Safety Profile of Upadacitinib up to 3 Years in Psoriatic Arthritis: An Integrated Analysis of Two Pivotal Phase 3 Trials

Gerd R Burmester, Kevin Winthrop, Ricardo Blanco, Peter Nash, Philippe Goupille, Valderilio F Azevedo, Carlo Salvarani, Andrea Rubbert-Roth, Elizabeth Lesser, Ralph Lippe, Apinya Lertratanakul, Reva M Mccaskill, John Liu, Eric M Ruderman, Gerd R Burmester, Kevin Winthrop, Ricardo Blanco, Peter Nash, Philippe Goupille, Valderilio F Azevedo, Carlo Salvarani, Andrea Rubbert-Roth, Elizabeth Lesser, Ralph Lippe, Apinya Lertratanakul, Reva M Mccaskill, John Liu, Eric M Ruderman

Abstract

Introduction: This integrated analysis describes the safety profile of upadacitinib, an oral Janus kinase inhibitor, at 15 and 30 mg once daily for up to 3 years of exposure in patients with active psoriatic arthritis (PsA) who had a prior inadequate response or intolerance to ≥ 1 non-biologic or biologic disease-modifying antirheumatic drug.

Methods: Safety data were pooled and analyzed from two randomized, placebo-controlled phase 3 trials. Both trials evaluated upadacitinib 15 mg and 30 mg once daily, and one trial also evaluated adalimumab 40 mg every other week. Treatment-emergent adverse events (TEAEs) and laboratory data were summarized for four groups: pooled placebo, pooled upadacitinib 15 mg, pooled upadacitinib 30 mg, and adalimumab. TEAEs were reported as exposure-adjusted event rates (events per 100 patient-years [E/100 PY]) up to a data cut-off of June 29, 2020.

Results: A total of 2257 patients received ≥ 1 dose of upadacitinib 15 mg (N = 907) or 30 mg (N = 921) for 2504.6 PY of exposure or adalimumab (N = 429) for 549.7 PY of exposure. Upper respiratory tract infection, nasopharyngitis, and increased creatine phosphokinase (CPK) were the most common TEAEs with upadacitinib. Rates of malignancies, adjudicated major adverse cardiovascular events (MACEs) and venous thromboembolic events (VTEs), and deaths were similar across treatment groups. Rates of herpes zoster (HZ) and opportunistic infections (OI; excluding tuberculosis, HZ, and oral candidiasis) were higher with upadacitinib versus adalimumab. Serious infection, anemia, and CPK elevations were most frequent with upadacitinib 30 mg. Potentially clinically significant laboratory abnormalities were uncommon.

Conclusions: Upadacitinib 15 mg and adalimumab had similar safety profiles with the exception of HZ and OIs, consistent with what was observed in rheumatoid arthritis. Rates of malignancies, MACEs, VTEs, and deaths were comparable among patients receiving upadacitinib and adalimumab. No new safety risks emerged with longer-term exposure to upadacitinib.

Trial registration numbers: SELECT-PsA 1: NCT03104400; SELECT-PsA 2: NCT03104374.

Keywords: Adalimumab; JAK inhibitor; Psoriatic arthritis; SELECT-PsA 1; SELECT-PsA 2; Safety; Upadacitinib.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Event rates per 100 patient years for adverse events of special interest (a–l). Events are presented as exposure-adjusted event rates per 100 patient-years (E/100 PY), calculated as the total number of events adjusted for total exposure to UPA, ADA, or PBO. UPA 15 mg and UPA 30 mg QD groups include patients who were originally assigned to UPA and patients originally assigned to PBO who switched to UPA at week 24. aOpportunistic infections excluded tuberculosis, herpes zoster, and oral candidiasis. bMACE was defined as CV death, non-fatal MI, and non-fatal stroke. cVTE was defined as deep vein thrombosis and pulmonary embolism. ADA adalimumab, CPK creatine phosphokinase, E/100 PY event per 100 patient-years, EOW every other week, MACE major adverse cardiovascular event, MI myocardial infarction, NMSC non-melanoma skin cancer, PBO placebo, UPA upadacitinib, VTE venous thromboembolism
Fig. 2
Fig. 2
Mean changes in hemoglobin, platelets, neutrophils, and lymphocytes levels (ad). Baseline mean includes patients with non-missing baseline and at least one post-baseline value. ADA adalimumab, EOW every other week, UPA upadacitinib

