A proof-of-concept study with the tyrosine kinase inhibitor nilotinib in spondyloarthritis

Jacqueline E Paramarta, Maureen C Turina, Troy Noordenbos, Tanja F Heijda, Iris C Blijdorp, Nataliya Yeremenko, Dominique Baeten, Jacqueline E Paramarta, Maureen C Turina, Troy Noordenbos, Tanja F Heijda, Iris C Blijdorp, Nataliya Yeremenko, Dominique Baeten

Abstract

Background: To evaluate the immunomodulating and clinical effects of nilotinib, a tyrosine kinase inhibitor, in a proof-of-concept study in spondyloarthritis (SpA) assessing the mast cell as potential novel therapeutic target in this disease.

Methods: Twenty eight patients with active peripheral (pSpA) and/or axial SpA (axSpA) were included in a randomized, double-blind, placebo-controlled clinical trial (Trial registration: Trialregister.nl NTR2834). Patients were treated 1:1 with nilotinib or placebo for 12 weeks, followed by an open label extension for another 12 weeks. Paired synovial tissue biopsies, serum sampling and assessment of clinical symptoms were performed serially.

Results: In pSpA (n = 13) synovial inflammation appeared to diminish after 12 weeks of nilotinib treatment as evidenced by histopathology (decrease in number of infiltrating CD68+ and CD163+ macrophages and mast cells). Compared to placebo mRNA expression of c-Kit as mast cell marker (p = 0.037) and of pro-inflammatory cytokines such as IL-6 (p = 0.024) were reduced. The reduction of synovial inflammation was paralleled by a decrease in serum biomarkers of inflammation such as C-reactive protein (p = 0.024) and calprotectin (p = 0.055). Also clinical parameters such as patient's global assessment of disease activity (p = 0.031) and ankylosing spondylitis disease activity score (p = 0.031) showed improvement upon 12 weeks of nilotinib but not placebo treatment. This improvement was further augmented at week 24. In contrast to pSpA, neither serum biomarkers of inflammation nor clinical parameters improved upon nilotinib treatment in axSpA. During the trial one serious adverse event occurred, which was considered unrelated to the study drug.

Conclusions: This small proof-of-concept study suggests that nilotinib treatment modulates inflammation and clinical symptoms in pSpA. A similar effect was not seen in axSpA.

Trial registration: trialregister.nl registration code NTR2834 registered 31 March 2011.

Keywords: Mast cells; Nilotinib; Randomized controlled trial; Spondyloarthritis; Tyrosine kinase inhibitor.

Figures

Fig. 1
Fig. 1
Synovial tissue mRNA expression in peripheral spondyloarthritis. Effect of nilotinib and placebo treatment on in vivo synovial tissue mRNA expression in peripheral spondyloarthritis as assessed by quantitative polymerase chain reaction. The panel represents the transcription of c-Kit, interleukin-6 (IL-6), IL-8, tumor necrosis factor (TNF), and IL-23 before and after treatment with nilotinib (ae) or placebo (fj). The lines connect the data points for each patient between weeks (week) 0 and 12
Fig. 2
Fig. 2
Serum biomarkers in peripheral spondyloarthritis. Effect of nilotinib and placebo on serum biomarkers of patients with peripheral spondyloarthritis. The panel represents the C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), calprotectin and matrix metalloproteinase-3 (MMP-3) levels before (week 0) and after treatment (week 12) with nilotinib (ad) or placebo (eh). The lines connect the data points for each patient. *P value <0.05
Fig. 3
Fig. 3
Clinical disease activity in peripheral spondyloarthritis. Changes in clinical disease activity parameters during treatment with nilotinib or placebo from week 0 until week 12, and during the open label extension phase with nilotinib from week 12 until week 24 in patients with peripheral spondyloarthritis. The panel represents the median (interquartile range) in patient’s global assessment of disease activity (a), physician’s global assessment of disease activity (b), swollen joint count (c), tender joint count (d), ankylosing spondylitis disease activity score (ASDAS) (e), and the percentage of patients achieving ASDAS clinically important improvement (f). *P value <0.05 compared to baseline
Fig. 4
Fig. 4
Serum biomarkers in axial spondyloarthritis. Effect of nilotinib and placebo on serum biomarkers of patients with axial spondyloarthritis. The panel represents the C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), calprotectin and matrix metalloproteinase-3 (MMP-3) levels before (week 0) and after treatment (week 12) with nilotinib (ad) or placebo (eh). The lines connect the data points for each patient. *P value <0.05
Fig. 5
Fig. 5
Clinical disease activity in axial spondyloarthritis. Changes in clinical disease activity parameters during treatment with nilotinib or placebo from week 0 until week 12, and during the open label extension phase with nilotinib from week 12 until week 24 in patients with axial spondyloarthritis. The panel represents the median (interquartile range) in patient’s global assessment of disease activity (a), physician’s global assessment of disease activity (b), bath ankylosing spondylitis disease activity index (BASDAI) (c), ankylosing spondylitis disease activity score (ASDAS) (d), and the percentage of patients achieving a BASDAI50 response (e), and ASDAS clinically important improvement (f). *P value <0.05 compared to baseline

