Upadacitinib improves patient-reported outcomes in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying antirheumatic drugs: results from SELECT-NEXT

Vibeke Strand, Janet Pope, Namita Tundia, Alan Friedman, Heidi S Camp, Aileen Pangan, Arijit Ganguli, Mahesh Fuldeore, Debbie Goldschmidt, Michael Schiff, Vibeke Strand, Janet Pope, Namita Tundia, Alan Friedman, Heidi S Camp, Aileen Pangan, Arijit Ganguli, Mahesh Fuldeore, Debbie Goldschmidt, Michael Schiff

Abstract

Background: To evaluate the effect of upadacitinib on patient-reported outcomes (PROs) in patients with RA who had an inadequate response to csDMARDs.

Methods: Patients in SELECT-NEXT, a randomised controlled trial, were on a background of csDMARDs and received upadacitinib 15 mg and 30 mg or placebo daily for 12 weeks. PROs included Patient Global Assessment of Disease Activity (PtGA), pain, Health Assessment Questionnaire-Disability Index (HAQ-DI), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), duration and severity of morning (AM) joint stiffness, Short Form 36 Health Survey (SF-36), and Work Instability Scale for RA (RA-WIS). Least squares mean (LSM) changes were based on mixed-effect repeated measure models. Percentages of patients reporting improvements ≥ minimum clinically important differences (MCIDs) and scores ≥ normative values and number needed to treat (NNT) were determined; group comparisons used chi-square tests.

Results: Data from 661 patients were analysed. Compared with placebo, patients receiving upadacitinib reported statistically significant improvements (both doses, P < 0.05) in PtGA, pain, HAQ-DI, FACIT-F, duration and severity of AM stiffness, SF-36 (PCS and 6/8 domains), and RA-WIS at week 12. Significantly, more upadacitinib-treated patients (both doses, P < 0.05) reported improvements ≥ MCID in PtGA, pain, HAQ-DI, FACIT-F, AM stiffness, SF-36 (PCS and 4 or 7/8 domains), and RA-WIS and scores ≥ normative values in HAQ-DI, FACIT-F, and SF-36 (PCS and 4 or 5/8 domains). For most PROs, the incremental NNT with upadacitinib to report clinically meaningful improvement from baseline ranged from 4 to 8 patients.

Conclusions: Upadacitinib 15 mg or 30 mg daily for 12 weeks resulted in significant and clinically meaningful improvements in global disease activity, pain, physical function, fatigue, duration and severity of AM stiffness, HRQOL, and work instability among csDMARD-IR patients with RA.

Trial registration: Clinicaltrials.gov, NCT02675426. Retrospectively registered 5 February 2016.

Keywords: Health-related quality of life; JAK inhibitor; Patient-reported outcomes; Rheumatoid arthritis; Treatment outcomes.

Conflict of interest statement

VS is a consultant for AbbVie, Amgen, AstraZeneca, BMS, Celgene, Genentech, GSK, Janssen, Lilly, Novartis, Pfizer, Regeneron, Sanofi, and UCB, and is involved in the advisory boards for AbbVie, Amgen, AstraZeneca, BMS, Celgene, Genentech, GSK, Janssen, Lilly, Novartis, Pfizer, Regeneron, Sanofi, and UCB. JP is a consultant for AbbVie, Amgen, BMS, Celltrion, GSK, Lilly, Merck, Novartis, Pfizer, Roche, Sandoz, Sanofi, and UCB. NT, AF, HSC, AP, AG, and MF are employees of AbbVie and may own AbbVie stock or stock options. DG is an employee of Analysis Group, Inc., which received consulting fees from AbbVie for this study. MS is a consultant for AbbVie, BMS, Eli Lilly, JNJ, and UCB, and is a member of the speaker bureau for AbbVie and BMS.