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(Suppl 2):14–17.
    1. Coates LC, Kavanaugh A, Mease PJ, et al. Group for research and assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68(5):1060–1071.
    1. Singh JA, Guyatt G, Ogdie A, et al. Special article: 2018 American College of Rheumatology/National Psoriasis Foundation guideline for the treatment of psoriatic arthritis. Arthritis Rheumatol. 2019;71(1):5–32.
    1. Gossec L, Baraliakos X, Kerschbaumer A, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis. 2020;79(6):700–712.
    1. Parmentier JM, Voss J, Graff C, et al. In vitro and in vivo characterization of the JAK1 selectivity of upadacitinib (ABT-494) BMC Rheumatol. 2018;2:23.
    1. European Medicines Agency. Upadacitinib summary of product characteristics. 2021. . Accessed 6 Apr 2021
    1. Food and Drug Administration. Upadacitinib prescribing information. 2019. . Accessed 6 Apr 2021
    1. Genovese MC, Fleischmann R, Combe B, et al. Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. Lancet. 2018;391(10139):2513–2524.
    1. Smolen JS, Pangan AL, Emery P, et al. Upadacitinib as monotherapy in patients with active rheumatoid arthritis and inadequate response to methotrexate (SELECT-MONOTHERAPY): a randomised, placebo-controlled, double-blind phase 3 study. Lancet. 2019;393(10188):2303–2311.
    1. Fleischmann R, Pangan AL, Song IH, et al. Upadacitinib versus placebo or adalimumab in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase III, double-blind, randomized controlled trial. Arthritis Rheumatol. 2019;71(11):1788–1800.
    1. van Vollenhoven R, Takeuchi T, Pangan AL, et al. Efficacy and safety of upadacitinib monotherapy in methotrexate-naive patients with moderately-to-severely active rheumatoid arthritis (SELECT-EARLY): a multicenter, multi-country, randomized, double-blind, active comparator-controlled trial. Arthritis Rheumatol. 2020;72(10):1607–1620.
    1. Burmester GR, Kremer JM, Van den Bosch F, 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(10139):2503–2512.
    1. Mease PJ, Lertratanakul A, Anderson JK, et al. Upadacitinib for psoriatic arthritis refractory to biologics: SELECT-PsA 2. Ann Rheum Dis. 2021;80:312–320.
    1. McInnes IB, Anderson JK, Magrey M, et al. Trial of upadacitinib and adalimumab for psoriatic arthritis. N Engl J Med. 2021;384(13):1227–1239.
    1. van der Heijde D, Song I-H, Pangan AL, et al. Efficacy and safety of upadacitinib in patients with active ankylosing spondylitis (SELECT-AXIS 1): a multicentre, randomised, double-blind, placebo-controlled, phase 2/3 trial. Lancet. 2019;394(10214):2108–2117.
    1. McInnes I, Kato K, Magrey M, et al. Long-term safety and effectiveness of upadacitinib in patients with psoriatic arthritis: results at 56 weeks from the SELECT-PsA 1 study. Ann Rheum Dis. 2021;80(Suppl 1):1288–1289.
    1. Mease PJ, Lertratanakul A, Papp KA, et al. Upadacitinib in patients with psoriatic arthritis and inadequate response to biologics: 56-week data from the randomized controlled phase 3 SELECT-PsA 2 study. Rheumatol Ther. 2021 doi: 10.1007/s40744-021-00305-z.
    1. Cohen SB, van Vollenhoven RF, Winthrop KL, et al. Safety profile of upadacitinib in rheumatoid arthritis: integrated analysis from the SELECT phase III clinical programme. Ann Rheum Dis. 2020;80(3):304–311.
    1. Ramiro S, Sepriano A, Chatzidionysiou K, et al. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2016 update of the EULAR recommendations for management of rheumatoid arthritis. Ann Rheum Dis. 2017;76(6):1101–1136.
    1. Desai RJ, Thaler KJ, Mahlknecht P, et al. Comparative risk of harm associated with the use of targeted immunomodulators: a systematic review. Arthritis Care Res (Hoboken) 2016;68(8):1078–1088.
    1. Mease PJ, Kavanaugh A, Reimold A, et al. Secukinumab in the treatment of psoriatic arthritis: efficacy and safety results through 3 years from the year 1 extension of the randomised phase III FUTURE 1 trial. RMD Open. 2018;4(2):e000723.
    1. Combe B, Rahman P, Kameda H, et al. Safety results of ixekizumab with 1822.2 patient-years of exposure: an integrated analysis of 3 clinical trials in adult patients with psoriatic arthritis. Arthritis Res Ther. 2020;22(1):14.
    1. Burmester GR, Curtis JR, Yun H, et al. An integrated analysis of the safety of tofacitinib in psoriatic arthritis across phase III and long-term extension studies with comparison to real-world observational data. Drug Saf. 2020;43(4):379–392.