References

    1. Dougados M, Baeten D. Spondyloarthritis. Lancet. 2011;377:2127–2137. doi: 10.1016/S0140-6736(11)60071-8.
    1. Baraliakos X, Listing J, Brandt J, Zink A, Alten R, Burmester G, et al. Clinical response to discontinuation of anti-TNF therapy in patients with ankylosing spondylitis after 3 years of continuous treatment with infliximab. Arthritis Res Ther. 2005;7:R439–R444. doi: 10.1186/ar1693.
    1. Paramarta JE, Heijda TF, Baeten DL. Fast relapse upon discontinuation of tumour necrosis factor blocking therapy in patients with peripheral spondyloarthritis. Ann Rheum Dis. 2013;72:1581–1582. doi: 10.1136/annrheumdis-2013-203327.
    1. Braun J, Baraliakos X, Hermann KG, Deodhar A, van der Heijde D, Inman R, et al. The effect of two golimumab doses on radiographic progression in ankylosing spondylitis: results through 4 years of the GO-RAISE trial. Ann Rheum Dis. 2014;73:1107–1113. doi: 10.1136/annrheumdis-2012-203075.
    1. van der Heijde D, Landewe R, Einstein S, Ory P, Vosse D, Ni L, et al. Radiographic progression of ankylosing spondylitis after up to 2 years of treatment with etanercept. Arthritis Rheum. 2008;58:1324–1331. doi: 10.1002/art.23471.
    1. van der Heijde D, Landewe R, Baraliakos X, Houben H, Tubergen AV, Williamson P, et al. Radiographic findings following 2 years of infliximab therapy in patients with ankylosing spondylitis. Arthritis Rheum. 2008;58:3063–3070. doi: 10.1002/art.23901.
    1. van der Heijde D, Salonen D, Weissman BN, Landewe R, Maksymowych WP, Kupper H, et al. Assessment of radiographic progression in the spines of patients with ankylosing spondylitis treated with adalimumab for up to 2 years. Arthritis Res Ther. 2009;11:R127. doi: 10.1186/ar2794.
    1. Finzel S, Kraus S, Schmidt S, Hueber A, Rech J, Engelke K, et al. Bone anabolic changes progress in psoriatic arthritis patients despite treatment with methotrexate or tumour necrosis factor inhibitors. Ann Rheum Dis. 2013;72:1176–1181. doi: 10.1136/annrheumdis-2012-201580.
    1. Sandler C, Lindstedt KA, Joutsiniemi S, Lappalainen J, Juutilainen T, Kolah J, et al. Selective activation of mast cells in rheumatoid synovial tissue results in production of TNF-alpha, IL-1beta and IL-1Ra. Inflamm Res. 2007;56:230–239. doi: 10.1007/s00011-007-6135-1.
    1. Woolley DE, Tetlow LC. Mast cell activation and its relation to proinflammatory cytokine production in the rheumatoid lesion. Arthritis Res. 2000;2:65–74. doi: 10.1186/ar70.
    1. Hueber AJ, Asquith DL, Miller AM, Reilly J, Kerr S, Leipe J, et al. Mast cells express IL-17A in rheumatoid arthritis synovium. J Immunol. 2010;184:3336–3340. doi: 10.4049/jimmunol.0903566.
    1. Eklund KK. Mast cells in the pathogenesis of rheumatic diseases and as potential targets for anti-rheumatic therapy. Immunol Rev. 2007;217:38–52. doi: 10.1111/j.1600-065X.2007.00504.x.
    1. Noordenbos T, Yeremenko N, Gofita I, van de Sande M, Tak PP, Canete JD, et al. Interleukin-17-positive mast cells contribute to synovial inflammation in spondylarthritis. Arthritis Rheum. 2012;64:99–109. doi: 10.1002/art.33396.
