Serum levels of interleukin-6 and interleukin-10 correlate with total skin thickness score in patients with systemic sclerosis. J Dermatol Sci. 2001;27:140–146.
  • De Lauretis A, Sestini P, Pantelidis P, et al. Serum interleukin 6 is predictive of early functional decline and mortality in interstitial lung disease associated with systemic sclerosis. J Rheumatol. 2013;40:435–446.
  • Shima Y, Kuwahara Y, Murota H, et al. The skin of patients with systemic sclerosis softened during the treatment with anti-IL-6 receptor antibody tocilizumab. Rheumatology. 2010;49:2408–2412.
  • Shima Y, Hosen N, Hirano T, et al. Expansion of range of joint motion following treatment of systemic sclerosis with tocilizumab. Mod Rheumatol. 2015;25:134–137.
  • Kondo M, Murakawa Y, Matsumura T, et al. A case of overlap syndrome successfully treated with tocilizumab: a hopeful treatment strategy for refractory dermatomyositis? Rheumatology (Oxford). 2014;53:1907–1908.
  • Elhai M, Meunier M, Matucci-Cerinic M, et al. Outcomes of patients with systemic sclerosis-associated polyarthritis and myopathy treated with tocilizumab or abatacept: A EUSTAR observational study. Ann Rheum Dis. 2013;72:1217–1220.
  • Szodoray P, Varoczy L, Szegedi G, et al. Autologous stem cell transplantation in autoimmune and rheumatic diseases: from the molecular background to clinical applications. Scand J Rheumatol. 2010;39:1–11.
  • Baraut J, Grigore EI, Jean-Louis F, et al. Peripheral blood regulatory T cells in patients with diffuse systemic sclerosis (SSc) before and after autologous hematopoietic SCT: A pilot study. Bone Marrow Transplant. 2014;49:349–354.
  • Farge D, Passweg J, Van Laar JM, et al. Autologous stem cell transplantation in the treatment of systemic sclerosis: report from the EBMT/EULAR registry. Ann Rheum Dis. 2004;63:974–981.
  • Nash RA, McSweeney PA, Crofford LJ, et al. High-dose immunosuppressive therapy and autologous hematopoietic cell transplantation for severe systemic sclerosis: long-term follow-up of the US multicenter pilot study. Blood. 2007;110:1388–1396.
  • Vonk MC, Marjanovic Z, Van Den Hoogen FHJ, et al. Long-term follow-up results after autologous haematopoietic stem cell transplantation for severe systemic sclerosis. Ann Rheum Dis. 2008;67:98–104.
  • Burt RK, Han X, Quigley K, et al. Cardiac safe hematopoietic stem cell transplantation for systemic sclerosis with poor cardiac function: a pilot safety study that decreases neutropenic interval to 5 days. Bone Marrow Transplant. 2020. DOI:https://doi.org/10.1038/s41409-020-0978-2
  • Bhattacharyya S, Ishida W, Wu M, et al. A non-Smad mechanism of fibroblast activation by transforming growth factor-β via c-Abl and Egr-1: selective modulation by imatinib mesylate. Oncogene. 2009;28:1285–1297.
  • Daniels CE, Wilkes MC, Edens M, et al. Imatinib mesylate inhibits the profibrogenic activity of TGF-β and prevents bleomycin-mediated lung fibrosis. J Clin Invest. 2004;114:1308–1316.
  • Huang J, Beyer C, Palumbo-Zerr K, et al. Nintedanib inhibits fibroblast activation and ameliorates fibrosis in preclinical models of systemic sclerosis. Ann Rheum Dis. 2016;75:883–890.
  • Huang J, Maier C, Zhang Y, et al. Nintedanib inhibits macrophage activation and ameliorates vascular and fibrotic manifestations in the Fra2 mouse model of systemic sclerosis. Ann Rheum Dis. 2017;76:1941–1948.
  • Dees C, Beyer C, Distler A, et al. Stimulators of soluble guanylate cyclase (sGC) inhibit experimental skin fibrosis of different aetiologies. Ann Rheum Dis. 2015;74:1621–1625.
  • Beyer C, Zenzmaier C, Palumbo-Zerr K, et al. Stimulation of the soluble guanylate cyclase (sGC) inhibits fibrosis by blocking non-canonical TGFβ signalling. Ann Rheum Dis. 2015;74:1408–1416.
  • Du J, Paz K, Flynn R, et al. Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production. Blood. 2017;129:2570–2580.
