Tofacitinib in Patients With Psoriatic Arthritis and Metabolic Syndrome: A Post hoc Analysis of Phase 3 Studies

Christopher T Ritchlin, Jon T Giles, Alexis Ogdie, Juan J Gomez-Reino, Philip Helliwell, Pamela Young, Cunshan Wang, Joseph Wu, Ana Belén Romero, John Woolcott, Lori Stockert, Christopher T Ritchlin, Jon T Giles, Alexis Ogdie, Juan J Gomez-Reino, Philip Helliwell, Pamela Young, Cunshan Wang, Joseph Wu, Ana Belén Romero, John Woolcott, Lori Stockert

Abstract

Objective: Metabolic syndrome (MetS) is a cluster of concurrent risk factors for cardiovascular disease and type 2 diabetes. This post hoc analysis explored key efficacy and safety endpoints in patients with psoriatic arthritis (PsA) and MetS treated with tofacitinib.

Methods: Tofacitinib 5 and 10 mg twice daily and placebo data were pooled from two Phase 3 studies (OPAL Broaden [12 months; ClinicalTrials.gov identifier NCT01877668]; OPAL Beyond [6 months; ClinicalTrials.gov identifier NCT01882439]); patients received one background conventional synthetic disease-modifying antirheumatic drug. Patients were stratified by baseline presence/absence of MetS. Efficacy and safety were reported to month 3 (tofacitinib and placebo) and 6 (tofacitinib only). Efficacy outcomes included: American College of Rheumatology (ACR)20/50/70, Health Assessment Questionnaire-Disability Index (HAQ-DI) response, Psoriasis Area Severity Index (PASI)75 response, and enthesitis/dactylitis resolution rates; and changes from baseline (Δ) in C-reactive protein, HAQ-DI, Patient's/Physician's Global Assessment of Arthritis, and patient-reported outcomes. Safety outcomes included treatment-emergent all-causality adverse events (AEs), Δ in lipid/hepatic values, and liver parameter increases.

Results: Of 710 patients, 41.4% (n = 294) had baseline MetS. All efficacy outcomes improved with both tofacitinib doses versus placebo, to month 3; tofacitinib efficacy was consistent to month 6, regardless of MetS status. MetS did not appear to affect the incidence of AEs or Δ in lipid/hepatic values with tofacitinib up to month 3 or 6. Arterial thromboembolism and myocardial infarction (adjudicated major adverse cardiovascular events) were each reported once in tofacitinib-treated patients with MetS.

Conclusion: Regardless of baseline MetS status, tofacitinib showed greater efficacy versus placebo in patients with active PsA. The tofacitinib safety profile appeared similar in patients with versus without MetS.

© 2020 The Authors. ACR Open Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.