    1. Baeten D, Baraliakos X, Braun J, Sieper J, Emery P, van der Heijde D, et al. Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial. Lancet. 2013;382:1705–1713. doi: 10.1016/S0140-6736(13)61134-4.
    1. McInnes IB, Sieper J, Braun J, Emery P, van der Heijde D, Isaacs JD, et al. Efficacy and safety of secukinumab, a fully human anti-interleukin-17A monoclonal antibody, in patients with moderate-to-severe psoriatic arthritis: a 24-week, randomised, double-blind, placebo-controlled, phase II proof-of-concept trial. Ann Rheum Dis. 2014;73:349–356. doi: 10.1136/annrheumdis-2012-202646.
    1. Baeten DL, Kuchroo VK. How cytokine networks fuel inflammation: interleukin-17 and a tale of two autoimmune diseases. Nat Med. 2013;19:824–825. doi: 10.1038/nm.3268.
    1. Clegg DO, Reda DJ, Abdellatif M. Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondyloarthropathies: a Department of Veterans Affairs Cooperative Study. Arthritis Rheum. 1999;42:2325–2329. doi: 10.1002/1529-0131(199911)42:11<2325::AID-ANR10>;2-C.
    1. Barrett KE, Tashof TL, Metcalfe DD. Inhibition of IgE-mediated mast cell degranulation by sulphasalazine. Eur J Pharmacol. 1985;107:279–281. doi: 10.1016/0014-2999(85)90071-8.
    1. Bissonnette EY, Enciso JA, Befus AD. Inhibitory effects of sulfasalazine and its metabolites on histamine release and TNF-alpha production by mast cells. J Immunol. 1996;156:218–223.
    1. Giles FJ, O’Dwyer M, Swords R. Class effects of tyrosine kinase inhibitors in the treatment of chronic myeloid leukemia. Leukemia. 2009;23:1698–1707. doi: 10.1038/leu.2009.111.
    1. Weisberg E, Manley PW, Breitenstein W, Bruggen J, Cowan-Jacob SW, Ray A, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005;7:129–141. doi: 10.1016/j.ccr.2005.01.007.
    1. Juurikivi A, Sandler C, Lindstedt KA, Kovanen PT, Juutilainen T, Leskinen MJ, et al. Inhibition of c-kit tyrosine kinase by imatinib mesylate induces apoptosis in mast cells in rheumatoid synovia: a potential approach to the treatment of arthritis. Ann Rheum Dis. 2005;64:1126–1131. doi: 10.1136/ard.2004.029835.
    1. Eklund KK, Remitz A, Kautiainen H, Reitamo S, Leirisalo-Repo M. Three months treatment of active spondyloarthritis with imatinib mesylate: an open-label pilot study with six patients. Rheumatology (Oxford) 2006;45:1573–1575. doi: 10.1093/rheumatology/kel365.
    1. Saglio G, Kim DW, Issaragrisil S, le Coutre P, Etienne G, Lobo C, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010;362:2251–2259. doi: 10.1056/NEJMoa0912614.
    1. Kantarjian HM, Giles F, Gattermann N, Bhalla K, Alimena G, Palandri F, et al. Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood. 2007;110:3540–3546. doi: 10.1182/blood-2007-03-080689.
    1. Dougados M, van der Linden S, Juhlin R, Huitfeldt B, Amor B, Calin A, et al. The European Spondyloarthropathy Study Group preliminary criteria for the classification of spondyloarthropathy. Arthritis Rheum. 1991;34:1218–1227. doi: 10.1002/art.1780341003.