  • Schaefer CJ, Ruhrmund DW, Pan L, et al. Antifibrotic activities of pirfenidone in animal models. Eur RespirRev. 2011;20:85–97.
  • Nakazato H, Oku H, Yamane S, et al. A novel anti-fibrotic agent pirfenidone suppresses tumor necrosis factor-α at the translational level. Eur J Pharmacol. 2002;446:177–185.
  • Iyer SN, Hyde DM, Giri SN. Anti-inflammatory effect of pirfenidone in the bleomycin- hamster model of lung inflammation. Inflammation. 2000;24:477–491.
  • Miura Y, Saito T, Fujita K, et al. Clinical experience with pirfenidone in five patients with scleroderma-related interstitial lung disease. Sarcoidosis Vasc Diffus Lung Dis. 2014;31:235–238.
  • Fraticelli P, Gabrielli B, Pomponio G, et al. Low-dose oral imatinib in the treatment of systemic sclerosis interstitial lung disease unresponsive to cyclophosphamide: A phase II pilot study. Arthritis Res Ther. 2014;16:R144.
  • Pope J, McBain D, Petrlich L, et al. Imatinib in active diffuse cutaneous systemic sclerosis: results of a six-month, randomized, double-blind, placebo-controlled, proof-of-concept pilot study at a single center. Arthritis Rheum. 2011;63:3547–3551.
  • Manley PW, Drueckes P, Fendrich G, et al. Extended kinase profile and properties of the protein kinase inhibitor nilotinib. Biochim Biophys Acta - Proteins Proteomics. 2010;1804:445–453.
  • Akhmetshina A, Dees C, Pileckyte M, et al. Dual inhibition of c‐abl and PDGF receptor signaling by dasatinib and nilotinib for the treatment of dermal fibrosis. Faseb J. 2008;22:2214–2222.
  • Gordon JK, Martyanov V, Magro C, et al. Nilotinib (TasignaTM) in the treatment of early diffuse systemic sclerosis: an open-label, pilot clinical trial. Arthritis Res Ther. 2015;17:213.
  • Martyanov V, Kim GHJ, Hayes W, et al. Novel lung imaging biomarkers and skin gene expression subsetting in dasatinib treatment of systemic sclerosis-associated interstitial lung disease. PLoS One. 2017;12:e0187580.
  • Nemeth JA, Nakada MT, Trikha M, et al. Alpha-v integrins as therapeutic targets in oncology. Cancer Invest. 2007;25:632–646.
  • Asano Y, Ihn H, Yamane K, et al. Increased expression of integrin αvβ5 induces the myofibroblastic differentiation of dermal fibroblasts. Am J Pathol. 2006;168:499–510.
  • Asano Y, Ihn H, Yamane K, et al. Increased expression of integrin α v β 3 contributes to the establishment of autocrine TGF-β signaling in scleroderma fibroblasts. J Immunol. 2005;175:7708–7718.
  • Wirth M, Heidenreich A, Gschwend JE, et al. A multicenter phase 1 study of EMD 525797 (DI17E6), a novel humanized monoclonal antibody targeting αv integrins, in progressive castration-resistant prostate cancer with bone metastases after chemotherapy. Eur Urol. 2014;65:897–904.
  • Hussain M, Le Moulec S, Gimmi C, et al. Differential effect on bone lesions of targeting integrins: randomized phase II trial of abituzumab in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res. 2016;22:3192–3200.
  • Pellom ST, Dudimah DF, Thounaojam MC, et al. Modulatory effects of bortezomib on host immune cell functions. Immunotherapy. 2015;7:1011–1022.
  • Mutlu GM, Budinger GRS, Wu M, et al. Proteasomal inhibition after injury prevents fibrosis by modulating TGF-β1 signalling. Thorax. 2012;67:139–146.
  • Bachhuber BG, Sarembock IJ, Gimple LW, et al. α-Thrombin induces transforming growth factor-β1 mrna and protein in cultured vascular smooth muscle cells via a proteolytically activated receptor. J Vasc Res. 1997;34:41–48.
  • Chambers RC, Leoni P, Blanc-Brude OP, et al. Thrombin is a potent inducer of connective tissue growth factor production via proteolytic activation of protease-activated receptor-1. J Biol Chem. 2000;275:35584–35591.
  • Bogatkevich GS, Tourkina E, Silver RM, et al. Thrombin differentiates normal lung fibroblasts to a myofibroblast phenotype via the proteolytically activated Receptor-1 and a protein Kinase C-dependent pathway. J Biol Chem. 2001;276:45184–45192.
