Abstract
Systemic sclerosis (SSc) remains a challenging and enigmatic systemic autoimmune disease, owing to its complex pathogenesis, clinical and molecular heterogeneity, and the lack of effective disease-modifying treatments. Despite a century of research in SSc, the interconnections among microvascular dysfunction, autoimmune phenomena and tissue fibrosis in SSc remain unclear. The absence of validated biomarkers and reliable animal models complicates diagnosis and treatment, contributing to high morbidity and mortality. Advances in the past 5 years, such as single-cell RNA sequencing, next-generation sequencing, spatial biology, transcriptomics, genomics, proteomics, metabolomics, microbiome profiling and artificial intelligence, offer new avenues for identifying the early pathogenetic events that, once treated, could change the clinical history of SSc. Collaborative global efforts to integrate these approaches are crucial to developing a comprehensive, mechanistic understanding and enabling personalized therapies. Challenges include disease classification, clinical heterogeneity and the establishment of robust biomarkers for disease activity and progression. Innovative clinical trial designs and patient-centred approaches are essential for developing effective treatments. Emerging therapies, including cell-based and fibroblast-targeting treatments, show promise. Global cooperation, standardized protocols and interdisciplinary research are vital for advancing SSc research and improving patient outcomes. The integration of advanced research techniques holds the potential for important breakthroughs in the diagnosis, treatment and care of individuals with SSc.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$189.00 per year
only $15.75 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout




Similar content being viewed by others
Change history
03 March 2025
A Correction to this paper has been published: https://doi.org/10.1038/s41584-025-01231-y
References
Denton, C. P. & Khanna, D. Systemic sclerosis. Lancet 390, 1685–1699 (2017).
Volkmann, E. R., Andreasson, K. & Smith, V. Systemic sclerosis. Lancet 401, 304–318 (2023).
Elhai, M. et al. Mapping and predicting mortality from systemic sclerosis. Ann. Rheum. Dis. 76, 1897–1905 (2017).
Park, E. H., Strand, V., Oh, Y. J., Song, Y. W. & Lee, E. B. Health-related quality of life in systemic sclerosis compared with other rheumatic diseases: a cross-sectional study. Arthritis Res. Ther. 21, 61 (2019).
Osler, W. On diffuse scleroderma: with special reference to diagnosis and to the use of thyroid-gland extract. J. Cutan. Dis. 16, 49 (1898).
Matsui, S. Pathology and pathogenesis of universal scleroderma. Mitt. Med. Fak. Univ. Zu Tokyo 31, 55 (1924).
Bi, X., Mills, T. & Wu, M. Animal models in systemic sclerosis: an update. Curr. Opin. Rheumatol. 35, 364–370 (2023).
Inventiva. Inventiva announces results from phase IIb clinical trial with lanifibranor in systemic sclerosis. GlobeNewswire https://www.globenewswire.com/news-release/2019/02/18/1733864/0/en/Inventiva-Announces-Results-From-Phase-IIb-Clinical-Trial-with-Lanifibranor-in-Systemic-Sclerosis.html (18 February 2019).
Roofeh, D. et al. Tocilizumab prevents progression of early systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol. 73, 1301–1310 (2021).
Khanna, D. et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir. Med. 8, 963–974 (2020).
Distler, O. et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. N. Engl. J. Med. 380, 2518–2528 (2019).
Abraham, D., Lescoat, A. & Stratton, R. Emerging diagnostic and therapeutic challenges for skin fibrosis in systemic sclerosis. Mol. Aspects Med. 96, 101252 (2024).
Rice, L. M. et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J. Clin. Invest. 125, 2795–2807 (2015).
Milano, A. et al. Molecular subsets in the gene expression signatures of scleroderma skin. PLoS ONE 3, e2696 (2008).
Korman, B. Evolving insights into the cellular and molecular pathogenesis of fibrosis in systemic sclerosis. Transl. Res. 209, 77–89 (2019).
Asano, Y. The pathogenesis of systemic sclerosis: an understanding based on a common pathologic cascade across multiple organs and additional organ-specific pathologies. J. Clin. Med. 9, 2687 (2020).
Gabrielli, A., Avvedimento, E. V. & Krieg, T. Scleroderma. N. Engl. J. Med. 360, 1989–2003 (2009).
Dees, C., Chakraborty, D. & Distler, J. H. W. Cellular and molecular mechanisms in fibrosis. Exp. Dermatol. 30, 121–131 (2021).
