Tofacitinib blocks IFN-regulated biomarker genes in skin fibroblasts and keratinocytes in a systemic sclerosis trial

Dinesh Khanna, Cristina Padilla, Lam C Tsoi, Vivek Nagaraja, Puja P Khanna, Tracy Tabib, J Michelle Kahlenberg, Amber Young, Suiyuan Huang, Johann E Gudjonsson, David A Fox, Robert Lafyatis, Dinesh Khanna, Cristina Padilla, Lam C Tsoi, Vivek Nagaraja, Puja P Khanna, Tracy Tabib, J Michelle Kahlenberg, Amber Young, Suiyuan Huang, Johann E Gudjonsson, David A Fox, Robert Lafyatis

Abstract

BACKGROUNDSystemic sclerosis (SSc) is an autoimmune, connective tissue disease characterized by vasculopathy and fibrosis of the skin and internal organs.METHODSWe randomized 15 participants with early diffuse cutaneous SSc to tofacitinib 5 mg twice a day or matching placebo in a phase I/II double-blind, placebo-controlled trial. The primary outcome measure was safety and tolerability at or before week 24. To understand the changes in gene expression associated with tofacitinib treatment in each skin cell population, we compared single-cell gene expression in punch skin biopsies obtained at baseline and 6 weeks following the initiation of treatment.RESULTSTofacitinib was well tolerated; no participants experienced grade 3 or higher adverse events before or at week 24. Trends in efficacy outcome measures favored tofacitnib. Baseline gene expression in fibroblast and keratinocyte subpopulations indicated IFN-activated gene expression. Tofacitinib inhibited IFN-regulated gene expression in SFRP2/DPP4 fibroblasts (progenitors of myofibroblasts) and in MYOC/CCL19, representing adventitial fibroblasts (P < 0.05), as well as in the basal and keratinized layers of the epidermis. Gene expression in macrophages and DCs indicated inhibition of STAT3 by tofacitinib (P < 0.05). No clinically meaningful inhibition of T cells and endothelial cells in the skin tissue was observed.CONCLUSIONThese results indicate that mesenchymal and epithelial cells of a target organ in SSc, not the infiltrating lymphocytes, may be the primary focus for therapeutic effects of a Janus kinase inhibitor.TRIAL REGISTRATIONClinicalTrials.gov NCT03274076.FUNDINGPfizer, NIH/National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) R01 AR070470, NIH/NIAMS K24 AR063120, Taubman Medical Research Institute and NIH P30 AR075043, and NIH/NIAMS K01 AR072129.

Keywords: Clinical Trials; Rheumatology; Skin.