  • Wienen W, Stassen JM, Priepke H, et al. In-vitro profile and ex-vivo anticoagulant activity of the direct thrombin inhibitor dabigatran and its orally active prodrug, dabigatran etexilate. Thromb Haemost. 2007;98:155–162.
  • Bogatkevich GS, Ludwicka-Bradley A, Silver RM. Dabigatran, a direct thrombin inhibitor, demonstrates antifibrotic effects on lung fibroblasts. Arthritis Rheum. 2009;60:3455–3464.
  • Bogatkevich GS, Ludwicka-Bradley A, Nietert PJ, et al. Antiinflammatory and antifibrotic effects of the oral direct thrombin inhibitor dabigatran etexilate in a murine model of interstitial lung disease. Arthritis Rheum. 2011;63:1416–1425.
  • Silver RM, Wilson DA, Akter T, et al. Safety and tolerability of thrombin inhibition in scleroderma-associated interstitial lung disease. ACR Open Rheumatol. 2019;1:403–411.
  • Penttinen RP, Kobayashi S, Bornstein P. Transforming growth factor β increases mRNA for matrix proteins both in the presence and in the absence of changes in mRNA stability. Proc Natl Acad Sci U S A. 1988;85:1105–1108.
  • Denton CP, Merkel PA, Furst DE, et al. Recombinant human anti-transforming growth factor β1 antibody therapy in systemic sclerosis: A multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum. 2007;56:323–333.
  • Rice LM, Padilla CM, McLaughlin SR, et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J Clin Invest. 2015;125:2795–2807.
  • Murray LA, Zhang H, Oak SR, et al. Targeting interleukin-13 with tralokinumab attenuates lung fibrosis and epithelial damage in a humanized SCID idiopathic pulmonary fibrosis model. Am J Respir Cell Mol Biol. 2014;50:985–994.
  • Burstein SH, Zurier RB. Cannabinoids, endocannabinoids, and related analogs in inflammation. Aaps J. 2009;11:109–119.
  • Garcia-Gonzalez E, Selvi E, Balistreri E, et al. Cannabinoids inhibit fibrogenesis in diffuse systemic sclerosis fibroblasts. Rheumatology. 2009;48:1050–1056.
  • Balistreri E, Garcia-Gonzalez E, Selvi E, et al. The cannabinoid WIN55,212-2 abrogates dermal fibrosis in scleroderma bleomycin model. Ann Rheum Dis. 2011;70:695–699.
  • Spiera R, Hummers L, Chung L, et al. Safety and efficacy of lenabasum in a phase 2 randomized, placebo-controlled trial in adults with systemic sclerosis. Arthritis Rheumatol. 2020. DOI:https://doi.org/10.1002/art.41294
  • Sokol SY. Spatial and temporal aspects of Wnt signaling and planar cell polarity during vertebrate embryonic development. Semin Cell Dev Biol. 2015;42:78–85.
  • Mirabelli CK, Nusse R, Tuveson DA, et al. Perspectives on the role of Wnt biology in cancer. Sci Signal. 2019;12:eaay4494.
  • Wei J, Melichian D, Komura K, et al. Canonical Wnt signaling induces skin fibrosis and subcutaneous lipoatrophy: A novel mouse model for scleroderma? Arthritis Rheum. 2011;63:1707–1717.
  • Wei J, Fang F, Lam AP, et al. Wnt/β-catenin signaling is hyperactivated in systemic sclerosis and induces Smad-dependent fibrotic responses in mesenchymal cells. Arthritis Rheum. 2012;64:2734–2745.
  • Frost J, Estivill X, Ramsay M, et al. Dysregulation of the Wnt signaling pathway in South African patients with diffuse systemic sclerosis. Clin Rheumatol. 2019;38:933–938.
  • Dees C, Akhmetshina A, Zerr P, et al. Platelet-derived serotonin links vascular disease and tissue fibrosis. J Exp Med. 2011;208:961–972.
  • Chaturvedi S, Misra DP, Prasad N, et al. 5-HT 2 and 5-HT 2B antagonists attenuate pro-fibrotic phenotype in human adult dermal fibroblasts by blocking TGF-β1 induced non-canonical signaling pathways including STAT3: implications for fibrotic diseases like scleroderma. Int J Rheum Dis. 2018;21:2128–2138.