Volkmann, E. R. & Varga, J. Emerging targets of disease-modifying therapy for systemic sclerosis. Nat. Rev. Rheumatol. 15, 208–224 (2019).
Gronberg, C. et al. Combined inhibition of IL-1, IL-33 and IL-36 signalling by targeting IL1RAP ameliorates skin and lung fibrosis in preclinical models of systemic sclerosis. Ann. Rheum. Dis. 83, 1156–1168 (2024).
Huang, M. et al. Single-cell transcriptomes and chromatin accessibility of endothelial cells unravel transcription factors associated with dysregulated angiogenesis in systemic sclerosis. Ann. Rheum. Dis. 83, 1335–1344 (2024).
Gaydosik, A. M. et al. Single-cell transcriptome analysis identifies skin-specific T-cell responses in systemic sclerosis. Ann. Rheum. Dis. 80, 1453–1460 (2021).
Zhu, H. et al. Fibroblast subpopulations in systemic sclerosis: functional implications of individual subpopulations and correlations with clinical features. J. Invest. Dermatol. 144, 1251–1261.e13 (2024).
Teaw, S., Hinchcliff, M. & Cheng, M. A review and roadmap of the skin, lung and gut microbiota in systemic sclerosis. Rheumatology 60, 5498–5508 (2021).
Yao, Q., Tan, W. & Bai, F. Gut microbiome and metabolomics in systemic sclerosis: feature, link and mechanisms. Front. Immunol. 15, 1475528 (2024).
Tan, T. C., Noviani, M., Leung, Y. Y. & Low, A. H. L. The microbiome and systemic sclerosis: a review of current evidence. Best. Pract. Res. Clin. Rheumatol. 35, 101687 (2021).
Russo, E. et al. The differential crosstalk of the skin–gut microbiome axis as a new emerging actor in systemic sclerosis. Rheumatology 63, 226–234 (2024).
Volkmann, E. R. Is there a role for the microbiome in systemic sclerosis? Expert. Rev. Clin. Immunol. 19, 237–240 (2023).
Volkmann, E. R. et al. Longitudinal characterisation of the gastrointestinal tract microbiome in systemic sclerosis. Eur. Med. J. 7, 110–118 (2020).
Kim, S. J. et al. Gut microbe-derived metabolite trimethylamine N-oxide activates PERK to drive fibrogenic mesenchymal differentiation. iScience 25, 104669 (2022).
Lopez-Isac, E. et al. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways. Nat. Commun. 10, 4955 (2019).
Acosta-Herrera, M. et al. Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Ann. Rheum. Dis. 78, 311–319 (2019).
Gourh, P. et al. HLA and autoantibodies define scleroderma subtypes and risk in African and European Americans and suggest a role for molecular mimicry. Proc. Natl Acad. Sci. USA 117, 552–562 (2020).
Hinchcliff, M. et al. Cellular and molecular diversity in scleroderma. Semin. Immunol. 58, 101648 (2021).
Ouchene, L. et al. Toward understanding of environmental risk factors in systemic sclerosis. J. Cutan. Med. Surg. 25, 188–204 (2021).
Vijayraghavan, S. et al. Widespread mutagenesis and chromosomal instability shape somatic genomes in systemic sclerosis. Nat. Commun. 15, 8889 (2024).
McMahan, Z. H. et al. Systemic sclerosis gastrointestinal dysmotility: risk factors, pathophysiology, diagnosis and management. Nat. Rev. Rheumatol. 19, 166–181 (2023).
Denton, C. P., Wells, A. U. & Coghlan, J. G. Major lung complications of systemic sclerosis. Nat. Rev. Rheumatol. 14, 511–527 (2018).
Humbert, M. et al. Pathology and pathobiology of pulmonary hypertension: state of the art and research perspectives. Eur. Respir. J. 53, 1801887 (2019).
Bahi, M., Li, C., Wang, G. & Korman, B. D. Systemic sclerosis-associated pulmonary arterial hypertension: from bedside to bench and back again. Int. J. Mol. Sci. 25, 4728 (2024).
Distler, J. H. W., Riemekasten, G. & Denton, C. P. The exciting future for scleroderma: what therapeutic pathways are on the horizon? Rheum. Dis. Clin. North. Am. 49, 445–462 (2023).