Figures

Figure 1
Figure 1
Response rate (SE) for ACR20 (A), ACR50 (B), ACR70 (C), PASI75a (D), and HAQ‐DI responseb (E), enthesitis resolutionc (F) and dactylitis resolutiond (G) rates at months 3 and 6, by treatment group and baseline MetS status; pooled data from OPAL Broaden and OPAL Beyond. aPASI was assessed only in patients with baseline BSA ≥ 3% and PASI > 0. bHAQ‐DI response is defined as a decrease ≥ 0.35 among patients with baseline HAQ‐DI score ≥ 0.35. cEnthesitis resolution was assessed only in patients with baseline LEI > 0. dDactylitis resolution was assessed only in patients with baseline DSS > 0. A patient with a missing value was considered a nonresponder. Abbreviations: ACR, American College of Rheumatology; BID, twice daily; BSA, body surface area; DSS, Dactylitis Severity Score; HAQ‐DI, Health Assessment Questionnaire‐Disability Index; LEI, Leeds Enthesitis Index; MetS, metabolic syndrome; N, number of patients with nonmissing baseline MetS; PASI, Psoriasis Area and Severity Index; SE, standard error.
Figure 2
Figure 2
LS mean (SE) change from baseline in HAQ‐DI (A), PGA (VAS, mm) (B), PtGA (VAS, mm) (C), Pain (VAS, mm) (D), and FACIT‐F total score (E) at months 3 and 6 by treatment group and baseline MetS status, and mean (SE) change from baseline in CRP (F), at months 3 and 6, by treatment group, baseline MetS status, and baseline CRP ≤ 2.87 mg/L and > 2.87 mg/L; pooled data from OPAL Broaden and OPAL Beyond. Each of the endpoints (except CRP) were analyzed using a mixed model for repeated measures, with fixed effects of treatment, visit, study, treatment‐by‐visit, baseline MetS, treatment‐by‐baseline MetS, visit‐by‐baseline MetS, treatment‐by‐visit‐by‐baseline MetS interactions, geographical region, and baseline value. Missing values were not imputed. A common unstructured covariance matrix was used. LS mean (SE) changes from baseline were calculated at months 3 and 6 using two separate models. Results at month 3 were based on a model that included tofacitinib 5 mg BID, tofacitinib 10 mg BID, and placebo treatment groups; as the placebo treatment group was only included up to month 3 (end of placebo‐controlled period), the results at month 6 were based on a separate model that included only tofacitinib 5 mg BID and tofacitinib 10 mg BID treatment groups. Arithmetic mean (SE) was calculated for change from baseline in CRP by treatment group, baseline MetS status, and CRP cutoff. Abbreviations: Δ, change from baseline; BID, twice daily; CRP, C‐reactive protein; FACIT‐F, Functional Assessment of Chronic Illness Therapy‐Fatigue; HAQ‐DI, Health Assessment Questionnaire‐Disability Index; LS mean, least squares mean; MetS, metabolic syndrome; N, number of patients evaluable at months 3 and 6; PtGA, Patient’s Global Assessment of Arthritis; PGA, Physician’s Global Assessment of Arthritis; SE, standard error; VAS, Visual Analog Scale.

