Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study

Objective To assess the safety and efficacy of secukinumab, a fully human monoclonal anti-interleukin-17A antibody, in patients with rheumatoid arthritis (RA). Methods Patients (n=237) with inadequate response to methotrexate were randomly assigned to receive monthly subcutaneous injections of secukinumab 25 mg, 75 mg, 150 mg, 300 mg or placebo. The primary endpoint was the American College of Rheumatology 20% response (ACR20) at week 16. Results Demographics and baseline characteristics were comparable across all treatment groups. The primary efficacy endpoint was not achieved: the proportion of ACR20 responders at week 16 with secukinumab 25–300 mg was 36.0–53.7% versus placebo (34%). Disease activity score in 28 joints (DAS28)–C-reactive protein (CRP) was a secondary endpoint and clinically relevant decreases with secukinumab 75–300 mg were reported versus placebo. Serum high sensitivity CRP levels at week 16 were significantly reduced with secukinumab 75 mg, 150 mg and 300 mg doses versus placebo. The safety profile of secukinumab was consistent with that seen with other biological agents. Most adverse events (AE) were mild to moderate in severity. Infections were slightly more frequent with secukinumab than placebo. Six serious AE were reported: secukinumab 75 mg (one), secukinumab 300 mg (four) and placebo (one). Conclusions ACR20 response rates differed between secukinumab 75 mg, 150 mg and 300 mg doses and placebo; however, the primary efficacy endpoint was not achieved. Greater decreases in DAS28 were observed with secukinumab 75 mg, 150 mg and 300 mg than placebo. There were no unexpected safety signals and no specific organ-related toxicities. Further trials with secukinumab in the treatment of RA are warranted.

[1]  S. Nakae,et al.  Suppression of Immune Induction of Collagen-Induced Arthritis in IL-17-Deficient Mice 1 , 2003, The Journal of Immunology.

[2]  J. Banchereau,et al.  T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines , 1996, The Journal of experimental medicine.

[3]  P. Miossec,et al.  IL-17 as a future therapeutic target for rheumatoid arthritis , 2009, Nature Reviews Rheumatology.

[4]  L. Joosten,et al.  IL-17 derived from juxta-articular bone and synovium contributes to joint degradation in rheumatoid arthritis , 2001, Arthritis research.

[5]  I. Buchan,et al.  Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. , 2006, JAMA.

[6]  W. Gross,et al.  A phase II study of interferon-alpha for the treatment of refractory Churg-Strauss syndrome. , 2008, Clinical and experimental rheumatology.

[7]  K. Hillan,et al.  Pathways by which interleukin 17 induces articular cartilage breakdown in vitro and in vivo. , 2001, Cytokine.

[8]  M. Liang,et al.  The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. , 1988, Arthritis and rheumatism.

[9]  D. Furst The risk of infections with biologic therapies for rheumatoid arthritis. , 2010, Seminars in arthritis and rheumatism.

[10]  R. Chang,et al.  The American College of Rheumatology 1991 revised criteria for the classification of global functional status in rheumatoid arthritis. , 1992, Arthritis and rheumatism.

[11]  T. Martin,et al.  IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. , 1999, The Journal of clinical investigation.

[12]  V. Kuchroo,et al.  Interleukin-17 and type 17 helper T cells. , 2009, The New England journal of medicine.

[13]  T. Pincus,et al.  Contemporary disease modifying antirheumatic drugs (DMARD) in patients with recent onset rheumatoid arthritis in a US private practice: methotrexate as the anchor drug in 90% and new DMARD in 30% of patients. , 2002, The Journal of rheumatology.

[14]  A. Skapenko,et al.  Role of Th17 cells in human autoimmune arthritis. , 2010, Arthritis and rheumatism.

[15]  Kaleb Michaud,et al.  Epidemiological studies in incidence, prevalence, mortality, and comorbidity of the rheumatic diseases , 2009, Arthritis research & therapy.

[16]  T. Moseley,et al.  Interleukin-17 family and IL-17 receptors. , 2003, Cytokine & growth factor reviews.

[17]  L. Joosten,et al.  Treatment with a neutralizing anti-murine interleukin-17 antibody after the onset of collagen-induced arthritis reduces joint inflammation, cartilage destruction, and bone erosion. , 2004, Arthritis and rheumatism.

[18]  P. Tak,et al.  Effects of AIN457, a Fully Human Antibody to Interleukin-17A, on Psoriasis, Rheumatoid Arthritis, and Uveitis , 2010, Science Translational Medicine.

[19]  J. Barton,et al.  Patient preferences and satisfaction in the treatment of rheumatoid arthritis with biologic therapy , 2009, Patient preference and adherence.

[20]  T. Pincus,et al.  Mortality in rheumatoid arthritis: 2008 update. , 2008, Clinical and experimental rheumatology.

[21]  B. Kirkham,et al.  Reduction of joint inflammation and bone erosion in rat adjuvant arthritis by treatment with interleukin-17 receptor IgG1 Fc fusion protein. , 2002, Arthritis and rheumatism.

[22]  Yulan He,et al.  IL-17 stimulates the production and expression of proinflammatory cytokines, IL-beta and TNF-alpha, by human macrophages. , 1998, Journal of immunology.

[23]  P. Miossec Interleukin-17 in rheumatoid arthritis: if T cells were to contribute to inflammation and destruction through synergy. , 2003, Arthritis and rheumatism.

[24]  D. Furst,et al.  Description of the Efficacy and Safety of Three New Biologics in the Treatment of Rheumatoid Arthritis , 2010, The Korean journal of internal medicine.

[25]  K. Książek,et al.  Interleukin-17: a mediator of inflammatory responses , 2004, Cellular and Molecular Life Sciences CMLS.