Figures

Figure 1. Transcriptomes and proportions of cell…
Figure 1. Transcriptomes and proportions of cell populations of study participants.
Combined analysis of scRNA-Seq transcriptomes of skin from 15 participants with dcSSc at baseline and 6 weeks after treatment with tofacitinib (n = 10) or placebo (n = 5; A). Cell clusters (n = 49) are numbered with cell types based on known marker genes indicated to the right. The proportion of cells in each cluster by subgroups of participants: placebo-treated baseline (PBO-Bsl) and 6-week (PBO-W6) and tofacitinib-treated baseline (TOFA-Bsl) and 6-week (TOFA-W6) biopsies (B). Stars indicate pericyte and endothelial cell clusters showing increased proportions of cells after tofacitinib (P < 0.05 by paired 2-tailed t test corrected for multiple comparisons by Bonferroni’s method).
Figure 2. IPA of scRNA-Seq from fibroblast…
Figure 2. IPA of scRNA-Seq from fibroblast populations.
Pathway analysis of fibroblast scRNA-Seq data from baseline study biopsies (n = 15), analyzed together with scRNA-Seq data from previously described dcSSc (n = 12) and healthy skin (n = 10). Selected pathways from clusters 1 and 9, clustered with analogous cells in previous studies, representing SFRP2/DPP4 fibroblasts (A) and clusters 6 and 7 clustered with analogous cells in previous studies representing MYOC/CCL19 fibroblasts (B). Genes correlating with baseline mRSS (uncorrected P < 0.05) were included in the pathway analysis. Only selected significant pathways (–log P < 1.4) are indicated. SFRP2/DPP4. Yellow bars indicate positive associations with IPA-expected direction of regulation; blue bars show negative associations with expected direction of regulation; and gray bars indicate no expected direction of regulation.
Figure 3. Genes and pathways changing in…
Figure 3. Genes and pathways changing in tofacitinib-treated patient SFRP2/DPP4 fibroblasts.
Pathway analysis of scRNA-Seq data from tofacitinib-treated baseline compared with week 6 gene expression (n = 10) by SFRP2 fibroblasts (clusters 1 and 9; A). Average gene expression in these clusters (pseudo-bulk gene expression) showing decreased expression at week 6 compared with baseline were included in the IPA (uncorrected P < 0.05). Only selected significant pathways (–log P < 1.4) are indicated. IPA used right-sided Fisher’s exact test to calculate the significance scores (shown on the y axis). Blue bars indicate pathways downregulated (z score less than –2), orange bar upregulated (z score greater than 2), white bars without direction of regulation, and gray bars with no expected direction of regulation after tofacitinib compared to baseline. The intensity of the shading indicates the level of the z score. Heatmap of gene expression of the genes associated with the IFN pathway seen in A (B). Changes in inflammatory gene signatures at week 6 compared with baseline in the placebo and tofacitinib groups for SFRP2/DPP4 fibroblasts (C). Clustering of changes in pseudo-bulk gene expression in SFRP2/DPP4 fibroblasts at week 6 compared with baseline in tofacitinib-treated participants of all (filtered) genes (D), showing IFN-regulated genes clustering with STAT1 (indicated by a red star).
Figure 4. Genes and pathways changing in…
Figure 4. Genes and pathways changing in tofacitinib-treated patient CCL19/MYOC fibroblasts.
Pathway analysis of scRNA-Seq data from tofacitinib-treated patients at week 6 compared with baseline gene expression (n = 10) by CCL19/MYOC fibroblasts (clusters 6 and 7; A). Average gene expression in these clusters (pseudo-bulk gene expression) showing decreased expression at week 6 compared with baseline were included in the IPA (uncorrected P < 0.05). Only selected significant pathways (–log P < 1.4) are indicated. IPA used right-sided Fisher’s exact test to calculate the significance scores (shown on the y axis). Blue bars indicate pathways downregulated (z score less than –2), orange bar upregulated (z score greater than 2), white bars without direction of regulation, and gray bars with no expected direction of regulation after tofacitinib compared to baseline. The intensity of the shading indicates the level of the z score. Heatmap of gene expression of the genes associated with the IFN pathway seen in A (B). Changes in inflammatory gene signatures at week 6 in the placebo and tofacitinib groups for CCL19/MYOC fibroblasts (C). Clustering of changes in pseudo-bulk gene expression in CCL19/MYOC fibroblasts at 6 weeks compared with baseline in tofacitinib-treated participants of all (filtered) genes (D), showing IFN-regulated genes clustering with JAK2 and STAT1 (indicated by red stars).
Figure 5. Reduction of type I and…
Figure 5. Reduction of type I and type I IFN and other inflammatory signatures by tofacitinib in epidermal keratinocytes.
Changes in inflammatory signature by week 6 in the placebo and tofacitinib groups for basal (KRT14), differentiated (KRT10), and keratinized (FLG) epidermal keratinocytes (AC). Fold change was computed using median value in baseline versus week 6 groups with P value calculated using Wilcoxon rank sum test. (D) Dot plot showing the most significantly enriched functions among genes only downregulated in the tofacitinib (but not placebo) group. Only significant results (i.e., FDR ≤ 1%) are shown.
Figure 6. Downregulated expression of IFN-regulated genes…
Figure 6. Downregulated expression of IFN-regulated genes after tofacitinib in fibroblast populations.
Dot plots showing markers for the SFRP2 (cluster 1 and 9), MYOC (cluster 6), CCL19 (cluster 7), as well as CRABP1 (dermal papilla, cluster 36) and ANGPTL7 (cluster 41) fibroblasts. IFN-regulated genes (IFI35, IFITM1, IFITM3, OAS1, and MX1) decreased after tofacitinib in clusters 1/9 (indicated by aqua bar) and in clusters 6/7 (IFNAR2, ISG15, IFI44L, and OAS3), indicated by maroon bar), but not in myofibroblasts (A). The lack of effect of tofacitinib treatment on expression of genes associated with myofibroblast differentiation in individual participants (B).