Figures

Fig. 1
Fig. 1
Baseline and post-treatment scores at week 12 across all Short Form 36 Health Survey domains. Baseline (BL) and SF-36 domain scores are relative to age- and gender-adjusted norms (A/G norms) for the general US population. a PBO. b UPA 15 mg. c UPA 30 mg. d Combined. In the combined spydergrams, most of the UPA 30-mg results are covered up by the UPA 15-mg results. BL values and SF-36 domain scores were re-scored from 0 to 100. No further transformations were applied for this analysis. BP, Bodily Pain; GH, General Health; MH, Mental Health; PBO, placebo; PF, Physical Functioning; RE, Role-Emotional; RP, Role-Physical; SF, Social Functioning; UPA, upadacitinib; VT, Vitality; Wk, week
Fig. 2
Fig. 2
Percentage of patients reporting improvements ≥ MCID at week 12. a Patient’s Global Assessment of Disease Activity (PtGA), pain, Health Assessment Questionnaire-Disability Index (HAQ-DI), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), morning (AM) joint stiffness duration, AM stiffness severity, Work Instability Scale for RA (RA-WIS), and Short Form 36 Health Survey (SF-36). b SF-36 individual domains. Baseline values and SF-36 domains were re-scored from 0 to 100. ***P < 0.001, **P < 0.01, *P < 0.5 for upadacitinib versus placebo. BP, Bodily Pain; GH, General Health; MCID, minimum clinically important difference; MCS, mental component summary; MH, Mental Health; NNT, number needed to treat; PBO, placebo; PCS, physical component summary; PF, Physical Functioning; RE, Role-Emotional; RP, Role-Physical; SF, Social Functioning; UPA, upadacitinib; VAS, visual analogue scale; VT, Vitality
Fig. 3
Fig. 3
Patients reporting scores ≥ normative values at baseline and week 12. a Health Assessment Questionnaire-Disability Index (HAQ-DI), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), Work Instability Scale for RA (RA-WIS), and Short Form 36 Health Survey (SF-36) PCS and MCS. b SF-36 individual domains. Baseline values and SF-36 domains were re-scored from 0 to 100. ***P < 0.001, **P < 0.01, *P < 0.5 for upadacitinib versus placebo. BL, baseline; BP, Bodily Pain; GH, General Health; MCS, mental component summary; MH, Mental Health; PBO, placebo; PCS, physical component summary; PF, Physical Functioning; RE, Role-Emotional; RP, Role-Physical; SF, Social Functioning; UPA, upadacitinib; VT, Vitality; Wk, week