    1. Baeten D, Van den Bosch F, Elewaut D, Stuer A, Veys EM, De Keyser F. Needle arthroscopy of the knee with synovial biopsy sampling: technical experience in 150 patients. Clin Rheumatol. 1999;18:434–441. doi: 10.1007/s100670050134.
    1. Gerlag DM, Tak PP. How to perform and analyse synovial biopsies. Best Pract Res Clin Rheumatol. 2009;23:221–232. doi: 10.1016/j.berh.2009.01.006.
    1. Baeten D, Moller HJ, Delanghe J, Veys EM, Moestrup SK, De Keyser F. Association of CD163+ macrophages and local production of soluble CD163 with decreased lymphocyte activation in spondyloarthropathy synovitis. Arthritis Rheum. 2004;50:1611–1623. doi: 10.1002/art.20174.
    1. Baeten D, Kruithof E, De Rycke L, Boots AM, Mielants H, Veys EM, et al. Infiltration of the synovial membrane with macrophage subsets and polymorphonuclear cells reflects global disease activity in spondyloarthropathy. Arthritis Res Ther. 2005;7:R359–R369. doi: 10.1186/ar1501.
    1. Kruithof E, Baeten D, De Rycke L, Vandooren B, Foell D, Roth J, et al. Synovial histopathology of psoriatic arthritis, both oligo- and polyarticular, resembles spondyloarthropathy more than it does rheumatoid arthritis. Arthritis Res Ther. 2005;7:R569–R580. doi: 10.1186/ar1698.
    1. Vandooren B, Kruithof E, Yu DT, Rihl M, Gu J, De Rycke L, et al. Involvement of matrix metalloproteinases and their inhibitors in peripheral synovitis and down-regulation by tumor necrosis factor alpha blockade in spondyloarthropathy. Arthritis Rheum. 2004;50:2942–2953. doi: 10.1002/art.20477.
    1. De Rycke L, Baeten D, Foell D, Kruithof E, Veys EM, Roth J, et al. Differential expression and response to anti-TNFalpha treatment of infiltrating versus resident tissue macrophage subsets in autoimmune arthritis. J Pathol. 2005;206:17–27. doi: 10.1002/path.1758.
    1. Lukas C, Landewe R, Sieper J, Dougados M, Davis J, Braun J, et al. Development of an ASAS-endorsed disease activity score (ASDAS) in patients with ankylosing spondylitis. Ann Rheum Dis. 2009;68:18–24. doi: 10.1136/ard.2008.094870.
    1. Machado P, Landewe R, Lie E, Kvien TK, Braun J, Baker D, et al. Ankylosing spondylitis disease activity score (ASDAS): defining cut-off values for disease activity states and improvement scores. Ann Rheum Dis. 2011;70:47–53. doi: 10.1136/ard.2010.138594.
    1. Garrett S, Jenkinson T, Kennedy LG, Whitelock H, Gaisford P, Calin A. A new approach to defining disease status in ankylosing spondylitis: the bath ankylosing spondylitis disease activity index. J Rheumatol. 1994;21:2286–2291.
    1. Braun J, Davis J, Dougados M, Sieper J, van der Linden S, van der Heijde D. First update of the international ASAS consensus statement for the use of anti-TNF agents in patients with ankylosing spondylitis. Ann Rheum Dis. 2006;65:316–320. doi: 10.1136/ard.2005.040758.
