An open-label study to evaluate biomarkers and safety in systemic sclerosis patients treated with paquinimod

Roger Hesselstrand, Jörg H W Distler, Gabriela Riemekasten, Dirk M Wuttge, Marie Törngren, Helén C Nyhlén, Fredrik Andersson, Helena Eriksson, Birgitta Sparre, Helén Tuvesson, Oliver Distler, Roger Hesselstrand, Jörg H W Distler, Gabriela Riemekasten, Dirk M Wuttge, Marie Törngren, Helén C Nyhlén, Fredrik Andersson, Helena Eriksson, Birgitta Sparre, Helén Tuvesson, Oliver Distler

Abstract

Objectives: To evaluate the changes in disease-related biomarkers and safety of paquinimod, an oral immunomodulatory compound, in patients with systemic sclerosis (SSc).

Methods: In this open-label, single-arm, multicenter study, SSc patients with a rapidly progressive disease received paquinimod for 8 weeks. Blood and skin biopsies were collected at baseline, during treatment, and at follow-up for the analyses of type I interferon (IFN) activity, chemokine (C-C motif) ligand 2 (CCL2), and the number of myofibroblasts. The safety of paquinimod was evaluated throughout the study.

Results: Nine SSc patients were enrolled and completed the study treatment with paquinimod at 3 mg/day for 8 weeks. After the treatment, a reduction of type I IFN activity in the plasma from one patient with elevated baseline IFN activity was recorded. A trend towards reduced IFN activity in the skin after treatment was also observed in patients. The serum level of CCL2 was reduced in 7 of 9 patients after paquinimod treatment. There was a median reduction of 10% of the number of myofibroblasts in skin biopsies at week 8 compared to baseline. No change in modified Rodnan skin score and quality of life was detected in the study. Reported adverse events (AEs) were mild to moderate and expected with the most common being arthralgia (n = 3) and headache (n = 3), and C-reactive protein (CRP) increase.

Conclusions: Analysis of biomarkers before and after treatment suggest reduced type I IFN activity and reduced number of myofibroblasts in lesional skin. Paquinimod was overall well tolerated with mild to moderate and expected AEs.

Trial registration: ClinicalTrials.gov, NCT01487551 . Registered on 7 September 2011.

Keywords: Clinical trial; Paquinimod; Skin fibrosis; Systemic sclerosis.

Conflict of interest statement

RH received funding from Active Biotech AB related to the study. JHWD has consultancy relationships with Actelion, Active Biotech, Anamar, ARXX, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, JB Therapeutics, Medac, Pfizer, RuiYi, and UCB. JHWD has received research funding from Anamar, Active Biotech, Array Biopharma, ARXX, aTyr, BMS, Bayer Pharma, Boehringer Ingelheim, Celgene, Galapagos, GSK, Inventiva, Novartis, Sanofi-Aventis, RedX, and UCB. JHWD is a stock owner of 4D Science.

GR and DMW declare that they have no competing interests.

MT, HT, and HE are employees and stock owners of Active Biotech AB.

HCN is a former employee at Active Biotech AB and owns stocks in Active Biotech AB.

FA and BS are former employees at Active Biotech AB.

OD has/had consultancy relationship and/or has received research funding from Abbvie, Actelion, Acceleron Pharma, Amgen, AnaMar, Baecon Discovery, Blade Therapeutics, Bayer, Boehringer Ingelheim, Catenion, Competitive Drug Development International Ltd., CSL Behring, ChemomAb, Curzion Pharmaceuticals, Ergonex, Galapagos NV, Glenmark Pharmaceuticals, GSK, Inventiva, Italfarmaco, iQone, iQvia, Lilly, medac, Medscape, Mitsubishi Tanabe Pharma, MSD, Novartis, Pfizer, Roche, Sanofi, Target BioScience, and UCB in the area of potential treatments of scleroderma and its complications. He has a patent mir-29 for the treatment of systemic sclerosis issued (US8247389, EP2331143).

