Relationships between psoriatic arthritis composite measures of disease activity with patient-reported outcomes in phase 3 studies of tofacitinib

Laura C Coates, Andrew G Bushmakin, Oliver FitzGerald, Dafna D Gladman, Lara Fallon, Joseph C Cappelleri, Ming-Ann Hsu, Philip S Helliwell, Laura C Coates, Andrew G Bushmakin, Oliver FitzGerald, Dafna D Gladman, Lara Fallon, Joseph C Cappelleri, Ming-Ann Hsu, Philip S Helliwell

Abstract

Background: In psoriatic arthritis (PsA), further understanding of the relationships between clinical measures and patient-reported outcomes (PROs) is needed. This post hoc analysis evaluated associations between minimal disease activity (MDA) as a continuous outcome (termed ScoreMDA) or Psoriatic Arthritis Disease Activity Score (PASDAS) with selected PROs not included in the composite measures.

Methods: Data from two phase 3 studies of tofacitinib in PsA (OPAL Broaden [NCT01877668; N = 422]; OPAL Beyond [NCT01882439; N = 394]) were included. MDA (binary outcome) was defined as meeting ≥5/7 criteria. For ScoreMDA, each criterion was assigned a value (1 = true; 0 = false; score range, 0-7; scores ≥5 indicated MDA). For PASDAS (score range, 0-10), higher scores indicated worse disease activity. PROs analyzed included Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), Patient's Assessment of Arthritis Pain visual analog scale (Pain VAS), and EuroQoL-Five Dimensions-Three Level Health Questionnaire visual analog scale (EQ-5D-3L VAS) and utility index. Relationships were evaluated using repeated measures regression models.

Results: Similar, approximately linear relationships were confirmed between PASDAS or ScoreMDA and PROs in both studies. In OPAL Broaden and OPAL Beyond, a one-point difference in PASDAS was associated with clinically relevant differences in PROs, including EQ-5D-3L VAS (- 6.7 mm, - 6.9 mm), Pain VAS (9.9 mm, 10.7 mm), and FACIT-F (- 2.8, - 3.3). A one-point difference in ScoreMDA was associated with clinically relevant differences in PROs, including EQ-5D-3L VAS (5.0 mm, 5.5 mm) and FACIT-F (1.9, 2.7) in OPAL Broaden and OPAL Beyond, respectively.

Conclusions: Linear associations between PASDAS or ScoreMDA and PROs provide interpretable and quantifiable metrics between composite clinical measures and PROs, highlighting the importance of these measures in understanding the relevance of treat-to-target goals in PsA.

Trial registration: ClinicalTrials.gov, NCT01877668 . Registered on June 12, 2013. ClinicalTrials.gov, NCT01882439 . Registered on June 18, 2013.

Keywords: Minimal disease activity; Patient-reported outcomes; Psoriatic arthritis.

Conflict of interest statement

LCC is funded by a National Institute for Health Research (NIHR) Clinician Scientist award. The research was supported by the NIHR Oxford Biomedical Research Centre. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR, or the Department of Health. LCC has also received honoraria and/or research funding from AbbVie, Amgen, Biogen, Celgene, Eli Lilly, Galapagos, Gilead, Janssen, Novartis, Pfizer Inc, and UCB. OF has received research grants from AbbVie, BMS, Eli Lilly, Novartis, and Pfizer Inc; is a consultant for BMS, Celgene, Eli Lilly, Janssen, and Pfizer Inc; and has been on speaker bureaus for AbbVie, Janssen, and Pfizer Inc. DDG has received research grants from AbbVie, Amgen, Celgene, Eli Lilly, Novartis, Pfizer Inc, and UCB, and is a consultant for AbbVie, Amgen, BMS, Celgene, Eli Lilly, Galapagos, Gilead, Janssen, Novartis, Pfizer Inc, and UCB. AGB, LF, JCC, and M-AH are stockholders and employees of Pfizer Inc. PSH has received research grants from AbbVie, Janssen, and Novartis, and is a consultant for AbbVie, Amgen, Celgene, Galapagos, Pfizer Inc, and UCB.