References

    1. Denton CP, Khanna D. Systemic sclerosis. Lancet. 2017;390(10103):1685–1699. doi: 10.1016/S0140-6736(17)30933-9.
    1. Roumm AD, et al. Lymphocytes in the skin of patients with progressive systemic sclerosis. Quantification, subtyping, and clinical correlations. Arthritis Rheum. 1984;27(6):645–653. doi: 10.1002/art.1780270607.
    1. Maehara T, et al. Cytotoxic CD4+ T lymphocytes may induce endothelial cell apoptosis in systemic sclerosis. J Clin Invest. 2020;130(5):2451–2464. doi: 10.1172/JCI131700.
    1. Kalogerou A, et al. Early T cell activation in the skin from patients with systemic sclerosis. Ann Rheum Dis. 2005;64(8):1233–1235. doi: 10.1136/ard.2004.027094.
    1. Stifano G, Christmann RB. Macrophage involvement in systemic sclerosis: do we need more evidence? Curr Rheumatol Rep. 2016;18(1):2. doi: 10.1007/s11926-015-0554-8.
    1. Schwartz DM, et al. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol. 2016;12(1):25–36. doi: 10.1038/nrrheum.2015.167.
    1. Hodge JA, et al. The mechanism of action of tofacitinib — an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 2016;34(2):318–328.
    1. Meyer DM, et al. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690,550, in rat adjuvant-induced arthritis. J Inflamm (Lond) 2010;7:41. doi: 10.1186/1476-9255-7-41.
    1. Emori T, et al. Role of JAK-STAT signaling in the pathogenic behavior of fibroblast-like synoviocytes in rheumatoid arthritis: effect of the novel JAK inhibitor peficitinib. Eur J Pharmacol. 2020;882:173238. doi: 10.1016/j.ejphar.2020.173238.
    1. Migita K, et al. Inhibition of Janus kinase/signal transducer and activator of transcription (JAK/STAT) signalling pathway in rheumatoid synovial fibroblasts using small molecule compounds. Clin Exp Immunol. 2013;174(3):356–363. doi: 10.1111/cei.12190.
    1. Wang W, et al. The JAK/STAT pathway is activated in systemic sclerosis and is effectively targeted by tofacitinib. J Scleroderma Relat Disord. 2020;5(1):40–50. doi: 10.1177/2397198319865367.
    1. Aung WW, et al. Immunomodulating role of the JAKs inhibitor tofacitinib in a mouse model of bleomycin-induced scleroderma. J Dermatol Sci. 2020;101(3):174–184.
    1. Lescoat A, 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. doi: 10.1016/j.bcp.2020.114103.
    1. Tabib T, et al. Myofibroblast transcriptome indicates SFRP2 hi fibroblast progenitors in systemic sclerosis skin. Nat Commun. 2021;12(1):4384. doi: 10.1038/s41467-021-24607-6.
    1. Farina G, et al. A four-gene biomarker predicts skin disease in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheum. 2010;62(2):580–588. doi: 10.1002/art.27220.
    1. Rice LM, et al. A longitudinal biomarker for the extent of skin disease in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheumatol. 2015;67(11):3004–3015. doi: 10.1002/art.39287.
    1. Lehtonen A, et al. Interferons up-regulate STAT1, STAT2, and IRF family transcription factor gene expression in human peripheral blood mononuclear cells and macrophages. J Immunol. 1997;159(2):794–803.
    1. Barley RD, et al. Aberrant p21WAF1-dependent growth arrest as the possible mechanism of abnormal resistance to ultraviolet light cytotoxicity in Li-Fraumeni syndrome fibroblast strains heterozygous for TP53 mutations. Oncogene. 1998;17(5):533–543. doi: 10.1038/sj.onc.1202271.
    1. Ashburner M, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000;25(1):25–29. doi: 10.1038/75556.
    1. The Gene Ontology Consortium. The Gene Ontology resource: 20 years and still GOing strong. Nucleic Acids Res. 2019;47(d1):D330–D338. doi: 10.1093/nar/gky1055.
    1. Gur C, et al. LGR5 expressing skin fibroblasts define a major cellular hub perturbed in scleroderma. Cell. 2022;185(8):1373–1388. doi: 10.1016/j.cell.2022.03.011.
    1. Makino K, et al. Blockade of PDGF receptors by crenolanib has therapeutic effect in patient fibroblasts and in preclinical models of systemicsclerosis. J Invest Dermatol. 2017;137(8):1671–1681. doi: 10.1016/j.jid.2017.03.032.