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Plasma levels of paquinimod in each individual patient were analyzed at weeks 2, 4 and 8, and at follow-up week 12, by using LC-MS/MS
Fig. 2
Fig. 2
A Type I IFN activity in plasma in each individual patient was analyzed at baseline, weeks 4 and 8, and at follow-up week 12, by using a functional receptor assay. B The gene expression of five type I IFN responsive genes, IFI44, IFI44L, IFI6, IFI27, and RSAD2, in the skin was analyzed by real-time PCR. The forest plot shows mean fold changes (log2) of each gene after 8 weeks of treatment. The dashes indicate mean fold changes (log2) after 8 weeks of treatment with paquinimod, and the horizontal bars show 95% CIs
Fig. 3
Fig. 3
Changes in the levels of CCL2 in the serum during the whole study in each individual patient. The recorded baseline values ranged between 427 and 1309 pg/mL. A reduction was observed in 7 out of 9 patients during treatment; the mean (shown in red) reduction was 18% (p = 0.07)
Fig. 4
Fig. 4
Changes in normalized gene expression of CCR2 in the skin. The mean fold reduction was 0.59 (p = 0.019) (shown in red) after 8 weeks of treatment
Fig. 5
Fig. 5
Changes of the number of myofibroblasts in skin biopsies from each individual patient treated with paquinimod. The myofibroblasts were analyzed by immunohistochemistry and detected by using an antibody recognizing α-smooth muscle actin (αSMA) in skin biopsies taken at baseline and week 8. The mean (shown in red) reduction was 8% (p = 0.023)
Fig. 6
Fig. 6
Modified Rodnan Skin Score (mRSS) at baseline, at weeks 4 and 8, and at follow-up in each individual patient treated with paquinimod