Figures

Fig. 1
Fig. 1
Estimated relationship between PASDAS/Pain VAS (a) and ScoreMDA/SF-36v2 PCS (b). Data from OPAL Broaden (N = 422). SF-36v2 PCS: norm-based scores were used (a score of 50 representing the mean for the general population, with higher scores indicating less impairment); ScoreMDA: continuous MDA with values from 0 to 7 (0–4, no MDA; 5–7, MDA). Arrows indicate the direction of improvement. MDA, minimal disease activity; Pain VAS, Patient’s Assessment of Arthritis Pain visual analog scale; PASDAS, Psoriatic Arthritis Disease Activity Score; PCS, Physical Component Summary; SF-36v2, Short Form-36 Health Survey Version 2, acute
Fig. 2
Fig. 2
Estimated relationships between PASDAS and FACIT-F total score (a), Pain VAS (b), and HAQ-DI (c). Data from OPAL Broaden (N = 422) and OPAL Beyond (N = 394); all treatment groups from each phase 3 study were pooled for analysis. Arrows indicate the direction of improvement. FACIT-F, Functional Assessment of Chronic Illness Therapy-Fatigue; HAQ-DI, Health Assessment Questionnaire-Disability Index; Pain VAS, Patient’s Assessment of Arthritis Pain visual analog scale; PASDAS, Psoriatic Arthritis Disease Activity Score; VAS visual analog scale
Fig. 3
Fig. 3
Estimated relationships between PASDAS and EQ-5D-3L VAS (a) and EQ-5D-3L UI (b). Data from OPAL Broaden (N = 422) and OPAL Beyond (N = 394); all treatment groups from each phase 3 study were pooled for analysis. Arrows indicate the direction of improvement. EQ-5D-3L, EuroQoL-Five Dimensions-Three Level Health Questionnaire; PASDAS, Psoriatic Arthritis Disease Activity Score; UI, utility index; VAS, visual analog scale
Fig. 4
Fig. 4
Estimated relationships between ScoreMDA and FACIT-F total score (a), SF-36v2 PCS (b), EQ-5D-3L VAS (c), and EQ-5D-3L UI (d). Data from OPAL Broaden (N = 422) and OPAL Beyond (N = 394); all treatment groups from each phase 3 study were pooled for analysis. SF-36v2 PCS: norm-based scores were used (a score of 50 representing the mean for the general population, with higher scores indicating less impairment); ScoreMDA: continuous MDA with values from 0 to 7 (0–4, no MDA; 5–7, MDA). EQ-5D-3L, EuroQoL-Five Dimensions-Three Level Health Questionnaire; FACIT-F, Functional Assessment of Chronic Illness Therapy-Fatigue; MDA, minimal disease activity; PCS, Physical Component Summary; SF-36v2, Short Form-36 Health Survey Version 2, acute; UI, utility index, VAS, visual analog scale

