Ixekizumab, an interleukin-17A specific monoclonal antibody, for the treatment of biologic-naive patients with active psoriatic arthritis: results from the 24-week randomised, double-blind, placebo-controlled and active (adalimumab)-controlled period of the phase III trial SPIRIT-P1

Philip J Mease, Désirée van der Heijde, Christopher T Ritchlin, Masato Okada, Raquel S Cuchacovich, Catherine L Shuler, Chen-Yen Lin, Daniel K Braun, Chin H Lee, Dafna D Gladman, SPIRIT-P1 Study Group, N Barkham, L Bessette, R Blanco Alonso, E J Box, M Brooks, R Burgos Vargas, V Chandran, R Dhar, A Fernandez Nebro, R Fleischmann, K Flint, J Forstot, F Galvan Villegas, F Garcia-Fructuoso, O Garmish, M Geneva-Popova, P Geusens, G Gladstein, H Goto, E Griep, R Harrell, A Hou, M Howell, A Kivitz, S Klein, A Kolczewska, M Korkosz, D Lopez Montilla, C Lue, Y Makino, Jl Marenco de la Fuente, H Marzo-Ortega, T Miyamura, E Mueller, L Myasoutova, F Navarro Sarabia, A Ostor, K Papp, L Podrazilova, G Pulka, E-K Raussi, J Rosa, E Roussou, A Rychlewska-Hanczewska, K Sada, L Sedova, D Sikes, S Solomon, M Stack, M Stanislavchuk, K Suzuki, H Tahir, J Tälli, A Toncheva, A Turkiewicz, I Valter, S Vladeva, S Wolfe, N Zyablova, Philip J Mease, Désirée van der Heijde, Christopher T Ritchlin, Masato Okada, Raquel S Cuchacovich, Catherine L Shuler, Chen-Yen Lin, Daniel K Braun, Chin H Lee, Dafna D Gladman, SPIRIT-P1 Study Group, N Barkham, L Bessette, R Blanco Alonso, E J Box, M Brooks, R Burgos Vargas, V Chandran, R Dhar, A Fernandez Nebro, R Fleischmann, K Flint, J Forstot, F Galvan Villegas, F Garcia-Fructuoso, O Garmish, M Geneva-Popova, P Geusens, G Gladstein, H Goto, E Griep, R Harrell, A Hou, M Howell, A Kivitz, S Klein, A Kolczewska, M Korkosz, D Lopez Montilla, C Lue, Y Makino, Jl Marenco de la Fuente, H Marzo-Ortega, T Miyamura, E Mueller, L Myasoutova, F Navarro Sarabia, A Ostor, K Papp, L Podrazilova, G Pulka, E-K Raussi, J Rosa, E Roussou, A Rychlewska-Hanczewska, K Sada, L Sedova, D Sikes, S Solomon, M Stack, M Stanislavchuk, K Suzuki, H Tahir, J Tälli, A Toncheva, A Turkiewicz, I Valter, S Vladeva, S Wolfe, N Zyablova

Abstract

Objective: To assess the safety and efficacy of ixekizumab, a monoclonal antibody that inhibits interleukin-17A, in a double-blind phase III trial enrolling patients with active psoriatic arthritis (PsA).

Methods: Patients naive to biologic therapy with active PsA were randomised to subcutaneous injections of placebo (N=106), adalimumab 40 mg once every 2 weeks (active reference; N=101), ixekizumab 80 mg once every 2 weeks (IXEQ2W) (N=103), or ixekizumab 80 mg once every 4 weeks (IXEQ4W) (N=107). Both ixekizumab regimens included a 160-mg starting dose. The primary objective was to assess the superiority of IXEQ2W or IXEQ4W versus placebo as measured by the proportion of patients achieving an American College of Rheumatology 20 (ACR20) response at week 24.

Results: Significantly more patients treated with ixekizumab achieved an ACR20 response with IXEQ2W (62.1%) or IXEQ4W (57.9%) than placebo (30.2%) (p≤0.001; non-responder imputation method). Disease activity and functional disability were significantly improved with both ixekizumab doses versus placebo at weeks 12 and 24, and there was significantly less progression of structural damage at week 24 (p≤0.01). Clearance of plaque psoriasis was greater with ixekizumab than placebo (p≤0.001). Efficacy results with adalimumab, the active reference arm, showed significant improvements versus placebo. Treatment-emergent adverse events were more frequent with ixekizumab (65.7-66.4%) and adalimumab (64.4%) than placebo (47.2%) (p<0.05).

Conclusions: In biologic-naive patients with active PsA, ixekizumab treatment resulted in improvements in disease activity and physical function, as well as in the inhibition of structural damage progression. Overall, adverse events were more frequent in all active groups compared with placebo.

Trial registration number: NCT01695239; EudraCT2011-002326-49; Results.

Keywords: DMARDs (biologic); Psoriatic Arthritis; Spondyloarthritis; Treatment.