References

    1. Smolen JS, Aletaha D, McInnes IB. Rheumatoid arthritis. Lancet. 2016;388:2023–2038. doi: 10.1016/S0140-6736(16)30173-8.
    1. Hewlett S, Cockshott Z, Byron M, Kitchen K, Tipler S, Pope D, et al. Patients’ perceptions of fatigue in rheumatoid arthritis: overwhelming, uncontrollable, ignored. Arthritis Rheum. 2005;53:697–702. doi: 10.1002/art.21450.
    1. Kirwan JR, Hewlett S. Patient perspective: reasons and methods for measuring fatigue in rheumatoid arthritis. J Rheumatol. 2007;34:1171–1173.
    1. da Silva JA, Phillips S, Buttgereit F. Impact of impaired morning function on the lives and well-being of patients with rheumatoid arthritis. Scand J Rheumatol Suppl. 2011;125:6–11. doi: 10.3109/03009742.2011.566434.
    1. Halls S, Dures E, Kirwan J, Pollock J, Baker G, Edmunds A, et al. Stiffness is more than just duration and severity: a qualitative exploration in people with rheumatoid arthritis. Rheumatology (Oxford) 2015;54:615–622. doi: 10.1093/rheumatology/keu379.
    1. Pollard LC, Choy EH, Gonzalez J, Khoshaba B, Scott DL. Fatigue in rheumatoid arthritis reflects pain, not disease activity. Rheumatology (Oxford) 2006;45:885–889. doi: 10.1093/rheumatology/kel021.
    1. Matcham F, Scott IC, Rayner L, Hotopf M, Kingsley GH, Norton S, et al. The impact of rheumatoid arthritis on quality-of-life assessed using the SF-36: a systematic review and meta-analysis. Semin Arthritis Rheum. 2014;44:123–130. doi: 10.1016/j.semarthrit.2014.05.001.
    1. Strand V, Khanna D. The impact of rheumatoid arthritis and treatment on patients’ lives. Clin Exp Rheumatol. 2010;28:S32–S40.
    1. Taylor PC, Moore A, Vasilescu R, Alvir J, Tarallo M. A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: a current perspective. Rheumatol Int. 2016;36:685–695. doi: 10.1007/s00296-015-3415-x.
    1. Tang K, Beaton DE, Gignac MA, Lacaille D, Zhang W, Bombardier C. The Work Instability Scale for rheumatoid arthritis predicts arthritis-related work transitions within 12 months. Arthritis Care Res (Hoboken) 2010;62:1578–1587. doi: 10.1002/acr.20272.
    1. Sokka T, Kautiainen H, Pincus T, Verstappen SM, Aggarwal A, Alten R, et al. Work disability remains a major problem in rheumatoid arthritis in the 2000s: data from 32 countries in the QUEST-RA study. Arthritis Res Ther. 2010;12:R42. doi: 10.1186/ar2951.
    1. Westhoff G, Buttgereit F, Gromnica-Ihle E, Zink A. Morning stiffness and its influence on early retirement in patients with recent onset rheumatoid arthritis. Rheumatology (Oxford) 2008;47:980–984. doi: 10.1093/rheumatology/ken137.
    1. van Tuyl LH, Sadlonova M, Hewlett S, Davis B, Flurey C, Goel N, et al. The patient perspective on absence of disease activity in rheumatoid arthritis: a survey to identify key domains of patient-perceived remission. Ann Rheum Dis. 2017;76:855–861. doi: 10.1136/annrheumdis-2016-209835.
    1. Nikiphorou E, Radner H, Chatzidionysiou K, Desthieux C, Zabalan C, van Eijk-Hustings Y, et al. Patient global assessment in measuring disease activity in rheumatoid arthritis: a review of the literature. Arthritis Res Ther. 2016;18:251. doi: 10.1186/s13075-016-1151-6.
    1. Orbai AM, Bingham CO., 3rd Patient reported outcomes in rheumatoid arthritis clinical trials. Curr Rheumatol Rep. 2015;17:28. doi: 10.1007/s11926-015-0501-8.
    1. Boers M, Kirwan JR, Wells G, Beaton D, Gossec L, d’Agostino MA, et al. Developing core outcome measurement sets for clinical trials: OMERACT filter 2.0. J Clin Epidemiol. 2014;67:745–753. doi: 10.1016/j.jclinepi.2013.11.013.
    1. Kirwan JR, Tugwell PS. Overview of the patient perspective at OMERACT 10--conceptualizing methods for developing patient-reported outcomes. J Rheumatol. 2011;38:1699–1701. doi: 10.3899/jrheum.110388.