Lescoat, A., Varga, J., Matucci-Cerinic, M. & Khanna, D. New promising drugs for the treatment of systemic sclerosis: pathogenic considerations, enhanced classifications, and personalized medicine. Expert. Opin. Investig. Drugs 30, 635–652 (2021).
Peclat, T. R., Shi, B., Varga, J. & Chini, E. N. The NADase enzyme CD38: an emerging pharmacological target for systemic sclerosis, systemic lupus erythematosus and rheumatoid arthritis. Curr. Opin. Rheumatol. 32, 488–496 (2020).
Lescoat, A., Kato, H. & Varga, J. Emerging cellular and immunotherapies for systemic sclerosis: from mesenchymal stromal cells to CAR-T cells and vaccine-based approaches. Curr. Opin. Rheumatol. 35, 356–363 (2023).
Tsou, P. S., Varga, J. & O’Reilly, S. Advances in epigenetics in systemic sclerosis: molecular mechanisms and therapeutic potential. Nat. Rev. Rheumatol. 17, 596–607 (2021).
Khanna, D. et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): a phase 2, randomised, controlled trial. Lancet 387, 2630–2640 (2016).
Butler, E. A. et al. Generation of a core set of items to develop classification criteria for scleroderma renal crisis using consensus methodology. Arthritis Rheumatol. 71, 964–971 (2019).
Fairley, J. L., Ross, L. & Nikpour, M. Heart involvement in systemic sclerosis: emerging concepts. Curr. Opin. Rheumatol. 36, 393–400 (2024).
Batani, V., Dagna, L. & De Luca, G. Therapeutic strategies for primary heart involvement in systemic sclerosis. Rheumatol. Immunol. Res. 5, 72–82 (2024).
Usategui, A. et al. Evidence of telomere attrition and a potential role for DNA damage in systemic sclerosis. Immun. Ageing 19, 7 (2022).
Shi, B. et al. Senescent cells accumulate in systemic sclerosis skin. J. Invest. Dermatol. 143, 661–664 e665 (2023).
Tsou, P. S., Shi, B. & Varga, J. Role of cellular senescence in the pathogenesis of systemic sclerosis. Curr. Opin. Rheumatol. 34, 343–350 (2022).
Adler, B. L. et al. Autoantibodies targeting telomere-associated proteins in systemic sclerosis. Ann. Rheum. Dis. 80, 912–919 (2021).
Jia, M. et al. Transcriptional changes of the aging lung. Aging Cell 22, e13969 (2023).
Mebratu, Y. A. et al. The aged extracellular matrix and the profibrotic role of senescence-associated secretory phenotype. Am. J. Physiol. Cell Physiol. 325, C565–C579 (2023).
O’Reilly, S., Tsou, P. S. & Varga, J. Senescence and tissue fibrosis: opportunities for therapeutic targeting. Trends Mol. Med. 30, 1113–1125 (2024).
Xing, E., Billi, A. C. & Gudjonsson, J. E. Sex bias and autoimmune diseases. J. Invest. Dermatol. 142, 857–866 (2022).
Dou, D. R. et al. Xist ribonucleoproteins promote female sex-biased autoimmunity. Cell 187, 733–749.e16 (2024).
Yasuoka, H., Larregina, A. T., Yamaguchi, Y. & Feghali-Bostwick, C. A. Human skin culture as an ex vivo model for assessing the fibrotic effects of insulin-like growth factor binding proteins. Open. Rheumatol. J. 2, 17–22 (2008).
Aida-Yasuoka, K. et al. Estradiol promotes the development of a fibrotic phenotype and is increased in the serum of patients with systemic sclerosis. Arthritis Res. Ther. 15, R10 (2013).
Yamaguchi, Y. et al. A peptide derived from endostatin ameliorates organ fibrosis. Sci. Transl. Med. 4, 136ra171 (2012).
Watanabe, T. et al. A human skin model recapitulates systemic sclerosis dermal fibrosis and identifies COL22A1 as a TGFβ early response gene that mediates fibroblast to myofibroblast transition. Genes 10, 75 (2019).
Sharma, S. et al. E4 engages uPAR and enolase-1 and activates urokinase to exert antifibrotic effects. JCI Insight 6, e144935 (2021).
Mlakar, L. et al. Ameliorating fibrosis in murine and human tissues with END55, an endostatin-derived fusion protein made in plants. Biomedicines 10, 2861 (2022).