References

    1. Gladman DD, Antoni C, Mease P, Clegg DO, Nash P. Psoriatic arthritis: epidemiology, clinical features, course, and outcome. Ann Rheum Dis. 2005;64:ii14–ii17.
    1. Ogdie A, Weiss P. The epidemiology of psoriatic arthritis. Rheum Dis Clin North Am. 2015;41(4):545–68. 10.1016/j.rdc.2015.07.001.
    1. Ritchlin CT, Colbert RA, Gladman DD. Psoriatic arthritis. N Engl J Med. 2017;376(10):957–970. doi: 10.1056/NEJMra1505557.
    1. Mease P, Strand V, Gladman D. Functional impairment measurement in psoriatic arthritis: importance and challenges. Semin Arthritis Rheum. 2018;48(3):436–448. doi: 10.1016/j.semarthrit.2018.05.010.
    1. Gudu T, Gossec L. Quality of life in psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(5):405–417. doi: 10.1080/1744666X.2018.1468252.
    1. Helliwell P, Coates LC, FitzGerald O, Nash P, Soriano ER, Husni ME, et al. Disease-specific composite measures for psoriatic arthritis are highly responsive to a Janus kinase inhibitor treatment that targets multiple domains of disease. Arthritis Res Ther. 2018;20(1):242. doi: 10.1186/s13075-018-1739-0.
    1. Coates LC, Fransen J, Helliwell PS. Defining minimal disease activity in psoriatic arthritis: a proposed objective target for treatment. Ann Rheum Dis. 2010;69(01):48–53. doi: 10.1136/ard.2008.102053.
    1. Gossec L, McGonagle D, Korotaeva T, Lubrano E, de Miguel E, Østergaard M, et al. Minimal disease activity as a treatment target in psoriatic arthritis: a review of the literature. J Rheumatol. 2018;45(1):6–13. 10.3899/jrheum.170449.
    1. Helliwell PS, FitzGerald O, Fransen J, Gladman DD, Kreuger GG, Callis-Duffin K, et al. The development of candidate composite disease activity and responder indices for psoriatic arthritis (GRACE project). Ann Rheum Dis. 2013;72(6):986–91. 10.1136/annrheumdis-2012-201341.
    1. Orbai A-M, Ogdie A. Patient reported outcomes in psoriatic arthritis. Rheum Dis Clin North Am. 2016;42(2):265–83. 10.1016/j.rdc.2016.01.002.
    1. Ware JE, Kosinski M, Dewey JE. How to score version two of the SF-36 Health Survey. Lincoln: QualityMetric Incorporated; 2000.
    1. Cauli A, Gladman DD, Mathieu A, Olivieri I, Porru G, Tak PP, et al. Patient global assessment in psoriatic arthritis: a multicenter GRAPPA and OMERACT study. J Rheumatol. 2011;38(5):898–903. doi: 10.3899/jrheum.100857.
    1. Fries JF, Spitz PW, Young DY. The dimensions of health outcomes: the health assessment questionnaire, disability and pain scales. J Rheumatol. 1982;9(5):789–793.
    1. Chandran V, Bhella S, Schentag C, Gladman DD. Functional assessment of chronic illness therapy-fatigue scale is valid in patients with psoriatic arthritis. Ann Rheum Dis. 2007;66(7):936–939. doi: 10.1136/ard.2006.065763.
    1. EuroQoL Group. EuroQol--a new facility for the measurement of health-related quality of life. Health Policy. 1990;16(3):199–208.
    1. Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377(16):1537–50. 10.1056/NEJMoa1615975.
    1. Gladman D, Rigby W, Azevedo VF, Behrens F, Blanco R, Kaszuba A, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–36. 10.1056/NEJMoa1615977.
    1. Nash P, Coates LC, Kivitz AJ, Mease PJ, Gladman DD, Covarrubias-Cobos JA, et al. Safety and efficacy of tofacitinib in patients with active psoriatic arthritis: interim analysis of OPAL Balance, an open-label, long-term extension study. Rheumatol Ther. 2020;7(3):553–80. 10.1007/s40744-020-00209-4.
    1. Strand V, de Vlam K, Covarrubias-Cobos JA, Mease PJ, Gladman DD, Graham D, et al. Tofacitinib or adalimumab versus placebo: patient-reported outcomes from OPAL Broaden—a phase III study of active psoriatic arthritis in patients with an inadequate response to conventional synthetic disease-modifying antirheumatic drugs. RMD Open. 2019;5(1):e000806. 10.1136/rmdopen-2018-000806.
    1. Strand V, de Vlam K, Covarrubias-Cobos JA, Mease PJ, Gladman DD, Chen L, et al. Effect of tofacitinib on patient-reported outcomes in patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors in the phase III, randomised controlled trial: OPAL Beyond. RMD Open. 2019;5(1):e000808. 10.1136/rmdopen-2018-000808.
