Associations between Patient Global Assessment scores and pain, physical function, and fatigue in rheumatoid arthritis: a post hoc analysis of data from phase 3 trials of tofacitinib

Vibeke Strand, Jeffrey Kaine, Rieke Alten, Gene Wallenstein, Annette Diehl, Harry Shi, Rebecca Germino, Christopher W Murray, Vibeke Strand, Jeffrey Kaine, Rieke Alten, Gene Wallenstein, Annette Diehl, Harry Shi, Rebecca Germino, Christopher W Murray

Abstract

Background: Tofacitinib is an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis (RA). We examined the degree to which Patient Global Assessment of Disease Activity (PtGA) was driven by patient-reported assessments of pain (Pain), physical function, and fatigue in patients receiving tofacitinib 5 mg twice daily or placebo, each with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs).

Methods: This post hoc analysis used data pooled from three randomized controlled trials in csDMARD-inadequate responder (csDMARD-IR) patients (ORAL Scan: NCT00847613; ORAL Standard: NCT00853385; ORAL Sync: NCT00856544). Using subgroup analysis from 2 × 2 tables, associations between PtGA and Pain, Health Assessment Questionnaire-Disability Index (HAQ-DI), and Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) at month 3 were evaluated using Pearson's Phi correlation coefficients. To support the main analysis, associations between select patient-reported outcomes (PROs) were also evaluated in csDMARD-naïve (ORAL Start; NCT01039688) and biologic (b)DMARD-IR (ORAL Step; NCT00960440) patients.

Results: Across csDMARD-IR treatment groups, low disease activity (defined as PtGA ≤ 20 mm), and moderate (≥ 30%) and substantial (≥ 50%) improvements from baseline in PtGA were associated with mild Pain (Visual Analog Scale score ≤ 20 mm), and moderate (≥ 30%) and substantial (≥ 50%) improvements from baseline in Pain; lack of Pain improvement was associated with little/no improvement in PtGA. In contrast, large proportions of csDMARD-IR patients who reported PtGA improvements did not report HAQ-DI or FACIT-F scores ≥ normative values (≤ 0.25 and ≥ 43.5, respectively) or changes in HAQ-DI or FACIT-F scores ≥ minimum clinically important difference (≥ 0.22 and ≥ 4.0, respectively). Generally, PtGA and Pain outcomes were moderately-to-strongly correlated at month 3 in csDMARD-IR patients, with weaker correlations evident between PtGA and HAQ-DI/FACIT-F outcomes. Similar findings were generally evident in csDMARD-naïve and bDMARD-IR patients.

Conclusions: This analysis supports the role of Pain as a key driver of PtGA in RA; physical function and fatigue play lesser roles in patients' perceptions of disease activity. These findings corroborate the importance of improved PROs and attainment of low symptom states for optimizing patient care.

Trial registration: Clinicaltrials.gov: NCT00847613 (registered: February 19, 2009); NCT00853385 (registered: March 2, 2009); NCT00856544 (registered: March 5, 2009); NCT01039688 (registered: December 25, 2009); NCT00960440 (registered: August 17, 2009).

Keywords: Disability; Fatigue; Pain; Patient Global Assessment; Patient-reported outcomes; Physical function; Rheumatoid arthritis; Tofacitinib.

Conflict of interest statement

VS has received consulting fees or other remuneration from AbbVie, Amgen, Bristol-Myers Squibb, Corrona, Eli Lilly, Janssen, Novartis, Pfizer Inc, and UCB.

JK has participated in speakers’ bureaus and has consulted for Pfizer Inc.

RA has received research grants and speakers’ honoraria from Pfizer Inc.

GW was an employee and shareholder of Pfizer Inc at the time of the analysis.

AD, HS, RG, and CWM are employees of Pfizer Inc and own stock or stock options in Pfizer Inc.