Valenzi, E. et al. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann. Rheum. Dis. 78, 1379–1387 (2019).
Tabib, T. et al. Myofibroblast transcriptome indicates SFRP2hi fibroblast progenitors in systemic sclerosis skin. Nat. Commun. 12, 4384 (2021).
Clark, K. E. N. et al. Single-cell analysis reveals key differences between early-stage and late-stage systemic sclerosis skin across autoantibody subgroups. Ann. Rheum. Dis. 82, 1568–1579 (2023).
Ramos, P. S. et al. Integrative analysis of DNA methylation in discordant twins unveils distinct architectures of systemic sclerosis subsets. Clin. Epigenetics 11, 58 (2019).
Malaab, M. et al. Antifibrotic factor KLF4 is repressed by the miR-10/TFAP2A/TBX5 axis in dermal fibroblasts: insights from twins discordant for systemic sclerosis. Ann. Rheum. Dis. 81, 268–277 (2022).
Baker Frost, D., da Silveira, W., Hazard, E. S., Atanelishvili, I., Wilson, R. C., Flume, J., Day, K. L., Oates, J. C., Bogatkevich, G. S., Feghali-Bostwick, C., Hardiman, G. & Ramos, P. S. Differential DNA methylation landscape in skin fibroblasts from African Americans with systemic sclerosis. Genes 12, 2 (2021).
Mouawad, J. E. & Feghali-Bostwick, C. The molecular mechanisms of systemic sclerosis-associated lung fibrosis. Int. J. Mol. Sci. 24, 2963 (2023).
Mouawad, J. E. et al. Reduced Cathepsin L expression and secretion into the extracellular milieu contribute to lung fibrosis in systemic sclerosis. Rheumatology 62, 1306–1316 (2023).
Burbelo, P. D. et al. Autoantibodies are present before the clinical diagnosis of systemic sclerosis. PLoS ONE 14, e0214202 (2019).
Bellocchi, C., Chung, A. G. S. E. & Volkmann, E. R. Predicting the progression of very early systemic sclerosis: current insights. Open. Access. Rheumatol. 14, 171–186 (2022).
Lescoat, A. Very early diagnosis of systemic sclerosis: deciphering the heterogeneity of systemic sclerosis in the very early stages of the disease. J. Scleroderma Relat. Disord. 8, 3–6 (2023).
Wuttge, D. M. et al. Increased serum type I interferon activity in early systemic sclerosis patients is associated with antibodies against Sjögren’s syndrome antigens and nuclear ribonucleoprotein antigens. Scand. J. Rheumatol. 42, 235–240 (2013).
Andraos, R. et al. Autoantibodies associated with systemic sclerosis in three autoimmune diseases imprinted by type I interferon gene dysregulation: a comparison across SLE, primary Sjögren’s syndrome and systemic sclerosis. Lupus Sci. Med. 9, e000732 (2022).
Brkic, Z. et al. The interferon type I signature is present in systemic sclerosis before overt fibrosis and might contribute to its pathogenesis through high BAFF gene expression and high collagen synthesis. Ann. Rheum. Dis. 75, 1567–1573 (2016).
Feghali-Bostwick, C., Medsger, T. A. Jr & Wright, T. M. Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum. 48, 1956–1963 (2003).
Arora-Singh, R. K. et al. Autoimmune diseases and autoantibodies in the first degree relatives of patients with systemic sclerosis. J. Autoimmun. 35, 52–57 (2010).
Whitfield, M. L. et al. Systemic and cell type-specific gene expression patterns in scleroderma skin. Proc. Natl Acad. Sci. USA 100, 12319–12324 (2003).
Yang, M. et al. Clinical phenotypes of patients with systemic sclerosis with distinct molecular signatures in skin. Arthritis Care Res. 75, 1469–1480 (2023).
Franks, J. M. et al. A genomic meta-analysis of clinical variables and their association with intrinsic molecular subsets in systemic sclerosis. Rheumatology 62, 19–28 (2022).
Skaug, B. et al. Large-scale analysis of longitudinal skin gene expression in systemic sclerosis reveals relationships of immune cell and fibroblast activity with skin thickness and a trend towards normalisation over time. Ann. Rheum. Dis. 81, 516–523 (2022).
Skaug, B. et al. Global skin gene expression analysis of early diffuse cutaneous systemic sclerosis shows a prominent innate and adaptive inflammatory profile. Ann. Rheum. Dis. 79, 379–386 (2020).