    1. Taylor W, Gladman D, Helliwell P, Marchesoni A, Mease P, Mielants H. Classification criteria for psoriatic arthritis: development of new criteria from a large international study. Arthritis Rheum. 2006;54(8):2665–2673. doi: 10.1002/art.21972.
    1. Helliwell PS, Kavanaugh A. Comparison of composite measures of disease activity in psoriatic arthritis using data from an interventional study with golimumab. Arthritis Care Res (Hoboken) 2014;66(5):749–756. doi: 10.1002/acr.22204.
    1. Helliwell PS, FitzGerald O, Fransen J. Composite disease activity and responder indices for psoriatic arthritis: a report from the GRAPPA 2013 meeting on development of cutoffs for both disease activity states and response. J Rheumatol. 2014;41(6):1212–1217. doi: 10.3899/jrheum.140172.
    1. Coates LC, Helliwell PS. Defining low disease activity states in psoriatic arthritis using novel composite disease instruments. J Rheumatol. 2016;43(2):371–375. doi: 10.3899/jrheum.150826.
    1. Cappelleri JC, Zou KH, Bushmakin AG, Alvir JMJ, Alemayehu D, Symonds T Patient-reported outcomes: measurement, implementation and interpretation. New York: CRC Press; 2014.
    1. Fitzmaurice GM, Laird NM, Ware JH. Applied longitudinal analysis. 2nd ed. New Jersey: Wiley; 2011. 10.1002/9781119513469.
    1. Orbai AM, de Wit M, Mease PJ, Callis Duffin K, Elmamoun M, Tillett W, et al. Updating the psoriatic arthritis (PsA) core domain set: a report from the PsA Workshop at OMERACT 2016. J Rheumatol. 2017;44(10):1522–8. 10.3899/jrheum.160904.
    1. Coates LC, Gladman DD, Nash P, FitzGerald O, Kavanaugh A, Kvien TK, et al. Secukinumab provides sustained PASDAS-defined remission in psoriatic arthritis and improves health-related quality of life in patients achieving remission: 2-year results from the phase III FUTURE 2 study. Arthritis Res Ther. 2018;20(1):272. doi: 10.1186/s13075-018-1773-y.
    1. Coates LC, Nash P, Kvien TK, Gossec L, Mease PJ, Rasouliyan L, et al. Comparison of remission and low disease activity states with DAPSA, MDA and VLDA in a clinical trial setting in psoriatic arthritis patients: 2-year results from the FUTURE 2 study. Semin Arthritis Rheum. 2020;50(4):709–18. 10.1016/j.semarthrit.2020.03.015.
    1. Bojke L, Epstein D, Craig D, Rodgers M, Woolacott N, Yang H, et al. Modelling the cost-effectiveness of biologic treatments for psoriatic arthritis. Rheumatology (Oxford). 2011;50(Suppl 4):iv39–47.
    1. Whitehead SJ, Ali S. Health outcomes in economic evaluation: the QALY and utilities. Br Med Bull. 2010;96(1):5–21. doi: 10.1093/bmb/ldq033.
    1. Orbai A-M. Content validity of psoriatic arthritis composite indices: anchoring with the patient perspective and the core domain set. Rheumatology (Oxford) 2020;59(1):1–4. doi: 10.1093/rheumatology/kez372.
    1. Fei JZ, Perruccio AV, Ye JY, Gladman DD, Chandran V. The relationship between patient acceptable symptom state and disease activity in patients with psoriatic arthritis. Rheumatology (Oxford) 2020;59(1):69–76. doi: 10.1093/rheumatology/kez202.
    1. Coates LC, Orbai AM, Morita A, Benichou O, Kerr L, Adams DH, et al. Achieving minimal disease activity in psoriatic arthritis predicts meaningful improvements in patients’ health-related quality of life and productivity. BMC Rheumatol. 2018;2(1):24. 10.1186/s41927-018-0030-y.
    1. Cella D, Wilson H, Shalhoub H, Revicki DA, Cappelleri JC, Bushmakin AG, et al. Content validity and psychometric evaluation of Functional Assessment of Chronic Illness Therapy-Fatigue in patients with psoriatic arthritis. J Patient Rep Outcomes. 2019;3(1):30. 10.1186/s41687-019-0115-4.
    1. Mease PJ, Kavanaugh A, Coates LC, McInnes IB, Hojnik M, Zhang Y, et al. Prediction and benefits of minimal disease activity in patients with psoriatic arthritis and active skin disease in the ADEPT trial. RMD Open. 2017;3(1):e000415. doi: 10.1136/rmdopen-2016-000415.
    1. Queiro R, Cañete JD, Montilla C, Abad M, Montoro M, Gómez S, et al. Minimal disease activity and impact of disease in psoriatic arthritis: a Spanish cross-sectional multicenter study. Arthritis Res Ther. 2017;19(1):72. doi: 10.1186/s13075-017-1277-1.

Source: PubMed

3
Subskrybuj