    1. Sanderson T, Morris M, Calnan M, Richards P, Hewlett S. What outcomes from pharmacologic treatments are important to people with rheumatoid arthritis? Creating the basis of a patient core set. Arthritis Care Res (Hoboken) 2010;62:640–646. doi: 10.1002/acr.20034.
    1. Fautrel B, Alten R, Kirkham B, de la Torre I, Durand F, Barry J, et al. Call for action: how to improve use of patient-reported outcomes to guide clinical decision making in rheumatoid arthritis. Rheumatol Int. 2018;38:935–947. doi: 10.1007/s00296-018-4005-5.
    1. Burmester GR, Blanco R, Charles-Schoeman C, Wollenhaupt J, Zerbini C, Benda B, et al. Tofacitinib (CP-690,550) in combination with methotrexate in patients with active rheumatoid arthritis with an inadequate response to tumour necrosis factor inhibitors: a randomised phase 3 trial. Lancet. 2013;381:451–460. doi: 10.1016/S0140-6736(12)61424-X.
    1. Genovese MC, Kremer J, Zamani O, Ludivico C, Krogulec M, Xie L, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016;374:1243–1252. doi: 10.1056/NEJMoa1507247.
    1. Kavanaugh A, Kremer J, Ponce L, Cseuz R, Reshetko OV, Stanislavchuk M, et al. Filgotinib (GLPG0634/GS-6034), an oral selective JAK1 inhibitor, is effective as monotherapy in patients with active rheumatoid arthritis: results from a randomised, dose-finding study (DARWIN 2) Ann Rheum Dis. 2017;76:1009–1019. doi: 10.1136/annrheumdis-2016-210105.
    1. Westhovens R, Taylor PC, Alten R, Pavlova D, Enriquez-Sosa F, Mazur M, et al. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1) Ann Rheum Dis. 2017;76:998–1008. doi: 10.1136/annrheumdis-2016-210104.
    1. Burmester GR, Kremer JM, Van den Bosch F, Kivitz A, Bessette L, Li Y, et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2018;391:2503–2512. doi: 10.1016/S0140-6736(18)31115-2.
    1. Genovese MC, Smolen JS, Weinblatt ME, Burmester GR, Meerwein S, Camp HS, et al. Efficacy and safety of ABT-494, a selective JAK-1 inhibitor, in a phase IIb study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol. 2016;68:2857–2866. doi: 10.1002/art.39808.
    1. Bruce B, Fries JF. The Health Assessment Questionnaire (HAQ) Clin Exp Rheumatol. 2005;23:S14–S18.
    1. Ramey D, Fries JF, Singh G. The Health Assessment Questionnaire 1995 – status and review. 2. Philadelphia: Lippincott-Raven; 1996.
    1. Kitchen H, Hansen B, Abetz L, Højbjerre L, Strandber-Larsen M. Patient-reported outcome measures for rheumatoid arthritis: minimal important differences review. Arthritis Rheum. 2013;65(Suppl 10):abstract 2268.
    1. Strand V, Boers M, Idzerda L, Kirwan JR, Kvien TK, Tugwell PS, et al. It’s good to feel better but it’s better to feel good and even better to feel good as soon as possible for as long as possible. Response criteria and the importance of change at OMERACT 10. J Rheumatol. 2011;38:1720–1727. doi: 10.3899/jrheum.110392.
    1. Krishnan E, Sokka T, Hakkinen A, Hubert H, Hannonen P. Normative values for the Health Assessment Questionnaire disability index: benchmarking disability in the general population. Arthritis Rheum. 2004;50:953–960. doi: 10.1002/art.20048.
    1. Cella D, Yount S, Sorensen M, Chartash E, Sengupta N, Grober J. Validation of the functional assessment of chronic illness therapy fatigue scale relative to other instrumentation in patients with rheumatoid arthritis. J Rheumatol. 2005;32:811–819.
    1. Kavanaugh A, Gladman DD, Edwards CJ, Poder A, Lioté F, Bird P, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, is associated with long-term (104-week) improvmeent in fatigue in patients with psoriatic arthritis: pooled results from 3 phase III, randomized, controlled trials. Ann Rheum Dis. 2016;75(Suppl 2):346.