References

    1. Varga J, Trojanowska M, Kuwana M. Pathogenesis of systemic sclerosis: recent insights of molecular and cellular mechanisms and therapeutic opportunities. J Scleroderma Rel Disord. 2017;2(3):137–152. doi: 10.5301/jsrd.5000249.
    1. Hesselstrand R, Scheja A, Akesson A. Mortality and causes of death in a Swedish series of systemic sclerosis patients. Ann Rheum Dis. 1998;57(11):682–686. doi: 10.1136/ard.57.11.682.
    1. Hoffmann-Vold AM, Molberg O, Midtvedt O, Garen T, Gran JT. Survival and causes of death in an unselected and complete cohort of Norwegian patients with systemic sclerosis. J Rheumatol. 2013;40(7):1127–1133. doi: 10.3899/jrheum.121390.
    1. Jacobsen S, Halberg P, Ullman S, Van Venrooij WJ, Hoier-Madsen M, Wiik A, Petersen J. Clinical features and serum antinuclear antibodies in 230 Danish patients with systemic sclerosis. Br J Rheumatol. 1998;37(1):39–45. doi: 10.1093/rheumatology/37.1.39.
    1. Kowal-Bielecka O, Fransen J, Avouac J, Becker M, Kulak A, Allanore Y, Distler O, Clements P, Cutolo M, Czirjak L, Damjanov N, del Galdo F, Denton CP, Distler JHW, Foeldvari I, Figelstone K, Frerix M, Furst DE, Guiducci S, Hunzelmann N, Khanna D, Matucci-Cerinic M, Herrick AL, van den Hoogen F, van Laar J, Riemekasten G, Silver R, Smith V, Sulli A, Tarner I, Tyndall A, Welling J, Wigley F, Valentini G, Walker UA, Zulian F, Müller-Ladner U, EUSTAR Coauthors Update of EULAR recommendations for the treatment of systemic sclerosis. Ann Rheum Dis. 2017;76(8):1327–1339. doi: 10.1136/annrheumdis-2016-209909.
    1. Distler JHW, Gyorfi AH, Ramanujam M, Whitfield ML, Konigshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol. 2019;15(12):705–730. doi: 10.1038/s41584-019-0322-7.
    1. Ishikawa O, Ishikawa H. Macrophage infiltration in the skin of patients with systemic sclerosis. J Rheumatol. 1992;19(8):1202–1206.
    1. Kania G, Rudnik M, Distler O. Involvement of the myeloid cell compartment in fibrogenesis and systemic sclerosis. Nat Rev Rheumatol. 2019;15(5):288–302. doi: 10.1038/s41584-019-0212-z.
    1. van Laar JM, Farge D, Sont JK, Naraghi K, Marjanovic Z, Larghero J, Schuerwegh AJ, Marijt EW, Vonk MC, Schattenberg AV, Matucci-Cerinic M, Voskuyl AE, van de Loosdrecht A, Daikeler T, Kötter I, Schmalzing M, Martin T, Lioure B, Weiner SM, Kreuter A, Deligny C, Durand JM, Emery P, Machold KP, Sarrot-Reynauld F, Warnatz K, Adoue DF, Constans J, Tony HP, del Papa N, Fassas A, Himsel A, Launay D, Lo Monaco A, Philippe P, Quéré I, Rich É, Westhovens R, Griffiths B, Saccardi R, van den Hoogen F, Fibbe WE, Socié G, Gratwohl A, Tyndall A, EBMT/EULAR Scleroderma Study Group Autologous hematopoietic stem cell transplantation vs intravenous pulse cyclophosphamide in diffuse cutaneous systemic sclerosis: a randomized clinical trial. Jama. 2014;311(24):2490–2498. doi: 10.1001/jama.2014.6368.
    1. O’Reilly S. Innate immunity in systemic sclerosis pathogenesis. Clin Sci. 2014;126(5):329–337. doi: 10.1042/CS20130367.
    1. Skaug B, Assassi S. Type I interferon dysregulation in systemic sclerosis. Cytokine. 2020;132:154635. doi: 10.1016/j.cyto.2018.12.018.
    1. Brkic Z, van Bon L, Cossu M, van Helden-Meeuwsen CG, Vonk MC, Knaapen H, van den Berg W, Dalm VA, Van Daele PL, Severino A, 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. 2016;75(8):1567–1573. doi: 10.1136/annrheumdis-2015-207392.
    1. Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70(11):2029–2036. doi: 10.1136/ard.2011.150326.
    1. Wuttge DM, Lood C, Tufvesson E, Scheja A, Truedsson L, Bengtsson AA, Hesselstrand R. Increased serum type I interferon activity in early systemic sclerosis patients is associated with antibodies against Sjogren’s syndrome antigens and nuclear ribonucleoprotein antigens. Scand J Rheumatol. 2013;42(3):235–240. doi: 10.3109/03009742.2012.736532.
    1. Higashi-Kuwata N, Jinnin M, Makino T, Fukushima S, Inoue Y, Muchemwa FC, Yonemura Y, Komohara Y, Takeya M, Mitsuya H, Ihn H. Characterization of monocyte/macrophage subsets in the skin and peripheral blood derived from patients with systemic sclerosis. Arthritis Res Ther. 2010;12(4):R128. doi: 10.1186/ar3066.
