Translating Improvements with Ixekizumab in Clinical Trial Outcomes into Clinical Practice: ASAS40, Pain, Fatigue, and Sleep in Ankylosing Spondylitis

Philip Mease, Jessica A Walsh, Xenofon Baraliakos, Robert Inman, Kurt de Vlam, James Cheng-Chung Wei, Theresa Hunter, Gaia Gallo, David Sandoval, Fangyi Zhao, Yan Dong, Rebecca Bolce, Helena Marzo-Ortega, Philip Mease, Jessica A Walsh, Xenofon Baraliakos, Robert Inman, Kurt de Vlam, James Cheng-Chung Wei, Theresa Hunter, Gaia Gallo, David Sandoval, Fangyi Zhao, Yan Dong, Rebecca Bolce, Helena Marzo-Ortega

Abstract

Introduction: Ixekizumab, a humanized interleukin-17A antibody, has shown efficacy in ankylosing spondylitis (AS), with a greater proportion of ixekizumab-treated patients achieving an ASAS40 (Assessment of Spondyloarthritis International Society 40) endpoint compared to placebo. An ASAS40 response is a high standard that is not routinely used in clinical practice. The goals of this study were (a) to measure improvement in ixekizumab-treated patients in the four ASAS treatment response domains and in other patient-reported outcomes, and (b) to determine how the ASAS response was associated with changes in spinal pain at night, fatigue, sleep, and the Short Form 36-Item Physical Component Summary (SF-36 PCS).

Methods: The COAST-V and COAST-W trials were randomized, double-blind, controlled trials examining ixekizumab efficacy in patients with AS who were biologic disease-modifying antirheumatic drug (bDMARD)-naïve and tumor necrosis factor inhibitor (TNFi)-experienced, respectively. Data for the ASAS treatment response domains and other outcomes were collected through 16 weeks. Comparisons between treatment groups were made using a mixed-effects model for repeated measures. To determine how the ASAS response was associated with the changes in spinal pain at night, fatigue, sleep, and SF-36 PCS, comparisons were made between patient groups according to their level of treatment response (ASAS40 vs. ASAS20 vs. ASAS20 nonresponse) using analysis of covariance.

Results: Compared with placebo, patients treated with ixekizumab reported significantly greater improvement in the four ASAS treatment response domains and other outcomes (p < 0.05). Results were consistent for bDMARD-naïve and TNFi-experienced patients. Compared to ASAS20 nonresponders, patients who achieved ASAS40 reported significantly greater mean changes in spinal pain at night (1.0 vs. 5.1 for bDMARD-naïve; 0.5 vs. 5.4 for TNFi-experienced), fatigue (0.6 vs. 3.8 for bDMARD-naïve; 0.2 vs. 3.9 for TNFi-experienced), sleep quality (1.1 vs. 4.0 for bDMARD-naïve; 0.8 vs. 4.9 for TNFi-experienced), and SF-36 PCS (2.6 vs. 11.6 for bDMARD-naïve; 1.2 vs. 12.6 for TNFi-experienced) (p < 0.0001).

Conclusion: Patients with AS who were treated with ixekizumab reported greater improvements in multiple patient-reported outcomes than patients who received placebo. Importantly, achieving ASAS40 was associated with a 2.6-fold to 5.3-fold greater improvement in pain, fatigue, sleep, and quality of life for bDMARD-naïve patients, and a 5.1-fold to 18.5-fold greater improvement for TNFi-experienced patients, compared to ASAS20 nonresponders.

Trial registration: ClinicalTrials.gov identifiers: NCT02696785 and NCT02696798.

Funding: Eli Lilly and Company.

Keywords: ASAS40; Ankylosing spondylitis; Axial spondyloarthritis; Fatigue; Inflammation; Ixekizumab; Pain; Sleep.