Mehta, B. K. et al. Machine-learning classification identifies patients with early systemic sclerosis as abatacept responders via CD28 pathway modulation. JCI Insight 7, e155282 (2022).
Franks, J. M. et al. A machine learning classifier for assigning individual patients with systemic sclerosis to intrinsic molecular subsets. Arthritis Rheumatol. 71, 1701–1710 (2019).
Franks, J. M. et al. Machine learning predicts stem cell transplant response in severe scleroderma. Ann. Rheum. Dis. 79, 1608–1615 (2020).
Lofgren, S. et al. Integrated, multicohort analysis of systemic sclerosis identifies robust transcriptional signature of disease severity. JCI Insight 1, e89073 (2016).
Correia, C. et al. High-throughput quantitative histology in systemic sclerosis skin disease using computer vision. Arthritis Res. Ther. 22, 48 (2020).
Mahoney, J. M. et al. Systems level analysis of systemic sclerosis shows a network of immune and profibrotic pathways connected with genetic polymorphisms. PLoS Comput. Biol. 11, e1004005 (2015).
Taroni, J. N. et al. A novel multi-network approach reveals tissue-specific cellular modulators of fibrosis in systemic sclerosis. Genome Med. 9, 27 (2017).
Bhandari, R. et al. Profibrotic activation of human macrophages in systemic sclerosis. Arthritis Rheumatol. 72, 1160–1169 (2020).
Ma, F. et al. Systems-based identification of the Hippo pathway for promoting fibrotic mesenchymal differentiation in systemic sclerosis. Nat. Commun. 15, 210 (2024).
Soderberg, O. et al. Direct observation of individual endogenous protein complexes in situ by proximity ligation. Nat. Methods 3, 995–1000 (2006).
Chen, Y. & Guo, J. Multiplexed single-cell in situ protein profiling. ACS Meas. Sci. Au 2, 296–303 (2022).
Herrick, A. L. et al. Patterns and predictors of skin score change in early diffuse systemic sclerosis from the European Scleroderma Observational Study. Ann. Rheum. Dis. 77, 563–570 (2018).
Candia, J., Daya, G. N., Tanaka, T., Ferrucci, L. & Walker, K. A. Assessment of variability in the plasma 7k SomaScan proteomics assay. Sci. Rep. 12, 17147 (2022).
Wik, L. et al. Proximity extension assay in combination with next-generation sequencing for high-throughput proteome-wide analysis. Mol. Cell Proteom. 20, 100168 (2021).
Bjorkesten, J. et al. Stability of proteins in dried blood spot biobanks. Mol. Cell Proteom. 16, 1286–1296 (2017).
Bellocchi, C. et al. Proteomic aptamer analysis reveals serum markers that characterize preclinical systemic sclerosis (SSc) patients at risk for progression toward definite SSc. Arthritis Res. Ther. 25, 15 (2023).
Alvez, M. B. et al. Next generation pan-cancer blood proteome profiling using proximity extension assay. Nat. Commun. 14, 4308 (2023).
Ravaglia, C. & Poletti, V. Transbronchial lung cryobiopsy for the diagnosis of interstitial lung diseases. Curr. Opin. Pulm. Med. 28, 9–16 (2022).
Denton, C. P. et al. The 2024 British Society for Rheumatology guideline for management of systemic sclerosis. Rheumatology 63, 2956–2975 (2024).
Muller, F. et al. CD19 CAR T-cell therapy in autoimmune disease — a case series with follow-up. N. Engl. J. Med. 390, 687–700 (2024).
Merkt, W. et al. Third-generation CD19.CAR-T cell-containing combination therapy in Scl70+ systemic sclerosis. Ann. Rheum. Dis. 83, 543–546 (2024).
Bergmann, C. et al. Treatment of a patient with severe systemic sclerosis (SSc) using CD19-targeted CAR T cells. Ann. Rheum. Dis. 82, 1117–1120 (2023).
Zamanian, R. T. et al. Safety and efficacy of B-cell depletion with rituximab for the treatment of systemic sclerosis-associated pulmonary arterial hypertension: a multicenter, double-blind, randomized, placebo-controlled trial. Am. J. Respir. Crit. Care Med. 204, 209–221 (2021).
Nash, R. A. 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 110, 1388–1396 (2007).