    1. Ware JE, Jr, Sherbourne CD. The MOS 36-item Short-Form Health Survey (SF-36). I. Conceptual framework and item selection. Med Care. 1992;30:473–483. doi: 10.1097/00005650-199206000-00002.
    1. Brazier JE, Harper R, Jones NM, O’Cathain A, Thomas KJ, Usherwood T, et al. Validating the SF-36 health survey questionnaire: new outcome measure for primary care. BMJ. 1992;305:160–164. doi: 10.1136/bmj.305.6846.160.
    1. Herdman M, Gudex C, Lloyd A, Janssen M, Kind P, Parkin D, et al. Development and preliminary testing of the new five-level version of EQ-5D (EQ-5D-5L) Qual Life Res. 2011;20:1727–1736. doi: 10.1007/s11136-011-9903-x.
    1. Shaw JW, Johnson JA, Coons SJ. US valuation of the EQ-5D health states: development and testing of the D1 valuation model. Med Care. 2005;43:203–220. doi: 10.1097/00005650-200503000-00003.
    1. Gilworth G, Chamberlain MA, Harvey A, Woodhouse A, Smith J, Smyth MG, et al. Development of a work instability scale for rheumatoid arthritis. Arthritis Rheum. 2003;49:349–354. doi: 10.1002/art.11114.
    1. Revicki D, Cifaldi MA, Safikhani S, Chen N, Ganguli A. Responsiveness and minimal important difference of the rheumatoid arthritis-work instability scale (RA-WIS) Ann Rheum Dis. 2013;72:A867. doi: 10.1136/annrheumdis-2013-eular.2588.
    1. Ara R, Brazier J. Deriving an algorithm to convert the eight mean SF-36 dimension scores into a mean EQ-5D preference-based score from published studies (where patient level data are not available) Value Health. 2008;11:1131–1143. doi: 10.1111/j.1524-4733.2008.00352.x.
    1. Ara R, Brazier J. Predicting the short form-6D preference-based index using the eight mean short form-36 health dimension scores: estimating preference-based health-related utilities when patient level data are not available. Value Health. 2009;12:346–353. doi: 10.1111/j.1524-4733.2008.00428.x.
    1. Siwek J, Newman DH. Introducing medicine by the numbers: a collaboration of the NNT group and AFP. Am Fam Physician. 2015;91:434–435.
    1. Breedveld FC, Han C, Bala M, van der Heijde D, Baker D, Kavanaugh AF, et al. Association between baseline radiographic damage and improvement in physical function after treatment of patients with rheumatoid arthritis. Ann Rheum Dis. 2005;64:52–55. doi: 10.1136/ard.2003.017160.
    1. Wolfe F, Michaud K. Predicting depression in rheumatoid arthritis: the signal importance of pain extent and fatigue, and comorbidity. Arthritis Rheum. 2009;61:667–673. doi: 10.1002/art.24428.
    1. Lacaille D, White MA, Backman CL, Gignac MA. Problems faced at work due to inflammatory arthritis: new insights gained from understanding patients’ perspective. Arthritis Rheum. 2007;57:1269–1279. doi: 10.1002/art.23002.
    1. Reilly MC, Zbrozek AS, Dukes EM. The validity and reproducibility of a work productivity and activity impairment instrument. Pharmacoeconomics. 1993;4:353–365. doi: 10.2165/00019053-199304050-00006.
    1. Walter MJM, Kuijper TM, Hazes JMW, Weel AE, Luime JJ. Fatigue in early, intensively treated and tight-controlled rheumatoid arthritis patients is frequent and persistent: a prospective study. Rheumatol Int. 2018;38:1643–1650. doi: 10.1007/s00296-018-4102-5.
    1. van Steenbergen HW, Tsonaka R, Huizinga TW, Boonen A, van der Helm-van Mil AH. Fatigue in rheumatoid arthritis; a persistent problem: a large longitudinal study. RMD Open. 2015;1:e000041. doi: 10.1136/rmdopen-2014-000041.
    1. Nikolaus S, Bode C, Taal E, van de Laar MA. Fatigue and factors related to fatigue in rheumatoid arthritis: a systematic review. Arthritis Care Res (Hoboken) 2013;65:1128–1146. doi: 10.1002/acr.21949.

Source: PubMed

3
Abonneren