Conflict of interest statement

PJM reports grants and personal fees from Eli Lilly & Company during the conduct of the study. PJM also reports, outside the submitted work, grants, personal fees, and non-financial support from AbbVie, Amgen, Bristol Myers Squibb, Celgene, Crescendo, Genentech, Janssen, Pfizer and UCB Pharma; grants and personal fees from Merck and Novartis; and non-financial support from Corrona. DvdH reports personal fees from Eli Lilly & Company, during the conduct of the study. DvdH also reports personal fees from AbbVie, Amgen, AstraZeneca, Augurex, Bristol Myers Squibb, Celgene, Centocor, Chugai, Covagen, Daiichi, Galapagos, GlaxoSmithKline, Janssen, Merck, Novo Nordisk, Otsuka, Pfizer, Roche, Sanofi-Aventis, UCB Pharma and Vertex, outside the submitted work. DvdH is the director at Imaging Rheumatology BV. CTR reports personal fees from AbbVie, Amgen, Janssen, Novartis, UCB, Boerhinger Ingelheim, as well as grants from Amgen, outside the submitted work. MO reports grants and non-financial support from Eli Lilly & Company, during the conduct of this study. MO also reports non-financial support from Santen Pharmaceutical, Mitsubishi Tanabe Pharma, Pfizer and Abbott Japan, outside the submitted work. RSC and DKB were employees of Eli Lilly & Company, at the time this study was conducted. CLS, C-YL and CHL are employees of Eli Lilly & Company. DDG reports grants and personal fees from AbbVie, Amgen, Celgene, Janssen, Novartis, Pfizer and UCB, and personal fees from Eli Lilly & Company, outside the submitted work.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
Time course of ACR responses. The percentages of patients achieving ACR20 (A), ACR50 (B) and ACR70 (C) are shown. Patients with inadequate responses to treatment at week 16 or missing data were analysed as non-response up to week 24. aActive reference arm for comparison with placebo. The study was not powered to test equivalence or non-inferiority of ixekizumab versus adalimumab. ACR20/50/70, 20/50/70% American College of Rheumatology response; ADAQ2W, 40 mg adalimumab once every 2 weeks; IXEQ2W, 80 mg ixekizumab once every 2 weeks; IXEQ4W, 80 mg ixekizumab once every 4 weeks; PBO, placebo; Q2W, once every 2 weeks.
Figure 2
Figure 2
Time course of PASI responses. The percentages of patients achieving PASI 75 (A), PASI 90 (B) and PASI 100 (C) are shown. PASI was measured in patients with baseline psoriatic lesion(s) involving ≥3% body surface area. Patients with inadequate responses to treatment at week 16 or missing data were analysed as non-response up to week 24. aActive reference arm for comparison with placebo. The study was not powered to test equivalence or non-inferiority of ixekizumab versus adalimumab. PASI 75/90/100, Psoriasis Area and Severity Index Improvement Response for 75/90/100%; IXEQ2W, 80 mg ixekizumab once every 2 weeks; IXEQ4W, 80 mg ixekizumab once every 4 weeks; Q2W, once every 2 weeks.

