Upadacitinib pharmacokinetics and exposure-response analyses of efficacy and safety in psoriatic arthritis patients - Analyses of phase III clinical trials

Elena Muensterman, Benjamin Engelhardt, Sathej Gopalakrishnan, Jaclyn K Anderson, Mohamed-Eslam F Mohamed, Elena Muensterman, Benjamin Engelhardt, Sathej Gopalakrishnan, Jaclyn K Anderson, Mohamed-Eslam F Mohamed

Abstract

Upadacitinib is an oral Janus kinase inhibitor approved for the treatment of rheumatoid arthritis (RA) and recently approved by the European Medicines Agency for the treatment of psoriatic arthritis (PsA). The efficacy and safety profile of upadacitinib in PsA have been established in the SELECT-PsA program in two global phase III studies, which evaluated upadacitinib 15 and 30 mg q.d. The analyses described here characterized upadacitinib pharmacokinetics and exposure-response relationships for efficacy and safety endpoints using data from the SELECT-PsA studies. Upadacitinib pharmacokinetics in patients with PsA were characterized through a Bayesian population analysis approach and were comparable to pharmacokinetics in patients with RA. Exposure-response relationships for key efficacy and safety endpoints were characterized using data from 1916 patients with PsA. The percentage of patients achieving efficacy endpoints at week 12 (American College of Rheumatology [ACR]50 and ACR70), 16 and 24 (sIGA0/1) increased with increasing upadacitinib average plasma concentration over a dosing interval, whereas no clear exposure-response trend was observed for ACR20 at week 12 or ACR20/50/70 at week 24 within the range of plasma exposures evaluated in the phase III PsA studies. No clear trends for exposure-response relationships were identified for experiencing pneumonia, herpes zoster infection, hemoglobin less than 8 g/dl, lymphopenia (grade ≥ 3), or neutropenia (grade ≥ 3) after 24 weeks of treatment. Shallow relationships with plasma exposures were observed for serious infections and hemoglobin decrease greater than 2 g/dl from baseline at week 24. Based on exposure-response analyses, the upadacitinib 15 mg q.d. regimen is predicted to achieve robust efficacy in patients with PsA and to be associated with limited incidences of reductions in hemoglobin or occurrence of serious infections.

Trial registration: ClinicalTrials.gov NCT03104374 NCT03104400.

Conflict of interest statement

B.E., J.K.A., and M.F.M. are employees of AbbVie and may hold AbbVie stock or stock options. E.M. and S.G. are former employees of AbbVie and may hold stock or stock options.

© 2021 AbbVie Inc. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

FIGURE 1
FIGURE 1
Goodness‐of‐fit plots of upadacitinib pharmacokinetic model and visual predictive checks (VPCs) of upadacitinib concentration versus time since last dose for patients with PsA stratified by dose (data from phase III PsA studies). (a) Observed versus population predicted concentrations. (b) Observed versus individual predicted concentrations. (c) Conditional weighted residuals versus time since last dose. (d) Conditional weighted residuals versus population predicted concentrations. (e) VPCs 15 mg upadacitinib. (f) VPCs 15 mg upadacitinib. VPCs: The shaded blue areas represent the 95% prediction interval of the 2.5th and 97.5th percentiles of simulated concentrations, the red shaded areas represent the 95% prediction interval of the 50th percentile of simulated concentrations, the solid red line represents median of the observed concentrations and dashed red lines represent the 2.5th and 97.5th percentile of the observed concentrations. PsA, psoriatic arthritis
FIGURE 2
FIGURE 2
Observed and model‐predicted ACR50 and ACR70 responses (NRI) at week 12 and sIGA 0/1 responses at week 16 and week 24 versus upadacitinib Cavg (final models). The blue solid line represents median predicted response and the blue shaded area represent 95% confidence intervals of the predicted response. The dots and error bars represent median and 95% binomial confidence intervals of binned observed rates. ACR, American College of Rheumatology; Cavg, average plasma concentration
FIGURE 3
FIGURE 3
Model‐predicted ACR50 and ACR70 response for 15 mg q.d., 30 mg q.d. regimens and placebo at week 12 stratified by covariate subgroups. ACR, American College of Rheumatology; bDMARD, disease‐modifying antirheumatic drugs; BMI, body mass index; DMARD, modifying antirheumatic drugs
FIGURE 4
FIGURE 4
Observed and model‐predicted percentage of patients with PsA with serious infections, greater than 2 g/dl decrease in hemoglobin, or greater than 2 g/dl with hemoglobin less than the lower limit for normal up to week 24 (final model). The blue solid line represents median predicted response and the blue shaded area represent 95% confidence intervals of the predicted response. The dots and error bars represent median and 95% binomial confidence intervals of binned observed rates. LOCF, last observation carried forward; PsA, psoriatic arthritis