References

    1. Haroon M, Gallagher P, Heffernan E, FitzGerald O. High prevalence of metabolic syndrome and of insulin resistance in psoriatic arthritis is associated with the severity of underlying disease. J Rheumatol 2014;41:1357–65.
    1. Alberti KGMM, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA, et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009;120:1640–5.
    1. Mottillo S, Filion KB, Genest J, Joseph L, Pilote L, Poirier P, et al. The metabolic syndrome and cardiovascular risk: a systematic review and meta‐analysis. J Am Coll Cardiol 2010;56:1113–32.
    1. Meigs JB, Wilson PWF, Fox CS, Vasan RS, Nathan DM, Sullivan LM, et al. Body mass index, metabolic syndrome, and risk of type 2 diabetes or cardiovascular disease. J Clin Endocrinol Metab 2006;91:2906–12.
    1. Wisse BE. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol 2004;15:2792–800.
    1. Després J‐P, Lemieux I. Abdominal obesity and metabolic syndrome. Nature 2006;444:881–7.
    1. Coates LC, FitzGerald O, Helliwell PS, Paul C. Psoriasis, psoriatic arthritis, and rheumatoid arthritis: is all inflammation the same? . Semin Arthritis Rheum 2016;46:291–304.
    1. Versini M, Jeandel P‐Y, Rosenthal E, Shoenfeld Y. Obesity in autoimmune diseases: not a passive bystander. Autoimmun Rev 2014;13:981–1000.
    1. Haroon M, Rafiq Chaudhry AB, Fitzgerald O. Higher prevalence of metabolic syndrome in patients with psoriatic arthritis: a comparison with a control group of noninflammatory rheumatologic conditions. J Rheumatol 2016;43:463–4.
    1. Costa L, Caso F, Ramonda R, Del Puente A, Cantarini L, Darda MA, et al. Metabolic syndrome and its relationship with the achievement of minimal disease activity state in psoriatic arthritis patients: an observational study. Immunol Res 2015;61:147–53.
    1. Singh S, Facciorusso A, Singh AG, Vande Casteele N, Zarrinpar A, Prokop LJ, et al. Obesity and response to anti‐tumor necrosis factor‐α agents in patients with select immune‐mediated inflammatory diseases: a systematic review and meta‐analysis. PLoS One 2018;13:e0195123.
    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;377:1537–50.
    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;377:1525–36.
    1. Nash P, Coates LC, Kivitz AJ, Mease PJ, Gladman DD, Covarrubias‐Cobos JA, et al. SAT0293 Safety and efficacy of tofacitinib, an oral Janus kinase inhibitor, up to 36 months in patients with active psoriatic arthritis: data from the third interim analysis of OPAL Balance, an open‐label long‐term extension study [abstract]. Ann Rheum Dis 2018;77 (Suppl 2) .
    1. Ritchlin C, Ogdie A, Giles J, Gomez‐Reino J, Helliwell P, Stockert L, et al. Impact of baseline body mass index on the efficacy and safety of tofacitinib in patients with psoriatic arthritis [abstract]. Arthritis Rheumatol 2019;71 (Suppl 10) URL: .
    1. Felson DT, Anderson JJ, Boers M, Bombardier C, Furst D, Goldsmith C, et al. American College of Rheumatology. Preliminary definition of improvement in rheumatoid arthritis. Arthritis Rheum 1995;38:727–35.
    1. Mease PJ, Woolley JM, Bitman B, Wang BC, Globe DR, Singh A. Minimally important difference of Health Assessment Questionnaire in psoriatic arthritis: relating thresholds of improvement in functional ability to patient‐rated importance and satisfaction. J Rheumatol 2011;38:2461–5.
    1. Ware JE, Kosinski M Jr, Bjorner JB, Turner‐Bowker DM, Gandek B, Maruish ME. User’s manual for the SF‐36v2® Health Survey. 2nd ed Lincoln (RI): QualityMetric Incorporated; 2007.
    1. EuroQoL Group . EuroQol–a new facility for the measurement of health‐related quality of life. Health Policy 1990;16:199–208.
    1. Klingberg E, Bilberg A, Björkman S, Hedberg M, Jacobsson L, Forsblad‐d'Elia H, et al. Weight loss improves disease activity in patients with psoriatic arthritis and obesity: an interventional study. Arthritis Res Ther 2019;21:17.
    1. Strober B, Teller C, Yamauchi P, Miller JL, Hooper M, Yang Y‐C, et al. Effects of etanercept on C‐reactive protein levels in psoriasis and psoriatic arthritis. Br J Dermatol 2008;159:322–30.
    1. Migita K, Miyashita T, Izumi Y, Koga T, Komori A, Maeda Y, et al. Inhibitory effects of the JAK inhibitor CP690,550 on human CD4(+) T lymphocyte cytokine production. BMC Immunol 2011;12:51.
    1. Meyer DM, Jesson MI, Li X, Elrick MM, Funckes‐Shippy CL, Warner JD, et al. Anti‐inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP‐690,550, in rat adjuvant‐induced arthritis. J Inflamm (Lond) 2010;7:41.
    1. McInnes IB, Byers NL, Higgs RE, Lee J, Macias WL, Na S, et al. Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther 2019;21:183.
    1. Raychaudhuri SK, Abria C, Raychaudhuri SP. Regulatory role of the JAK STAT kinase signalling system on the IL‐23/IL‐17 cytokine axis in psoriatic arthritis. Ann Rheum Dis 2017;76:e36.
    1. Gladman DD, Charles‐Schoeman C, McInnes IB, Veale DJ, Thiers B, Nurmohamed M, et al. Changes in lipid levels and incidence of cardiovascular events following tofacitinib treatment in patients with psoriatic arthritis: a pooled analysis across phase III and long‐term extension studies. Arthritis Care Res (Hoboken) 2019;71:1387–95.
    1. European Medicines Agency . Xeljanz (tofacitinib): summary of product characteristics. 2020. URL: .
    1. US Food and Drug Administration . XELJANZ® (tofacitinib): highlights of prescribing information. 2019. URL: .
    1. Hamaguchi M, Kojima T, Takeda N, Nakagawa T, Taniguchi H, Fujii K, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med 2005;143:722–8.
    1. Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Metabolic syndrome: definitions and controversies. BMC Med 2011;9:48.
    1. O'Neill S, O'Driscoll L. Metabolic syndrome: a closer look at the growing epidemic and its associated pathologies. Obes Rev 2015;16:1–12.
    1. Charles‐Schoeman C, Wicker P, Gonzalez‐Gay MA, Boy M, Zuckerman A, Soma K, et al. Cardiovascular safety findings in patients with rheumatoid arthritis treated with tofacitinib, an oral Janus kinase inhibitor. Semin Arthritis Rheum 2016;46:261–71.

Source: PubMed

3
Subskrybuj