References

    1. Gladman DD. Psoriatic arthritis. Dermatol Ther 2009;22:40–55. 10.1111/j.1529-8019.2008.01215.x
    1. de Vlam K, Gottlieb AB, Mease PJ. Current concepts in psoriatic arthritis: pathogenesis and management. Acta Derm Venereol 2014;94:627–34. 10.2340/00015555-1833
    1. Gladman DD, Antoni C, Mease P, et al. . Psoriatic arthritis: epidemiology, clinical features, course, and outcome. Ann Rheum Dis 2005;64(Suppl 2):ii14–17. 10.1136/ard.2004.032482
    1. Leung YY, Tam LS, Kun EW, et al. . Psoriatic arthritis as a distinct disease entity. J Postgrad Med 2007;53:63–71. 10.4103/0022-3859.30334
    1. Raychaudhuri SP. Role of IL-17 in psoriasis and psoriatic arthritis. Clin Rev Allergy Immunol 2013;44:183–93. 10.1007/s12016-012-8307-1
    1. Jandus C, Bioley G, Rivals JP, et al. . Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum 2008;58:2307–17. 10.1002/art.23655
    1. Kagami S, Rizzo HL, Lee JJ, et al. . Circulating Th17, Th22, and Th1 cells are increased in psoriasis. J Invest Dermatol 2010;130:1373–83. 10.1038/jid.2009.399
    1. Lin AM, Rubin CJ, Khandpur R, et al. . Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J Immunol 2011;187:490–500. 10.4049/jimmunol.1100123
    1. Noordenbos T, Yeremenko N, Gofita I, et al. . Interleukin-17-positive mast cells contribute to synovial inflammation in spondylarthritis. Arthritis Rheum 2012;64:99–109. 10.1002/art.33396
    1. Menon B, Gullick NJ, Walter GJ, et al. . Interleukin-17+CD8+T cells are enriched in the joints of patients with psoriatic arthritis and correlate with disease activity and joint damage progression. 2014;66:1272–81. 10.1002/art.38376
    1. Leijten EF, van Kempen TS, Boes M, et al. . Brief report: enrichment of activated group 3 innate lymphoid cells in psoriatic arthritis synovial fluid. Arthritis Rheumatol 2015;67:2673–8. 10.1002/art.39261
    1. McInnes IB, Mease PJ, Kirkham B, et al. . Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015;386:1137–46. 10.1016/S0140-6736(15)61134-5
    1. Mease PJ, McInnes IB, Kirkham B, et al. . Secukinumab inhibition of interleukin-17A in patients with psoriatic arthritis. N Engl J Med 2015;373:1329–39. 10.1056/NEJMoa1412679
    1. Taylor W, Gladman D, Helliwell P, et al. . Classification criteria for psoriatic arthritis: development of new criteria from a large international study. Arthritis Rheum 2006;54:2665–73. 10.1002/art.21972
    1. Felson DT, Anderson JJ, Boers M, et al. . American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995;38:727–35. 10.1002/art.1780380602
    1. Feldman SR, Krueger GG. Psoriasis assessment tools in clinical trials. Ann Rheum Dis 2005;64(Suppl 2):ii65–8; discussion ii69–73 10.1136/ard.2004.031237
    1. Weisman S, Pollack CR, Gottschalk RW. Psoriasis disease severity measures: comparing efficacy of treatments for severe psoriasis. J Dermatolog Treat 2003;14:158–65. 10.1080/09546630310013360
    1. Fries JF, Spitz P, Kraines RG, et al. . Measurement of patient outcome in arthritis. Arthritis Rheum 1980;23:137–45. 10.1002/art.1780230202
    1. Fries JF, Spitz PW, Young DY. The dimensions of health outcomes: the health assessment questionnaire, disability and pain scales. J Rheumatol 1982;9: 789–93.
    1. van der Heijde D, Sharp J, Wassenberg S, et al. . Psoriatic arthritis imaging: a review of scoring methods. Ann Rheum Dis 2005;64(Suppl 2):ii61–4. 10.1136/ard.2004.030809
    1. Wells G, Becker JC, Teng J, et al. . Validation of the 28-joint Disease Activity Score (DAS28) and European League Against Rheumatism response criteria based on C-reactive protein against disease progression in patients with rheumatoid arthritis, and comparison with the DAS28 based on erythrocyte sedimentation rate. Ann Rheum Dis 2009;68:954–60. 10.1136/ard.2007.084459
    1. Healy PJ, Helliwell PS. Measuring clinical enthesitis in psoriatic arthritis: assessment of existing measures and development of an instrument specific to psoriatic arthritis. Arthritis Rheum 2008;59:686–91. 10.1002/art.23568
    1. Helliwell PS, Firth J, Ibrahim GH, et al. . Development of an assessment tool for dactylitis in patients with psoriatic arthritis. J Rheumatol 2005;32:1745–50.
    1. Healy PJ, Helliwell PS. Measuring dactylitis in clinical trials: which is the best instrument to use? J Rheumatol 2007;34:1302–6.
    1. Kimball AB, Naegeli AN, Edson-Heredia E, et al. . Psychometric properties of the itch numeric rating scale in patients with moderate-to-severe plaque psoriasis. Br J Dermatol 2016;175:157–62.10.1111/bjd.14464
    1. Rich P, Scher RK. Nail psoriasis severity index: a useful tool for evaluation of nail psoriasis. J Am Acad Dermatol 2003;49:206–12. 10.1067/S0190-9622(03)00910-1
    1. Mease PJ, Woolley JM, Bitman B, et al. . Minimally important difference of Health Assessment Questionnaire in psoriatic arthritis: relating thresholds of improvement in functional ability to patient-rated importance and satisfaction. J Rheumatol 2011;38:2461–5. 10.3899/jrheum.110546
    1. Common Terminology Criteria for Adverse Events (CTCAE), version 4.03 Bethesda, MD: National Cancer Institute, 2006. (accessed 14 Jun 2010).
    1. Leonardi C, Matheson R, Zachariae C, et al. . Anti-interleukin-17 monoclonal antibody ixekizumab in chronic plaque psoriasis. N Engl J Med 2012;366: 1190–9. 10.1056/NEJMoa1109997
    1. Griffiths CE, Reich K, Lebwohl M, et al. . Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet 2015;386:541–51. 10.1016/S0140-6736(15)60125-8
    1. Mease PJ, Genovese MC, Greenwald MW, et al. . Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med 2014;370: 2295–306. 10.1056/NEJMoa1315231
    1. Huang W, Na L, Fidel PL, et al. . Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis 2004;190:624–31. 10.1086/422329
    1. Chimenti MS, Ballanti E, Perricone C, et al. . Immunomodulation in psoriatic arthritis: focus on cellular and molecular pathways. Autoimmun Rev 2013;12:599–606. 10.1016/j.autrev.2012.10.002
    1. Yamamoto T. Angiogenic and inflammatory properties of psoriatic arthritis. ISRN Dermatol 2013;2013:630620 10.1155/2013/630620

Source: PubMed

Подписаться