Figures

Fig. 1
Fig. 1
Associations between LDA and Pain, HAQ-DI, and FACIT-F outcomes at month 3 in csDMARD-IR patients. Proportions of csDMARD-IR patients at month 3 who did/did not report a mild Pain (VAS score ≤ 20 mm), b moderate improvements in Pain (≥ 30% decreases from baseline), c substantial improvements in Pain (≥ 50% decreases from baseline), d HAQ-DI scores ≥ normative values (≤ 0.25), or e FACIT-F scores ≥ normative values (≥ 43.5), stratified by LDA status (PtGA VAS score ≤ 20 mm). Denominators represent the number of patients who did/did not report LDA, respectively. Abbreviations: BID twice daily, CI confidence interval, csDMARD conventional synthetic disease-modifying antirheumatic drug, FACIT-F Functional Assessment of Chronic Illness Therapy-Fatigue, HAQ-DI Health Assessment Questionnaire-Disability Index, IR inadequate responder, LDA low disease activity, PtGA Patient Global Assessment of Disease Activity, VAS Visual Analog Scale
Fig. 2
Fig. 2
Associations between moderate improvements in PtGA and Pain, HAQ-DI, and FACIT-F outcomes at month 3 in csDMARD-IR patients. Proportions of csDMARD-IR patients at month 3 who did/did not report a mild Pain (VAS score ≤ 20 mm), b moderate improvements in Pain (≥ 30% decreases from baseline), c substantial improvements in Pain (≥ 50% decreases from baseline), d HAQ-DI scores ≥ normative values (≤ 0.25), or e FACIT-F scores ≥ normative values (≥ 43.5), stratified by reporting of moderate improvements in PtGA (≥ 30% decreases from baseline). Denominators represent the number of patients who did/did not report moderate PtGA improvements, respectively. Abbreviations: BID twice daily, CI confidence interval, csDMARD conventional synthetic disease-modifying antirheumatic drug, FACIT-F Functional Assessment of Chronic Illness Therapy-Fatigue, HAQ-DI Health Assessment Questionnaire-Disability Index, IR inadequate responder, PtGA Patient Global Assessment of Disease Activity, VAS Visual Analog Scale
Fig. 3
Fig. 3
Associations between substantial improvements in PtGA and Pain, HAQ-DI, and FACIT-F outcomes at month 3 in csDMARD-IR patients. Proportions of csDMARD-IR patients at month 3 who did/did not report a mild Pain (VAS score ≤ 20 mm), b moderate improvements in Pain (≥ 30% decreases from baseline), c substantial improvements in Pain (≥ 50% decreases from baseline), d HAQ-DI scores ≥ normative values (≤ 0.25), or e FACIT-F scores ≥ normative values (≥ 43.5), stratified by reporting of substantial improvements in PtGA (≥ 50% decreases from baseline). Denominators represent the number of patients who did/did not report substantial PtGA improvements, respectively. Abbreviations: BID twice daily, CI confidence interval, csDMARD conventional synthetic disease-modifying antirheumatic drug, FACIT-F Functional Assessment of Chronic Illness Therapy-Fatigue, HAQ-DI Health Assessment Questionnaire-Disability Index, IR inadequate responder, PtGA Patient Global Assessment of Disease Activity, VAS Visual Analog Scale