    1. Jonsson S, Andersson G, Fex T, Fristedt T, Hedlund G, Jansson K, Abramo L, Fritzson I, Pekarski O, Runstrom A, et al. Synthesis and biological evaluation of new 1,2-dihydro-4-hydroxy-2-oxo-3-quinolinecarboxamides for treatment of autoimmune disorders: structure-activity relationship. J Med Chem. 2004;47(8):2075–2088. doi: 10.1021/jm031044w.
    1. Vollmer TL, Sorensen PS, Selmaj K, Zipp F, Havrdova E, Cohen JA, Sasson N, Gilgun-Sherki Y, Arnold DL, Group BS A randomized placebo-controlled phase III trial of oral laquinimod for multiple sclerosis. J Neurol. 2014;261(4):773–783. doi: 10.1007/s00415-014-7264-4.
    1. Comi G, Jeffery D, Kappos L, Montalban X, Boyko A, Rocca MA, Filippi M, Group AS Placebo-controlled trial of oral laquinimod for multiple sclerosis. N Engl J Med. 2012;366(11):1000–1009. doi: 10.1056/NEJMoa1104318.
    1. Bjork P, Bjork A, Vogl T, Stenstrom M, Liberg D, Olsson A, Roth J, Ivars F, Leanderson T. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides. PLoS Biol. 2009;7(4):e97. doi: 10.1371/journal.pbio.1000097.
    1. Foell D, Roth J. Proinflammatory S100 proteins in arthritis and autoimmune disease. Arthritis Rheum. 2004;50(12):3762–3771. doi: 10.1002/art.20631.
    1. Nikitorowicz-Buniak J, Shiwen X, Denton CP, Abraham D, Stratton R. Abnormally differentiating keratinocytes in the epidermis of systemic sclerosis patients show enhanced secretion of CCN2 and S100A9. J Investigative Dermatol. 2014;134(11):2693–2702. doi: 10.1038/jid.2014.253.
    1. Xu X, Wu WY, Tu WZ, Chu HY, Zhu XX, Liang MR, Xue Y, Wang JC, Zou HJ. Increased expression of S100A8 and S100A9 in patients with diffuse cutaneous systemic sclerosis. A correlation with organ involvement and immunological abnormalities. Clin Rheumatol. 2013;32(10):1501–1510. doi: 10.1007/s10067-013-2305-4.
    1. Helmersson S, Sundstedt A, Deronic A, Leanderson T, Ivars F. Amelioration of experimental autoimmune encephalomyelitis by the quinoline-3-carboxamide paquinimod: reduced priming of proinflammatory effector CD4(+) T cells. Am J Pathol. 2013;182(5):1671–1680. doi: 10.1016/j.ajpath.2013.01.032.
    1. Bengtsson AA, Sturfelt G, Lood C, Ronnblom L, van Vollenhoven RF, Axelsson B, Sparre B, Tuvesson H, Ohman MW, Leanderson T. Pharmacokinetics, tolerability, and preliminary efficacy of paquinimod (ABR-215757), a new quinoline-3-carboxamide derivative: studies in lupus-prone mice and a multicenter, randomized, double-blind, placebo-controlled, repeat-dose, dose-ranging study in patients with systemic lupus erythematosus. Arthritis Rheum. 2012;64(5):1579–1588. doi: 10.1002/art.33493.
    1. Yan L, Bjork P, Butuc R, Gawdzik J, Earley J, Kim G, Hofmann Bowman MA. Beneficial effects of quinoline-3-carboxamide (ABR-215757) on atherosclerotic plaque morphology in S100A12 transgenic ApoE null mice. Atherosclerosis. 2013;228(1):69–79. doi: 10.1016/j.atherosclerosis.2013.02.023.
    1. Fransen Pettersson N, Deronic A, Nilsson J, Hannibal TD, Hansen L, Schmidt-Christensen A, Ivars F, Holmberg D. The immunomodulatory quinoline-3-carboxamide paquinimod reverses established fibrosis in a novel mouse model for liver fibrosis. PLoS One. 2018;13(9):e0203228. doi: 10.1371/journal.pone.0203228.
    1. Stenstrom M, Nyhlen HC, Torngren M, Liberg D, Sparre B, Tuvesson H, Eriksson H, Leanderson T. Paquinimod reduces skin fibrosis in tight skin 1 mice, an experimental model of systemic sclerosis. J Dermatol Sci. 2016;83(1):52–59. doi: 10.1016/j.jdermsci.2016.04.006.
    1. Bengtsson ASG, Rönnblom L, Gunnarsson I, Svenungsson E, Jacobsen S, Lood C, Sparre B, Wallén Öhman M, Tuvesson H, Leanderson T. Poster presented at Eular Annual European Congress of Rheumatology. 2011. An exploratory study to evaluate changes in disease activity and biomarkers during treatment with ABR-215757 in patients with mild active systemic lupus erythematosus (SLE)
    1. Masi A, RG, Medsger T. Preliminary criteria for the classification of systemic sclerosis (scleroderma). Subcommittee for scleroderma criteria of the American Rheumatism Association Diagnostic and Therapeutic Criteria Committee. Arthritis Rheum. 1980;23(5):581–590. doi: 10.1002/art.1780230510.