    1. Arron JR. Biomarkers in systemic sclerosis: mechanistic insights into pathogenesis and treatment. Curr Opin Rheumatol. 2021;33(6):480–485. doi: 10.1097/BOR.0000000000000827.
    1. Nazari B, et al. Altered dermal fibroblasts in systemic sclerosis display podoplanin and CD90. Am J Pathol. 2016;186(10):2650–2664. doi: 10.1016/j.ajpath.2016.06.020.
    1. Li Y, et al. Inducible interleukin 32 (IL-32) exerts extensive antiviral function via selective stimulation of interferon λ1 (IFN-λ1) J Biol Chem. 2013;288(29):20927–20941. doi: 10.1074/jbc.M112.440115.
    1. Netea MG, et al. Mycobacterium tuberculosis induces interleukin-32 production through a caspase- 1/IL-18/interferon-gamma-dependent mechanism. PLoS Med. 2006;3(8):e277. doi: 10.1371/journal.pmed.0030277.
    1. Jha AK, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419–430. doi: 10.1016/j.immuni.2015.02.005.
    1. Vangala JR, et al. Regulation of PSMB5 protein and β subunits of mammalian proteasome by constitutively activated signal transducer and activator of transcription 3 (STAT3): potential role in bortezomib-mediated anticancer therapy. J Biol Chem. 2014;289(18):12612–12622. doi: 10.1074/jbc.M113.542829.
    1. Saito Y, et al. AT-rich-interactive domain-containing protein 5A functions as a negative regulator of retinoic acid receptor-related orphan nuclear receptor γt-induced Th17 cell differentiation. Arthritis Rheumatol. 2014;66(5):1185–1194. doi: 10.1002/art.38324.
    1. Wolfle SJ, et al. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur J Immunol. 2011;41(2):413–424. doi: 10.1002/eji.201040979.
    1. Parackova Z, et al. Enhanced STAT3 phosphorylation and PD-L1 expression in myeloid dendritic cells indicate impaired IL-27Ralpha signaling in type 1 diabetes. Sci Rep. 2020;10(1):493. doi: 10.1038/s41598-020-57507-8.
    1. Lee SH, et al. HtrA2 suppresses autoimmune arthritis and regulates activation of STAT3. Sci Rep. 2016;6:39393. doi: 10.1038/srep39393.
    1. Goldberg L, et al. Alternative splicing of STAT3 is affected by RNA editing. DNA Cell Biol. 2017;36(5):367–376. doi: 10.1089/dna.2016.3575.
    1. Zhao Y, et al. Exhaustion and senescence: two crucial dysfunctional states of T cells in the tumor microenvironment. Cell Mol Immunol. 2020;17(1):27–35. doi: 10.1038/s41423-019-0344-8.
    1. Buggert M, et al. T-bet and Eomes are differentially linked to the exhausted phenotype of CD8+ T cells in HIV infection. PLoS Pathog. 2014;10(7):e1004251. doi: 10.1371/journal.ppat.1004251.
    1. Li J, et al. High levels of eomes promote exhaustion of anti-tumor CD8+ T cells. Front Immunol. 2018;9:2981. doi: 10.3389/fimmu.2018.02981.
    1. Stephen TL, et al. Transforming growth factor β-mediated suppression of antitumor T cells requires FoxP1 transcription factor expression. Immunity. 2014;41(3):427–439. doi: 10.1016/j.immuni.2014.08.012.
    1. Andrews LP, et al. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev. 2017;276(1):80–96. doi: 10.1111/imr.12519.
    1. Schoors S, et al. Fatty acid carbon is essential for dNTP synthesis in endothelial cells. Nature. 2015;520(7546):192–197. doi: 10.1038/nature14362.
    1. Nagaraja V, et al. Current and future outlook on disease modification and defining low disease activity in systemic sclerosis. Arthritis Rheumatol. 2020;72(7):1049–1058. doi: 10.1002/art.41246.
    1. Dekanty A, et al. Leukemia inhibitory factor induces DNA synthesis in Swiss mouse 3T3 cells independently of cyclin D1 expression through a mechanism involving MEK/ERK1/2 activation. J Biol Chem. 2006;281(10):6136–6143. doi: 10.1074/jbc.M505839200.
    1. Rosengren S, et al. The JAK inhibitor CP-690,550 (tofacitinib) inhibits TNF-induced chemokine expression in fibroblast-like synoviocytes: autocrine role of type I interferon. Ann Rheum Dis. 2012;71(3):440–447. doi: 10.1136/ard.2011.150284.
    1. LaBranche TP, et al. JAK inhibition with tofacitinib suppresses arthritic joint structural damage through decreased RANKL production. Arthritis Rheum. 2012;64(11):3531–3542. doi: 10.1002/art.34649.