References

    1. Strand V, Singh JA. Newer biological agents in rheumatoid arthritis: impact on health-related quality of life and productivity. Drugs. 2010;70:121–145. doi: 10.2165/11531980-000000000-00000.
    1. Carr A, Hewlett S, Hughes R, Mitchell H, Ryan S, Carr M, et al. Rheumatology outcomes: the patient’s perspective. J Rheumatol. 2003;30:880–883.
    1. Kirwan JR, Hewlett SE, Heiberg T, Hughes RA, Carr M, Hehir M, et al. Incorporating the patient perspective into outcome assessment in rheumatoid arthritis--progress at OMERACT 7. J Rheumatol. 2005;32:2250–2256.
    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. Felson DT, Anderson JJ, Boers M, Bombardier C, Chernoff M, Fried B, et al. The American College of Rheumatology preliminary core set of disease activity measures for rheumatoid arthritis clinical trials. The Committee on Outcome Measures in Rheumatoid Arthritis Clinical Trials. Arthritis Rheum. 1993;36:729–40.
    1. Gossec L, Paternotte S, Aanerud GJ, Balanescu A, Boumpas DT, Carmona L, et al. Finalisation and validation of the rheumatoid arthritis impact of disease score, a patient-derived composite measure of impact of rheumatoid arthritis: a EULAR initiative. Ann Rheum Dis. 2011;70:935–942. doi: 10.1136/ard.2010.142901.
    1. US Food and Drug Administration. Guidance for Industry. Patient-reported outcome measures: use in medical product development to support labeling claims 2009. . Accessed 25 Sept 2020.
    1. Kvitkina T, ten Haaf A, Reken S, McGauran N, Wieseler B. Patient-relevant outcomes and surrogates in the early benefit assessment of drugs: first experiences. Z Evid Fortbild Qual Gesundhwes. 2014;108:528–538. doi: 10.1016/j.zefq.2014.06.015.
    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. Ward MM. Clinical measures in rheumatoid arthritis: which are most useful in assessing patients? J Rheumatol. 1994;21:17–27.
    1. Kirwan JR, Minnock P, Adebajo A, Bresnihan B, Choy E, de Wit M, et al. Patient perspective: fatigue as a recommended patient centered outcome measure in rheumatoid arthritis. J Rheumatol. 2007;34:1174–1177.
    1. Khan NA, Spencer HJ, Abda E, Aggarwal A, Alten R, Ancuta C, et al. Determinants of discordance in patients’ and physicians’ rating of rheumatoid arthritis disease activity. Arthritis Care Res (Hoboken). 2012;64:206–214. doi: 10.1002/acr.20685.
    1. Studenic P, Radner H, Smolen JS, Aletaha D. Discrepancies between patients and physicians in their perceptions of rheumatoid arthritis disease activity. Arthritis Rheum. 2012;64:2814–2823. doi: 10.1002/art.34543.
    1. Desthieux C, Hermet A, Granger B, Fautrel B, Gossec L. Patient-physician discordance in global assessment in rheumatoid arthritis: a systematic literature review with metaanalysis. Arthritis Care Res (Hoboken) 2016;68:1767–1773. doi: 10.1002/acr.22902.
    1. Lee YC. Effect and treatment of chronic pain in inflammatory arthritis. Curr Rheumatol Rep. 2013;15:300. doi: 10.1007/s11926-012-0300-4.
    1. Barton JL, Imboden J, Graf J, Glidden D, Yelin EH, Schillinger D. Patient-physician discordance in assessments of global disease severity in rheumatoid arthritis. Arthritis Care Res (Hoboken). 2010;62:857–864. doi: 10.1002/acr.20132.
    1. Karpouzas GA, Ramadan SN, Cost CE, Draper TL, Hernandez E, Strand V, et al. Discordant patient-physician assessments of disease activity and its persistence adversely impact quality of life and work productivity in US Hispanics with rheumatoid arthritis. RMD Open. 2017;3:e000551. doi: 10.1136/rmdopen-2017-000551.
    1. Karpouzas GA, Strand V, Ormseth SR. Latent profile analysis approach to the relationship between patient and physician global assessments of rheumatoid arthritis activity. RMD Open. 2018;4:e000695. doi: 10.1136/rmdopen-2018-000695.