    1. Domsic RT, Rodriguez-Reyna T, Lucas M, Fertig N, Medsger TA., Jr Skin thickness progression rate: a predictor of mortality and early internal organ involvement in diffuse scleroderma. Ann Rheum Dis. 2011;70(1):104–109. doi: 10.1136/ard.2009.127621.
    1. Clements P, Lachenbruch P, Siebold J, White B, Weiner S, Martin R, Weinstein A, Weisman M, Mayes M, Collier D, et al. Inter and intraobserver variability of total skin thickness score (modified Rodnan TSS) in systemic sclerosis. J Rheumatol. 1995;22(7):1281–1285.
    1. Bengtsson A SG, Rönnblom L, van Vollenhoven R, Wallén Öhman M, Tuvesson H, Lando P, Edman K, Liberg D, Källberg E, Olsson A, Leanderson T: A phase I, dose escalation study to evaluate the tolerability of ABR-215757 in patients with systemic lupus erythematosus (SLE). Poster presented at EULAR (The European League Against Rheumatism) Conference, Copenhagen, Denmark 2009, June 10 (Active Biotech Doc. No. 0930104).
    1. Hayflick L. The establishment of a line (WISH) of human amnion cells in continuous cultivation. Exp Cell Res. 1961;23(1):14–20. doi: 10.1016/0014-4827(61)90059-3.
    1. Wu M, Assassi S. The role of type 1 interferon in systemic sclerosis. Front Immunol. 2013;4:266. doi: 10.3389/fimmu.2013.00266.
    1. Distler JH, Jungel A, Caretto D, Schulze-Horsel U, Kowal-Bielecka O, Gay RE, Michel BA, Muller-Ladner U, Kalden JR, Gay S, et al. Monocyte chemoattractant protein 1 released from glycosaminoglycans mediates its profibrotic effects in systemic sclerosis via the release of interleukin-4 from T cells. Arthritis Rheum. 2006;54(1):214–225. doi: 10.1002/art.21497.
    1. Distler O, Pap T, Kowal-Bielecka O, Meyringer R, Guiducci S, Landthaler M, et al. Overexpression of monocyte chemoattractant protein 1 in systemic sclerosis: role of platelet-derived growth factor and effects on monocyte chemotaxis and collagen synthesis. Arthritis Rheum. 2001;44(11):2665–78. 10.1002/1529-0131(200111)44:11<2665::AID-ART446>;2-S.
    1. Carulli MT, Ong VH, Ponticos M, Shiwen X, Abraham DJ, Black CM, Denton CP. Chemokine receptor CCR2 expression by systemic sclerosis fibroblasts: evidence for autocrine regulation of myofibroblast differentiation. Arthritis Rheum. 2005;52(12):3772–3782. doi: 10.1002/art.21396.
    1. Deronic A, Helmersson S, Leanderson T, Ivars F. The quinoline-3-carboxamide paquinimod (ABR-215757) reduces leukocyte recruitment during sterile inflammation: leukocyte- and context-specific effects. Int Immunopharmacol. 2014;18(2):290–297. doi: 10.1016/j.intimp.2013.12.008.
    1. Farina G, Lafyatis D, Lemaire R, Lafyatis R. 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. Assassi S, Mayes MD, Arnett FC, Gourh P, Agarwal SK, McNearney TA, Chaussabel D, Oommen N, Fischbach M, Shah KR, et al. Systemic sclerosis and lupus: points in an interferon-mediated continuum. Arthritis Rheum. 2010;62(2):589–598. doi: 10.1002/art.27224.
    1. Goldberg A, Geppert T, Schiopu E, Frech T, Hsu V, Simms RW, Peng SL, Yao Y, Elgeioushi N, Chang L, Wang B, Yoo S. Dose-escalation of human anti-interferon-alpha receptor monoclonal antibody MEDI-546 in subjects with systemic sclerosis: a phase 1, multicenter, open label study. Arthritis Res Ther. 2014;16(1):R57. doi: 10.1186/ar4492.
    1. Kissin EY, Merkel PA, Lafyatis R. Myofibroblasts and hyalinized collagen as markers of skin disease in systemic sclerosis. Arthritis Rheum. 2006;54(11):3655–3660. doi: 10.1002/art.22186.
    1. Lafyatis R, Kissin E, York M, Farina G, Viger K, Fritzler MJ, Merkel PA, Simms RW. B cell depletion with rituximab in patients with diffuse cutaneous systemic sclerosis. Arthritis Rheum. 2009;60(2):578–583. doi: 10.1002/art.24249.
    1. Enocsson H, Gullstrand B, Eloranta ML, Wettero J, Leonard D, Ronnblom L, Bengtsson AA, Sjowall C. C-reactive protein levels in systemic lupus erythematosus are modulated by the interferon gene signature and CRP gene polymorphism rs1205. Front Immunol. 2020;11:622326. doi: 10.3389/fimmu.2020.622326.

Source: PubMed

3
Subscribe