Figures

Fig. 1
Fig. 1
Least-squares mean (LSM) changes from baseline in the ASAS treatment response domains of a, b patient global, c, d spinal pain, e, f inflammation (BASDAI Q5 and Q6), and g function (BASFI) for a, c, e, g bDMARD-naïve patients (COAST-V) and b, d, f TNFi-experienced patients (COAST-W). Comparisons with PBO were made using a mixed-effects model for repeated measures. ***p < 0.001 vs. PBO; **p < 0.01 vs. PBO. The BASFI data for TNFi-experienced patients (COAST-W) were published previously [13]. ADA adalimumab, ASAS Assessment of Spondyloarthritis International Society, BASDAI Bath Ankylosing Spondylitis Disease Activity Index, BASFI Bath Ankylosing Spondylitis Functional Index, bDMARD biologic disease-modifying antirheumatic drugs, IXE ixekizumab, PBO placebo, Q2W, every 2 weeks, Q4W every 4 weeks, TNFi tumor necrosis factor inhibitor
Fig. 2
Fig. 2
Least-squares mean (LSM) changes from baseline in patient-reported outcomes of a, b spinal pain at night, c, d fatigue measured by BASDAI Q1, e, f fatigue measured by NRS, and g, h sleep quality measured by JSEQ for a, c, e, g bDMARD-naïve patients (COAST-V) and b, d, f, h TNFi-experienced patients (COAST-W). Comparisons with PBO were made using a mixed-effects model for repeated measures. ***p < 0.001 vs. PBO; **p < 0.01 vs. PBO; *p < 0.05 vs. PBO. ADA adalimumab, BASDAI Bath Ankylosing Spondylitis Disease Activity Index, bDMARD biologic disease-modifying antirheumatic drugs, IXE ixekizumab, JSEQ Jenkins Sleep Evaluation Questionnaire, NRS Numeric Rating Scale, PBO placebo, Q2W, every 2 weeks, Q4W every 4 weeks, TNFi tumor necrosis factor inhibitor
Fig. 3
Fig. 3
Least-squares mean (LSM) changes from baseline in a Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) and b Medical Outcomes Study 36-Item Short-Form Health Survey Physical Component Summary (SF-36 PCS) in bDMARD-naïve patients (COAST-V). Comparisons with PBO were made using a mixed-effects model for repeated measures. ***p < 0.001 vs. PBO; **p < 0.01 vs. PBO. BASDAI and SF-36 PCS data for TNFi-experienced patients (COAST-W) were published previously [13]. ADA adalimumab, bDMARD biologic disease-modifying antirheumatic drug, IXE ixekizumab, PBO placebo, Q2W every 2 weeks, Q4W every 4 weeks, TNFi tumor necrosis factor inhibitor
Fig. 4
Fig. 4
Association between ASAS20 and ASAS40 and LSM improvement from baseline for a spinal pain at night, b fatigue (NRS), c sleep quality (JSEQ), and d SF-36 PCS at week 16. Treatment groups, including the placebo and adalimumab arms, were pooled and separated by ASAS20 and ASAS40 achievement. Black bars indicate data from bDMARD-naïve patients (COAST-V); gray bars indicate data from TNFi-experienced patients (COAST-W). §p ≤ 0.001, achieved ASAS20 but not ASAS40 vs. ASAS20 not achieved; *p < 0.0001, ASAS40 achieved vs. ASAS20 not achieved; †p < 0.0001, ASAS40 achieved vs. achieved ASAS20 but not ASAS40. Results were compared using ANCOVA after correcting for baseline value, age, and gender. Missing data were imputed using nonresponder imputation for the ASAS treatment response criteria and using modified baseline observation carried forward for the outcomes at week 16. Numbers shown are LSM improvements from baseline. Fold difference = [(ASAS40 achieved/ASAS20 not achieved) − 1]. ANCOVA analysis of covariance, ASAS Assessment of Spondyloarthritis International Society, bDMARD biologic disease-modifying antirheumatic drug, JSEQ Jenkins Sleep Evaluation Questionnaire, LSM least-squares mean, n, number of non-missing observations, NRS Numeric Rating Scale, SF-36 PCS Medical Outcomes Study 36-Item Short-Form Health Survey Physical Component Summary, TNFi tumor necrosis factor inhibitor