    1. Smolen JS, Strand V, Koenig AS, Szumski A, Kotak S, Jones TV. Discordance between patient and physician assessments of global disease activity in rheumatoid arthritis and association with work productivity. Arthritis Res Ther. 2016;18:114. doi: 10.1186/s13075-016-1004-3.
    1. Busch-Dienstfertig M, González-Rodríguez S. IL-4, JAK-STAT signaling, and pain. JAKSTAT. 2013;2:e27638.
    1. Salaffi F, Giacobazzi G, Di Carlo M. Chronic pain in inflammatory arthritis: mechanisms, metrology, and emerging targets-a focus on the JAK-STAT pathway. Pain Res Manag. 2018;2018:8564215.
    1. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. 2017;77:521–46.
    1. Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, et al. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol. 2020;80:106210. doi: 10.1016/j.intimp.2020.106210.
    1. Ogdie A, de Vlam K, McInnes IB, Mease PJ, Baer P, Lukic T, et al. Efficacy of tofacitinib in reducing pain in patients with rheumatoid arthritis, psoriatic arthritis or ankylosing spondylitis. RMD Open. 2020;6:e001042. doi: 10.1136/rmdopen-2019-001042.
    1. Wallenstein GV, Kanik KS, Wilkinson B, Cohen S, Cutolo M, Fleishmann R, et al. Effects of the oral Janus kinase inhibitor tofacitinib on patient-reported outcomes in patients with active rheumatoid arthritis: results of two phase 2 randomised controlled trials. Clin Exp Rheumatol. 2016;34:430–442.
    1. Strand V, Kremer J, Wallenstein G, Kanik KS, Connell C, Gruben D, et al. Effects of tofacitinib monotherapy on patient-reported outcomes in a randomized phase 3 study of patients with active rheumatoid arthritis and inadequate responses to DMARDs. Arthritis Res Ther. 2015;17:307. doi: 10.1186/s13075-015-0825-9.
    1. Strand V, van Vollenhoven RF, Lee EB, Fleischmann R, Zwillich SH, Gruben D, et al. Tofacitinib or adalimumab versus placebo: patient-reported outcomes from a phase 3 study of active rheumatoid arthritis. Rheumatology (Oxford) 2016;55:1031–1041. doi: 10.1093/rheumatology/kev442.
    1. Strand V, Lee EB, Fleischmann R, Alten RE, Koncz T, Zwillich SH, et al. Tofacitinib versus methotrexate in rheumatoid arthritis: patient-reported outcomes from the randomised phase III ORAL Start trial. RMD Open. 2016;2:e000308. doi: 10.1136/rmdopen-2016-000308.
    1. Strand V, Kremer JM, Gruben D, Krishnaswami S, Zwillich SH, Wallenstein GV. Tofacitinib in combination with conventional disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis: patient-reported outcomes from a Phase III randomized controlled trial. Arthritis Care Res (Hoboken) 2017;69:592–598. doi: 10.1002/acr.23004.
    1. Strand V, Burmester GR, Zerbini CA, Mebus CA, Zwillich SH, Gruben D, et al. Tofacitinib with methotrexate in third-line treatment of patients with active rheumatoid arthritis: patient-reported outcomes from a phase III trial. Arthritis Care Res. 2015;67:475–483. doi: 10.1002/acr.22453.
    1. Strand V, Mysler E, Moots RJ, Wallenstein G, DeMasi R, Gruben D, et al. Patient-reported outcomes for tofacitinib with and without methotrexate, or adalimumab with methotrexate, in rheumatoid arthritis: a phase IIIB/IV trial. RMD Open. 2019:5:e001040.
    1. Wollenhaupt J, Silverfield J, Lee EB, Curtis JR, Wood SP, Soma K, et al. Safety and efficacy of tofacitinib, an oral Janus kinase inhibitor, for the treatment of rheumatoid arthritis in open-label, longterm extension studies. J Rheumatol. 2014;41:837–852. doi: 10.3899/jrheum.130683.
    1. Yamanaka H, Tanaka Y, Takeuchi T, Sugiyama N, Yuasa H, Toyoizumi S, et al. Tofacitinib, an oral Janus kinase inhibitor, as monotherapy or with background methotrexate, in Japanese patients with rheumatoid arthritis: an open-label, long-term extension study. Arthritis Res Ther. 2016;18:34. doi: 10.1186/s13075-016-0932-2.