Aschwanden, M. et al. Rapid improvement of nailfold capillaroscopy after intense immunosuppression for systemic sclerosis and mixed connective tissue disease. Ann. Rheum. Dis. 67, 1057–1059 (2008).
van Laar, J. M. et al. Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. JAMA 311, 2490–2498, (2014).
Sullivan, K. M. et al. Myeloablative autologous stem-cell transplantation for severe scleroderma. N. Engl. J. Med. 378, 35–47 (2018).
Gustafsson, K. et al. Clearing and replacing tissue-resident myeloid cells with an anti-CD45 antibody-drug conjugate. Blood Adv. 7, 6964–6973 (2023).
Xue, E. et al. Cellular-based therapies in systemic sclerosis: from hematopoietic stem cell transplant to innovative approaches. Cells 11, 3346 (2022).
Yang, H., Cheong, S., He, Y. & Lu, F. Mesenchymal stem cell-based therapy for autoimmune-related fibrotic skin diseases — systemic sclerosis and sclerodermatous graft-versus-host disease. Stem Cell Res. Ther. 14, 372 (2023).
Barde, F. et al. Induction of regulatory T cells and efficacy of low-dose interleukin-2 in systemic sclerosis: interventional open-label phase 1–phase 2a study. RMD Open 10, e003500 (2024).
Le Huu, D. et al. Donor-derived regulatory B cells are important for suppression of murine sclerodermatous chronic graft-versus-host disease. Blood 121, 3274–3283 (2013).
Liu, M. Effector and regulatory B-cell imbalance in systemic sclerosis: cooperation or competition? Clin. Rheumatol. 43, 2783–2789 (2024).
Morante-Palacios, O., Fondelli, F., Ballestar, E. & Martinez-Caceres, E. M. Tolerogenic dendritic cells in autoimmunity and inflammatory diseases. Trends Immunol. 42, 59–75 (2021).
Odell, I. D. et al. Epiregulin is a dendritic cell-derived EGFR ligand that maintains skin and lung fibrosis. Sci. Immunol. 7, eabq6691 (2022).
Dorst, D. N. et al. Fibroblast activation protein targeted photodynamic therapy selectively kills activated skin fibroblasts from systemic sclerosis patients and prevents tissue contraction. Int. J. Mol. Sci. 22, 12681 (2021).
Gur, C. et al. LGR5 expressing skin fibroblasts define a major cellular hub perturbed in scleroderma. Cell 185, 1373–1388 (2022).
Trinh-Minh, T. et al. Effect of anti-S100A4 monoclonal antibody treatment on experimental skin fibrosis and systemic sclerosis-specific transcriptional signatures in human skin. Arthritis Rheumatol. 76, 783–795 (2024).
Liang, M. et al. Attenuation of fibroblast activation and fibrosis by adropin in systemic sclerosis. Sci. Transl. Med. 16, eadd6570 (2024).
Huang, M. et al. Self-assembled human skin equivalents model macrophage activation of cutaneous fibrogenesis in systemic sclerosis. Arthritis Rheumatol. 74, 1245–1256 (2022).
Huang, M. et al. Systemic sclerosis dermal fibroblasts induce cutaneous fibrosis through lysyl oxidase-like 4: new evidence from three-dimensional skin-like tissues. Arthritis Rheumatol. 72, 791–801 (2020).
Denton, C. P. Challenges in systemic sclerosis trial design. Semin. Arthritis Rheum. 49, S3–S7 (2019).
Hoffmann-Vold, A. M. et al. Cohort enrichment strategies for progressive interstitial lung disease in systemic sclerosis from European scleroderma trials and research. Chest 163, 586–598 (2023).
Maurer, B. et al. Prediction of worsening of skin fibrosis in patients with diffuse cutaneous systemic sclerosis using the EUSTAR database. Ann. Rheum. Dis. 74, 1124–1131 (2015).
Khanna, D. et al. Riociguat in patients with early diffuse cutaneous systemic sclerosis (RISE-SSc): randomised, double-blind, placebo-controlled multicentre trial. Ann. Rheum. Dis. 79, 618–625 (2020).
Chung, L. et al. Safety and efficacy of abatacept in early diffuse cutaneous systemic sclerosis (ASSET): open-label extension of a phase 2, double-blind randomised trial. Lancet Rheumatol. 2, e743–e753 (2020).
Khanna, D. 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. 72, 125–136 (2020).