References

    1. Reveille JD. Epidemiology of spondyloarthritis in North America. Am J Med Sci. 2011;341(4):284–286. doi: 10.1097/MAJ.0b013e31820f8c99.
    1. Gran JT, Husby G, Hordvik M. Prevalence of ankylosing spondylitis in males and females in a young middle-aged population of Tromso, Northern Norway. Ann Rheum Dis. 1985;44(6):359–367. doi: 10.1136/ard.44.6.359.
    1. Rudwaleit M, van der Heijde D, Landewe R, et al. The development of Assessment of SpondyloArthritis international Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann Rheum Dis. 2009;68(6):777–783. doi: 10.1136/ard.2009.108233.
    1. Strand V, Singh JA. Patient burden of axial spondyloarthritis. J Clin Rheumatol. 2017;23(7):383–391. doi: 10.1097/RHU.0000000000000589.
    1. van der Heijde D, Ramiro S, Landewe R, et al. 2016 Update of the ASAS-EULAR management recommendations for axial spondyloarthritis. Ann Rheum Dis. 2017;76(6):978–91.
    1. Glintborg B, Ostergaard M, Krogh NS, Dreyer L, Kristensen HL, Hetland ML. Predictors of treatment response and drug continuation in 842 patients with ankylosing spondylitis treated with anti-tumour necrosis factor: results from 8 years’ surveillance in the Danish nationwide DANBIO registry. Ann Rheum Dis. 2010;69(11):2002–2008. doi: 10.1136/ard.2009.124446.
    1. Heiberg MS, Koldingsnes W, Mikkelsen K, et al. The comparative one-year performance of anti–tumor necrosis factor alpha drugs in patients with rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis: results from a longitudinal, observational, multicenter study. Arthritis Rheum. 2008;59(2):234–240. doi: 10.1002/art.23333.
    1. Inman RD, Davis JC, Jr, Heijde D, et al. Efficacy and safety of golimumab in patients with ankylosing spondylitis: results of a randomized, double-blind, placebo-controlled, phase III trial. Arthritis Rheum. 2008;58(11):3402–3412. doi: 10.1002/art.23969.
    1. van der Heijde D, Kivitz A, Schiff MH, 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(7):2136–2146. doi: 10.1002/art.21913.
    1. Dougados M, Baeten D. Spondyloarthritis. Lancet. 2011;377(9783):2127–2137. doi: 10.1016/S0140-6736(11)60071-8.
    1. Yeremenko N, Paramarta JE, Baeten D. The interleukin-23/interleukin-17 immune axis as a promising new target in the treatment of spondyloarthritis. Curr Opin Rheumatol. 2014;26(4):361–370. doi: 10.1097/BOR.0000000000000069.
    1. Liu L, Lu J, Allan BW, et al. Generation and characterization of ixekizumab, a humanized monoclonal antibody that neutralizes interleukin-17A. J Inflamm Res. 2016;9:39–50. doi: 10.2147/JIR.S100940.
    1. Deodhar A, Poddubnyy D, Pacheco-Tena C, et al. Efficacy and safety of ixekizumab in the treatment of radiographic axial spondyloarthritis: sixteen-week results of a phase III randomized, double-blind, placebo-controlled trial in patients with prior inadequate response or intolerance to tumor necrosis factor inhibitors. Arthritis Rheumatol. 2019;71(4):599–611. doi: 10.1002/art.40753.
    1. van der Heijde D, Cheng-Chung Wei J, Dougados M, et al. Ixekizumab, an interleukin-17A antagonist in the treatment of ankylosing spondylitis or radiographic axial spondyloarthritis in patients previously untreated with biological disease-modifying anti-rheumatic drugs (COAST-V): 16 week results of a phase 3 randomised, double-blind, active-controlled and placebo-controlled trial. Lancet. 2018;392(10163):2441–51.
    1. Sieper J, Rudwaleit M, Baraliakos X, et al. The Assessment of SpondyloArthritis international Society (ASAS) handbook: a guide to assess spondyloarthritis. Ann Rheum Dis. 2009;68(Suppl 2):ii1–44.v.
    1. Baeten D, Sieper J, Braun J, et al. Secukinumab, an interleukin-17A inhibitor, in ankylosing spondylitis. N Engl J Med. 2015;373(26):2534–2548. doi: 10.1056/NEJMoa1505066.
    1. Davis JC, van der Heijde DM, Braun J, et al. Sustained durability and tolerability of etanercept in ankylosing spondylitis for 96 weeks. Ann Rheum Dis. 2005;64(11):1557–1562. doi: 10.1136/ard.2004.035105.
    1. Lambert RG, Salonen D, Rahman P, et al. Adalimumab significantly reduces both spinal and sacroiliac joint inflammation in patients with ankylosing spondylitis: a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 2007;56(12):4005–4014. doi: 10.1002/art.23044.
    1. van der Heijde D, Dougados M, Landewe R, et al. Sustained efficacy, safety and patient-reported outcomes of certolizumab pegol in axial spondyloarthritis: 4-year outcomes from RAPID-axSpA. Rheumatology (Oxford). 2017;56(9):1498–509.
    1. van der Linden S, Valkenburg HA, Cats A. Evaluation of diagnostic criteria for ankylosing spondylitis. A proposal for modification of the New York criteria. Arthritis Rheum. 1984;27(4):361–368. doi: 10.1002/art.1780270401.
    1. Machado PM, Landewé R, van der Heijde D. Ankylosing Spondylitis Disease Activity Score (ASDAS): 2018 update of the nomenclature for disease activity states. Ann Rheum Dis. 2018;77:1539–1540.
    1. Naegeli AN, Flood E, Tucker J, Devlen J, Edson-Heredia E. The patient experience with fatigue and content validity of a measure to assess fatigue severity: qualitative research in patients with ankylosing spondylitis (AS) Health Qual Life Outcomes. 2013;11:192. doi: 10.1186/1477-7525-11-192.
    1. Deodhar A, Braun J, Inman RD, et al. Golimumab reduces sleep disturbance in patients with active ankylosing spondylitis: results from a randomized, placebo-controlled trial. Arthritis Care Res (Hoboken). 2010;62(9):1266–1271. doi: 10.1002/acr.20233.
    1. Helman CG. Disease versus illness in general practice. J R Coll Gen Pract. 1981;31(230):548–552.

Source: PubMed

3
Abonnere