    1. Wollenhaupt J, Lee EB, Curtis JR, Silverfield J, Terry K, Soma K, et al. Safety and efficacy of tofacitinib for up to 9.5 years in the treatment of rheumatoid arthritis: final results of a global, open-label, long-term extension study. Arthritis Res Ther. 2019;21:89. doi: 10.1186/s13075-019-1866-2.
    1. Kremer J, Li Z-G, Hall S, Fleischmann R, Genovese M, Martin-Mola E, et al. Tofacitinib in combination with nonbiologic disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis: a randomized trial. Ann Intern Med. 2013;159:253–261. doi: 10.7326/0003-4819-159-4-201308200-00006.
    1. van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, García Meijide JA, Wagner S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367:508–519. doi: 10.1056/NEJMoa1112072.
    1. van der Heijde D, Tanaka Y, Fleischmann R, Keystone E, Kremer J, Zerbini C, et al. Tofacitinib (CP-690,550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic study. Arthritis Rheum. 2013;65:559–570. doi: 10.1002/art.37816.
    1. Strand V, van der Heijde D, Tanaka Y, Keystone E, Kremer J, Zerbini CAF, et al. Tofacitinib in combination with methotrexate in patients with rheumatoid arthritis: patient-reported outcomes from the 24-month Phase 3 ORAL Scan study. Clin Exp Rheumatol. 2020;38:848–57.
    1. Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–324. doi: 10.1002/art.1780310302.
    1. Lee EB, Fleischmann R, Hall S, Wilkinson B, Bradley J, Gruben D, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med. 2014;370:2377–2386. doi: 10.1056/NEJMoa1310476.
    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. Wells GA, Boers M, Shea B, Brooks PM, Simon LS, Strand CV, et al. Minimal disease activity for rheumatoid arthritis: a preliminary definition. J Rheumatol. 2005;32:2016–2024.
    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. Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, et al. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. J Pain. 2008;9:105–121. doi: 10.1016/j.jpain.2007.09.005.
    1. Farrar JT, Young JP, Jr, LaMoreaux L, Werth JL, Poole RM. Clinical importance of changes in chronic pain intensity measured on an 11-point numerical pain rating scale. Pain. 2001;94:149–158. doi: 10.1016/S0304-3959(01)00349-9.
    1. Krishnan E, Sokka T, Häkkinen 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. Wells GA, Tugwell P, Kraag GR, Baker PR, Groh J, Redelmeier DA. Minimum important difference between patients with rheumatoid arthritis: the patient’s perspective. J Rheumatol. 1993;20:557–560.
    1. Montan I, Löwe B, Cella D, Mehnert A, Hinz A. General population norms for the Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue scale. Value Health. 2018;21:1313–1321. doi: 10.1016/j.jval.2018.03.013.
    1. Cella D, Lai JS, Chang CH, Peterman A, Slavin M. Fatigue in cancer patients compared with fatigue in the general United States population. Cancer. 2002;94:528–538. doi: 10.1002/cncr.10245.
    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. Stoffer MA, Smolen JS, Woolf A, Ambrozic A, Bosworth A, Carmona L, et al. Development of patient-centred standards of care for rheumatoid arthritis in Europe: the project. Ann Rheum Dis. 2014;73:902–5.
    1. Strand V, Wright GC, Bergman MJ, Tambiah J, Taylor PC. Patient expectations and perceptions of goal-setting strategies for disease management in rheumatoid arthritis. J Rheumatol. 2015;42:2046–2054. doi: 10.3899/jrheum.140976.
    1. ten Klooster PM, Veehof MM, Taal E, van Riel PL, van de Laar MA. Changes in priorities for improvement in patients with rheumatoid arthritis during 1 year of anti-tumour necrosis factor treatment. Ann Rheum Dis. 2007;66:1485–1490. doi: 10.1136/ard.2007.069765.