    1. Kruithof E, De Rycke L, Vandooren B, De Keyser F, FitzGerald O, McInnes I, et al. Identification of synovial biomarkers of response to experimental treatment in early-phase clinical trials in spondylarthritis. Arthritis Rheum. 2006;54:1795–1804. doi: 10.1002/art.21914.
    1. Paramarta JE, De Rycke L, Heijda TF, Ambarus CA, Vos K, Dinant HJ, et al. Efficacy and safety of adalimumab for the treatment of peripheral arthritis in spondyloarthritis patients without ankylosing spondylitis or psoriatic arthritis. Ann Rheum Dis. 2013;72:1793–1799. doi: 10.1136/annrheumdis-2012-202245.
    1. Bernink JH, Peters CP, Munneke M, te Velde AA, Meijer SL, Weijer K, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol. 2013;14:221–229. doi: 10.1038/ni.2534.
    1. Baeten D, Demetter P, Cuvelier CA, Kruithof E, Van Damme N, De Vos M, et al. Macrophages expressing the scavenger receptor CD163: a link between immune alterations of the gut and synovial inflammation in spondyloarthropathy. J Pathol. 2002;196:343–350. doi: 10.1002/path.1044.
    1. Vandooren B, Noordenbos T, Ambarus C, Krausz S, Cantaert T, Yeremenko N, et al. Absence of a classically activated macrophage cytokine signature in peripheral spondylarthritis, including psoriatic arthritis. Arthritis Rheum. 2009;60:966–975. doi: 10.1002/art.24406.
    1. Yeremenko N, Noordenbos T, Cantaert T, van Tok M, van de Sande M, Canete JD, et al. Disease-specific and inflammation-independent stromal alterations in spondylarthritis synovitis. Arthritis Rheum. 2013;65:174–185. doi: 10.1002/art.37704.
    1. Baeten D, Breban M, Lories R, Schett G, Sieper J. Are spondyloarthritides related but distinct conditions or a single disease with a heterogeneous phenotype? Arthritis Rheum. 2013;65:12–20. doi: 10.1002/art.37829.
    1. Appel H, Maier R, Wu P, Scheer R, Hempfing A, Kayser R, et al. Analysis of IL-17(+) cells in facet joints of patients with spondyloarthritis suggests that the innate immune pathway might be of greater relevance than the Th17-mediated adaptive immune response. Arthritis Res Ther. 2011;13:R95. doi: 10.1186/ar3370.
    1. Van den Bosch F, Kruithof E, Baeten D, Herssens A, De Keyser F, Mielants H, et al. Randomized double-blind comparison of chimeric monoclonal antibody to tumor necrosis factor alpha (infliximab) versus placebo in active spondyloarthropathy. Arthritis Rheum. 2002;46:755–765. doi: 10.1002/art.511.
    1. van der Heijde D, Kivitz A, Schiff MH, Sieper J, Dijkmans BA, Braun J, et al. Efficacy and safety of adalimumab in patients with ankylosing spondylitis: results of a multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2006;54:2136–2146. doi: 10.1002/art.21913.
    1. Sieper J, van der Heijde D, Dougados M, Mease PJ, Maksymowych WP, Brown MA, et al. Efficacy and safety of adalimumab in patients with non-radiographic axial spondyloarthritis: results of a randomised placebo-controlled trial (ABILITY-1) Ann Rheum Dis. 2013;72:815–822. doi: 10.1136/annrheumdis-2012-201766.

Source: PubMed

3
Abonner