    1. Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–243.
    1. Choy EH. Clinical significance of Janus kinase inhibitor selectivity. Rheumatology. 2018;58(6):953–962.
    1. Chen Y-H, Chen Y-M, Smolen JS, et al. FRI0164 Incidence rate and characterization of herpes zoster in patients with moderate-to-severe rheumatoid arthritis: an update from baricitinib clinical studies. Ann Rheum Dis. 2019;78(Suppl 2):755.
    1. Winthrop KL, Lebwohl M, Cohen AD, et al. Herpes zoster in psoriasis patients treated with tofacitinib. J Am Acad Dermatol. 2017;77(2):302–309.
    1. Curtis JR, Xie F, Yang S, et al. Risk for herpes zoster in tofacitinib-treated rheumatoid arthritis patients with and without concomitant methotrexate and glucocorticoids. Arthritis Care Res (Hoboken) 2019;71(9):1249–1254.
    1. European Medicines Agency. PRAC recommendations on signals. 2021. . Accessed 18 Aug 2021
    1. Food and Drug Administration. Initial safety trial results find increased risk of serious heart-related problems and cancer with arthritis and ulcerative colitis medicine Xeljanz, Xeljanz XR (tofacitinib). 2021. . Accessed 18 Aug 2021
    1. Food and Drug Administration. FDA requires warnings about increased risk of serious heart-related events, cancer, blood clots, and death for JAK inhibitors that treat certain chronic inflammatory conditions. 2021. . Accessed 10 Sept 2021
    1. Scott IC, Hider SL, Scott DL. Thromboembolism with Janus kinase (JAK) inhibitors for rheumatoid arthritis: How real is the risk? Drug Saf. 2018;41(7):645–653.
    1. Ogdie A, Kay McGill N, Shin DB, et al. Risk of venous thromboembolism in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a general population-based cohort study. Eur Heart J. 2017;39(39):3608–3614.
    1. Ogdie A, Yu Y, Haynes K, et al. Risk of major cardiovascular events in patients with psoriatic arthritis, psoriasis and rheumatoid arthritis: a population-based cohort study. Ann Rheum Dis. 2015;74(2):326–332.
    1. Charles-Schoeman C, DeMasi R, Valdez H, et al. Risk factors for major adverse cardiovascular events in phase III and long-term extension studies of tofacitinib in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019;71(9):1450–1459.
    1. Mease P, Charles-Schoeman C, Cohen S, et al. Incidence of venous and arterial thromboembolic events reported in the tofacitinib rheumatoid arthritis, psoriasis and psoriatic arthritis development programmes and from real-world data. Ann Rheum Dis. 2020;79(11):1400–1413.
    1. Taylor PC, Weinblatt ME, Burmester GR, et al. Cardiovascular safety during treatment with baricitinib in rheumatoid arthritis. Arthritis Rheumatol. 2019;71(7):1042–1055.
    1. Franks AL, Slansky JE. Multiple associations between a broad spectrum of autoimmune diseases, chronic inflammatory diseases and cancer. Anticancer Res. 2012;32(4):1119–1136.
    1. Vaengebjerg S, Skov L, Egeberg A, Loft ND. Prevalence, incidence, and risk of cancer in patients with psoriasis and psoriatic arthritis: a systematic review and meta-analysis. JAMA Dermatol. 2020;156(4):421–429.
    1. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;17(1):78.
    1. Rida MA, Chandran V. Challenges in the clinical diagnosis of psoriatic arthritis. Clin Immunol. 2020;214:108390.
    1. Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a randomized trial of patients with active ulcerative colitis. Gastroenterology. 2020;158(8):2139–49.e14.
    1. Aithal GP. Hepatotoxicity related to antirheumatic drugs. Nat Rev Rheumatol. 2011;7(3):139.
    1. Inokuma S, Kono H, Nakayama H, Yamazaki J. Immunoglobulin and lymphocyte decrease concurrent with adverse reactions induced by methotrexate for RA. Ann Rheum Dis. 2000;59:490.
    1. Gelfand JM, Wan J, Zhang H, et al. Risk of liver disease in patients with psoriasis, psoriatic arthritis, and rheumatoid arthritis receiving methotrexate: a population-based study. J Am Acad Dermatol. 2021;84(6):1636–1643.
    1. Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–1536.
    1. Smolen JS, Genovese MC, Takeuchi T, et al. Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol. 2019;46(1):7–18.
    1. Queeney K, Housley W, Sokolov J, Long A. FRI0131 Elucidating the mechanism underlying creatine phosphokinase upregulation with upadacitinib. Ann Rheum Dis. 2019;78(Suppl 2):734–735.

Source: PubMed

3
Abonner