    1. American College of Rheumatology Pain Management Task Force Report of the American College of Rheumatology Pain Management Task Force. Arthritis Care Res (Hoboken). 2010;62:590–599. doi: 10.1002/acr.20005.
    1. Lee YC, Cui J, Lu B, Frits ML, Iannaccone CK, Shadick NA, et al. Pain persists in DAS28 rheumatoid arthritis remission but not in ACR/EULAR remission: a longitudinal observational study. Arthritis Res Ther. 2011;13:R83. doi: 10.1186/ar3353.
    1. Olofsson T, Wallman JK, Jöud A, Schelin ME, Ernestam S, van Vollenhoven R, et al. Unacceptable, refractory pain despite inflammation control in early rheumatoid arthritis and its relation to treatment strategy: results from the randomised controlled SWEFOT trial. Ann Rheum Dis. 2018;77(Suppl 2):117.
    1. Edwards RR, Wasan AD, Bingham CO, 3rd, Bathon J, Haythornthwaite JA, Smith MT, et al. Enhanced reactivity to pain in patients with rheumatoid arthritis. Arthritis Res Ther. 2009;11:R61. doi: 10.1186/ar2684.
    1. Leffler AS, Kosek E, Lerndal T, Nordmark B, Hansson P. Somatosensory perception and function of diffuse noxious inhibitory controls (DNIC) in patients suffering from rheumatoid arthritis. Eur J Pain. 2002;6:161–176. doi: 10.1053/eujp.2001.0313.
    1. Rao JK, Weinberger M, Anderson LA, Kroenke K. Predicting reports of unmet expectations among rheumatology patients. Arthritis Rheum. 2004;51:215–221. doi: 10.1002/art.20246.
    1. Nelson EC, Eftimovska E, Lind C, Hager A, Wasson JH, Lindblad S. Patient reported outcome measures in practice. BMJ. 2015;350:g7818. doi: 10.1136/bmj.g7818.
    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. Anderson JK, Zimmerman L, Caplan L, Michaud K. Measures of rheumatoid arthritis disease activity: Patient (PtGA) and Provider (PrGA) Global Assessment of Disease Activity, Disease Activity Score (DAS) and Disease Activity Score with 28-Joint Counts (DAS28), Simplified Disease Activity Index (SDAI), Clinical Disease Activity Index (CDAI), Patient Activity Score (PAS) and Patient Activity Score-II (PASII), Routine Assessment of Patient Index Data (RAPID), Rheumatoid Arthritis Disease Activity Index (RADAI) and Rheumatoid Arthritis Disease Activity Index-5 (RADAI-5), Chronic Arthritis Systemic Index (CASI), Patient-Based Disease Activity Score With ESR (PDAS1) and Patient-Based Disease Activity Score without ESR (PDAS2), and Mean Overall Index for Rheumatoid Arthritis (MOI-RA). Arthritis Care Res (Hoboken). 2011;63 Suppl 11:S14-S36.
    1. Pincus T, Furer V, Keystone E, Yazici Y, Bergman MJ, Luijtens K. RAPID3 (Routine Assessment of Patient Index Data 3) severity categories and response criteria: Similar results to DAS28 (Disease Activity Score) and CDAI (Clinical Disease Activity Index) in the RAPID 1 (Rheumatoid Arthritis Prevention of Structural Damage) clinical trial of certolizumab pegol. Arthritis Care Res (Hoboken). 2011;63:1142–1149. doi: 10.1002/acr.20481.
    1. Pincus T, Swearingen CJ, Bergman MJ, Colglazier CL, Kaell AT, Kunath AM, et al. RAPID3 (Routine Assessment of Patient Index Data) on an MDHAQ (Multidimensional Health Assessment Questionnaire): agreement with DAS28 (Disease Activity Score) and CDAI (Clinical Disease Activity Index) activity categories, scored in five versus more than ninety seconds. Arthritis Care Res (Hoboken) 2010;62:181–189. doi: 10.1002/acr.20066.
    1. Pincus T, Swearingen CJ, Bergman M, Yazici Y. RAPID3 (Routine Assessment of Patient Index Data 3), a rheumatoid arthritis index without formal joint counts for routine care: proposed severity categories compared to disease activity score and clinical disease activity index categories. J Rheumatol. 2008;35:2136–2147. doi: 10.3899/jrheum.080182.

Source: PubMed

3
Abonnieren