Khanna, D. et al. The American College of Rheumatology provisional composite response index for clinical trials in early diffuse cutaneous systemic sclerosis. Arthritis Care Res. 68, 167–178 (2016).
Khanna, D., Huang, S., Lin, C. J. F. & Spino, C. New composite endpoint in early diffuse cutaneous systemic sclerosis: revisiting the provisional American College of Rheumatology Composite Response Index in Systemic Sclerosis. Ann. Rheum. Dis. 80, 641–650 (2021).
Scleroderma Research Foundation. About CONQUEST. https://srfcure.org/research/conquest/about-conquest/ (2024).
Morgan, N. D. et al. Clinical and serological features of systemic sclerosis in a multicenter African American cohort: analysis of the genome research in African American scleroderma patients clinical database. Medicine 96, e8980 (2017).
Spiera, R. et al. Safety and efficacy of lenabasum in a phase II, randomized, placebo-controlled trial in adults with systemic sclerosis. Arthritis Rheumatol. 72, 1350–1360 (2020).
Spiera, R. et al. Efficacy and safety of lenabasum, a cannabinoid type 2 receptor agonist, in a phase 3 randomized trial in diffuse cutaneous systemic sclerosis. Arthritis Rheumatol. 75, 1608–1618 (2023).
Allanore, Y. et al. A randomised, double-blind, placebo-controlled, 24-week, phase II, proof-of-concept study of romilkimab (SAR156597) in early diffuse cutaneous systemic sclerosis. Ann. Rheum. Dis. 79, 1600–1607 (2020).
Khanna, D. et al. A 24-week, phase IIa, randomized, double-blind, placebo-controlled study of ziritaxestat in early diffuse cutaneous systemic sclerosis. Arthritis Rheumatol. 75, 1434–1444 (2023).
Ebata, S. et al. Safety and efficacy of rituximab in systemic sclerosis (DESIRES): open-label extension of a double-blind, investigators-initiated, randomised, placebo-controlled trial. Lancet Rheumatol. 4, e546–e555 (2022).
Denton, C. P., Stevens, W., Kruger, N., Papadimitriou, M., Khong, F., Bradney, M., Kelly, D. & Lafyatis, R. FT011 for the treatment of systemic sclerosis. results from a phase II study. Arthritis Rheumatol. 75, 2593 (2023).
Acknowledgements
The authors thank K. Brennan for administrative support and meeting summary. Workshop support provided by The Edith Busch Foundation and The Fondazione di Medicina Molecolare e Terapia Cellulare, Università Politecnica delle Marche-Ancona, Italy.
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding authors
Ethics declarations
Competing interests
J.H.W.D. has consultancy relationships with AbbVie, Active Biotech, Anamar, ARXX, AstraZeneca, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, Genentech, GSK, Inventiva, Janssen, Novartis, Pfizer, Roche and UCB, and has received research funding from AbbVie, Anamar, Argenx, ARXX, BMS, Bayer Pharma, Boehringer Ingelheim, Cantargia, Celgene, CSL Behring, ExoTherapeutics, Galapagos, GSK, Inventiva, Kiniksa, Lassen, Novartis, Sanofi-Aventis, RedX, UCB and Zenasbio. J.H.W.D. is CEO of 4D Science and Scientific Lead of FibroCure. R.D. has received funding from and is a consultant for Aisa Pharma Inc and AstraZeneca. M.K. has received funding from and is a consultant for Boehringer Ingelheim, Mochida, Kissei, GSK, AstraZeneca, Mitsubishi Tanabe, Janssen, Biogen, Novartis, Chugai and Asahi Kasei Pharma. U.L. is the founder of Olink Proteomics and has stocks in Navinci and SampleFacts.
Peer review
Peer review information
Nature Reviews Rheumatology thanks Lorinda Chung, who co-reviewed with Brian Lee, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Abraham, D.J., Black, C.M., Denton, C.P. et al. An international perspective on the future of systemic sclerosis research. Nat Rev Rheumatol 21, 174–187 (2025). https://doi.org/10.1038/s41584-024-01217-2
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41584-024-01217-2
This article is cited by
-
The senescence-like activity of BMS-470539 is associated with anti-fibrotic actions in models of dermal fibrosis
Arthritis Research & Therapy (2025)
-
Medikamentöse Fibrosehemmung am Beispiel der systemischen Sklerose
Die Innere Medizin (2025)