  • Distler O, Maurer B, Vettori S, et al. The serotonin receptor 2 inhibitor terguride has beneficial effects on skin fibrosis: results from a phase 2 proof of concept study. Arthritis Rheumatol[Internet]. 2016;68:(suppl 10). [cited 2020 Jul 22]. Available from: https://acrabstracts.org/abstract/the-serotonin-receptor-2-inhibitor-terguride-has-beneficial-effects-on-skin-fibrosis-results-from-a-phase-2-proof-of-concept-study/
  • Ghosh AK, Bhattacharyya S, Wei J, et al. Peroxisome proliferator-activated receptor-γ abrogates Smad-dependent collagen stimulation by targeting the p300 transcriptional coactivator. Faseb J. 2009;23:2968–2977.
  • Wei J, Ghosh AK, Sargent JL, et al. PPARγ downregulation by TGFß in fibroblast and impaired expression and function in systemic sclerosis: a novel mechanism for progressive fibrogenesis. PLoS One. 2015;74(11):e13778.
  • Wu M, Melichian DS, Chang E, et al. Rosiglitazone abrogates bleomycin-induced scleroderma and blocks profibrotic responses through peroxisome proliferator-activated receptor-β. Am J Pathol. 2009;174:519–533.
  • Ruzehaji N, Frantz C, Ponsoye M, et al. Pan PPAR agonist IVA337 is effective in prevention and treatment of experimental skin fibrosis. Ann Rheum Dis. 2016;75:2175–2183.
  • Dees C, Tomcik M, Palumbo-Zerr K, et al. JAK-2 as a novel mediator of the profibrotic effects of transforming growth factor β in systemic sclerosis. Arthritis Rheum. 2012;64:3006–3015.
  • Lescoat A, Lelong M, Jeljeli M, et al. Combined anti-fibrotic and anti-inflammatory properties of JAK-inhibitors on macrophages in vitro and in vivo: perspectives for scleroderma-associated interstitial lung disease. Biochem Pharmacol. 2020;178:114103.
  • da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119:2204–2213.
  • Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res Ther. 2016;7:125.
  • Ruiz M, Cosenza S, Maumus M, et al. Therapeutic application of mesenchymal stem cells in osteoarthritis. Expert Opin Biol Ther. 2016;16:33–42.
  • Cras A, Farge D, Carmoi T, et al. Update on mesenchymal stem cell-based therapy in lupus and scleroderma. Arthritis Res Ther. 2015;17:301.
  • Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003;100:8407–8411.
  • Moodley Y, Atienza D, Manuelpillai U, et al. Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol. 2009;175:303–313.
  • Wu Y, Huang S, Enhe J, et al. Bone marrow-derived mesenchymal stem cell attenuates skin fibrosis development in mice. Int Wound J. 2014;11:701–710.
  • Maria ATJ, Toupet K, Bony C, et al. Antifibrotic, antioxidant, and immunomodulatory effects of mesenchymal stem cells in HOCl-induced systemic sclerosis. Arthritis Rheumatol. 2016;68:1013–1025.
  • Chen C, Wang D, Moshaverinia A, et al. Mesenchymal stem cell transplantation in tight-skin mice identifies miR-151-5p as a therapeutic target for systemic sclerosis. Cell Res. 2017;27:559–577.
  • Rozier P, Maria A, Goulabchand R, et al. Mesenchymal stem cells in systemic sclerosis: allogenic or autologous approaches for therapeutic use?. Front Immunol. 2018;9:2938.
  • Christopeit M, Schendel M, Föll J, et al. Marked improvement of severe progressive systemic sclerosis after transplantation of mesenchymal stem cells from an allogeneic haploidentical-related donor mediated by ligation of CD137L. Leukemia. 2008;22:1062–1064.
  • Keyszer G, Christopeit M, Fick S, et al. Treatment of severe progressive systemic sclerosis with transplantation of mesenchymal stromal cells from allogeneic related donors: report of five cases. Arthritis Rheum. 2011;63:2540–2542.
  • Granel B, Daumas A, Jouve E, et al. Safety, tolerability and potential efficacy of injection of autologous adipose-derived stromal vascular fraction in the fingers of patients with systemic sclerosis: an open-label phase I trial. Ann Rheum Dis. 2015;74:2175–2182.
  • Daumas A, Magalon J, Jouve E, et al. Long-term follow-up after autologous adipose-derived stromal vascular fraction injection into fingers in systemic sclerosis patients. Curr Res Transl Med. 2017;65:40–43.
  • Nihtyanova SI, Tang EC, Coghlan JG, et al. Improved survival in systemic sclerosis is associated with better ascertainment of internal organ disease: A retrospective cohort study. QJM. 2010;103:109–115.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.