    1. Dees C, et al. JAK-2 as a novel mediator of the profibrotic effects of transforming growth factor β in systemic sclerosis. Arthritis Rheum. 2012;64(9):3006–3015. doi: 10.1002/art.34500.
    1. Mizoguchi F, et al. Functionally distinct disease-associated fibroblast subsets in rheumatoid arthritis. Nat Commun. 2018;9(1):789. doi: 10.1038/s41467-018-02892-y.
    1. Valenzi E, et al. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann Rheum Dis. 2019;78(10):1379–1387. doi: 10.1136/annrheumdis-2018-214865.
    1. York MR, et al. A macrophage marker, Siglec-1, is increased on circulating monocytes in patients with systemic sclerosis and induced by type I interferons and toll-like receptor agonists. Arthritis Rheum. 2007;56(3):1010–1020. doi: 10.1002/art.22382.
    1. Farina GA, et al. Poly(I:C) drives type I IFN- and TGFβ-mediated inflammation and dermal fibrosis simulating altered gene expression in systemic sclerosis. J Invest Dermatol. 2010;130(11):2583–2593. doi: 10.1038/jid.2010.200.
    1. Barrat FJ, Lu TT. Role of type I interferons and innate immunity in systemic sclerosis: unbalanced activities on distinct cell types? Curr Opin Rheumatol. 2019;31(6):569–575. doi: 10.1097/BOR.0000000000000659.
    1. McCoy SS, et al. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta. Rheumatology (Oxford) 2017;56(11):1970–1981. doi: 10.1093/rheumatology/kex280.
    1. Wu M, Assassi S. The role of type 1 interferon in systemic sclerosis. Front Immunol. 2013;4:266.
    1. Basters A, et al. USP18 — a multifunctional component in the interferon response. Biosci Rep. 2018;38(6):BSR20180250. doi: 10.1042/BSR20180250.
    1. Schafer MJ, et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun. 2017;8:14532. doi: 10.1038/ncomms14532.
    1. Bueno M, et al. PINK1 deficiency impairs mitochondrial homeostasis and promotes lung fibrosis. J Clin Invest. 2015;125(2):521–538. doi: 10.1172/JCI74942.
    1. Summer R, et al. Activation of the mTORC1/PGC-1 axis promotes mitochondrial biogenesis and induces cellular senescence in the lung epithelium. Am J Physiol Lung Cell Mol Physiol. 2019;316(6):L1049–L1060. doi: 10.1152/ajplung.00244.2018.
    1. Birch J, Gil J. Senescence and the SASP: many therapeutic avenues. Genes Dev. 2020;34(23–24):1565–1576.
    1. Lin Y, Xu Z. Fibroblast senescence in idiopathic pulmonary fibrosis. Front Cell Dev Biol. 2020;8:593283. doi: 10.3389/fcell.2020.593283.
    1. Schafer MJ, et al. Targeting senescent cells in fibrosis: pathology, paradox, and practical considerations. Curr Rheumatol Rep. 2018;20(1):3. doi: 10.1007/s11926-018-0712-x.
    1. Piera-Velazquez S, Jimenez SA. Role of cellular senescence and NOX4-mediated oxidative stress in systemic sclerosis pathogenesis. Curr Rheumatol Rep. 2015;17(1):473. doi: 10.1007/s11926-014-0473-0.
    1. Luckhardt TR, Thannickal VJ. Systemic sclerosis-associated fibrosis: an accelerated aging phenotype? Curr Opin Rheumatol. 2015;27(6):571–576. doi: 10.1097/BOR.0000000000000219.
    1. Bernard K, et al. Metabolic reprogramming is required for myofibroblast contractility and differentiation. J Biol Chem. 2015;290(42):25427–25438. doi: 10.1074/jbc.M115.646984.
    1. Xie N, et al. Glycolytic reprogramming in myofibroblast differentiation and lung fibrosis. Am J Respir Crit Care Med. 2015;192(12):1462–1474. doi: 10.1164/rccm.201504-0780OC.
    1. Xie N, et al. Metabolic characterization and RNA profiling reveal glycolytic dependence of profibrotic phenotype of alveolar macrophages in lung fibrosis. Am J Physiol Lung Cell Mol Physiol. 2017;313(5):L834–L844. doi: 10.1152/ajplung.00235.2017.
    1. Van den Hoogen F, et al. 2013 classification criteria for systemic sclerosis: an American College of Rheumatology/European League against rheumatism collaborative initiative. Arthritis Rheum. 2013;65(11):2737–2747. doi: 10.1002/art.38098.
    1. LeRoy EC, Medsger TA., Jr Criteria for the classification of early systemic sclerosis. J Rheumatol. 2001;28(7):1573–1576.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888–1902. doi: 10.1016/j.cell.2019.05.031.

Source: PubMed

3
Prenumerera