References

    1. Mohamed MF, Klünder B, Camp HS, Othman AA. Exposure‐response analyses of upadacitinib efficacy in phase II trials in rheumatoid arthritis and basis for phase III dose selection. Clin Pharmacol Ther. 2019;106:1319‐1327.
    1. Duarte GV, Faillace C, Freire de Carvalho J. Psoriatic arthritis. Best Pract Res Clin Rheumatol. 2012;26:147‐156.
    1. Gladman DD, Chandran V. Observational cohort studies: lessons learnt from the University of Toronto Psoriatic Arthritis Program. Rheumatology (Oxford, England). 2011;50:25‐31.
    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;79:700‐712.
    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 Care Res (Hoboken). 2019;71:2‐29.
    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. Mohamed MF, Beck D, Camp HS, Othman AA. Preferential inhibition of JAK1 relative to JAK3 by upadacitinib: exposure‐response analyses of ex vivo data from 2 phase 1 clinical trials and comparison to tofacitinib. J Clin Pharmacol. 2020;60:188‐197.
    1. Norman P. Selective JAK inhibitors in development for rheumatoid arthritis. Expert Opin Investig Drugs. 2014;23:1067‐1077.
    1. RINVOQ™ (upadacitinib) [US package insert]. (2019).
    1. McInnes IB, Anderson JK, Magrey M, et al. Trial of upadacitinib and adalimumab for psoriatic arthritis. N Engl J Med. 2021;384:1227‐1239.
    1. Mease PJ, Lertratanakul A, Anderson JK, et al. Upadacitinib for psoriatic arthritis refractory to biologics: SELECT‐PsA 2. Ann Rheum Dis. 2020;80:312‐320.
    1. Sandborn WJ, Feagan BG, Loftus EV, et al. Efficacy and safety of upadacitinib in a randomized trial of patients with Crohn’s disease. Gastroenterology. 2020;158:2123‐2138.e2128.
    1. Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a randomized trial of patients With active ulcerative colitis. Gastroenterology. 2020;158:2139‐2149.e14. 10.1053/j.gastro.2020.02.030. Erratum in: Gastroenterology. 2020 Sep;159(3):1192.
    1. Guttman‐Yassky E, Thaçi D, Pangan AL, et al. Upadacitinib in adults with moderate to severe atopic dermatitis: 16‐week results from a randomized, placebo‐controlled trial. J Allergy Clin Immunol. 2020;145:877‐884.
    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;394:2108‐2117.
    1. Baker KF, Isaacs JD. Novel therapies for immune‐mediated inflammatory diseases: what can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn’s disease and ulcerative colitis? Ann Rheum Dis. 2018;77:175‐187.
    1. Mease P, Hall S, FitzGerald O, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377:1537‐1550.
    1. Mohamed MF, Zeng J, Marroum PJ, Song IH, Othman AA. Pharmacokinetics of upadacitinib with the clinical regimens of the extended‐release formulation utilized in rheumatoid arthritis phase 3 trials. Clin Pharmacol Drug Dev. 2019;8:208‐216.
    1. Klünder B, Mittapalli RK, Mohamed M‐EF, et al. Population pharmacokinetics of upadacitinib using the immediate‐release and extended‐release formulations in healthy subjects and subjects with rheumatoid arthritis: analyses of phase I‐III clinical trials. Clin Pharmacokinet. 2019;58:1045‐1058.
    1. Klunder B, Mohamed MF, Othman AA. Population pharmacokinetics of upadacitinib in healthy subjects and subjects with rheumatoid arthritis: analyses of phase I and II clinical trials. Clin Pharmacokinet. 2018;57:977‐988.
    1. Mohamed MF, Jungerwirth S, Asatryan A, Jiang P, Othman AA. Assessment of effect of CYP3A inhibition, CYP induction, OATP1B inhibition, and high‐fat meal on pharmacokinetics of the JAK1 inhibitor upadacitinib. Br J Clin Pharmacol. 2017;83:2242‐2248.
    1. Mohamed MF, Trueman S, Feng T, Anderson J, Marbury TC, Othman AA. Characterization of the effect of renal impairment on upadacitinib pharmacokinetics. J Clin Pharmacol. 2019;59:856‐862.
    1. Trueman S, Mohamed MF, Feng T, Lacerda AP, Marbury T, Othman AA. Characterization of the effect of hepatic impairment on upadacitinib pharmacokinetics. J Clin Pharmacol. 2019;59:1188‐1194.
    1. Mohamed M‐EF, Klünder B, Othman AA. Clinical pharmacokinetics of upadacitinib: review of data relevant to the rheumatoid arthritis indication. Clin Pharmacokinet. 2020;59:531‐544.
    1. Mohamed MF, Klunder B, Lacerda AP, Othman AA. Exposure‐response analyses for upadacitinib efficacy and safety in the Crohn’s disease CELEST study and bridging to the extended‐release formulation. Clin Pharmacol Ther. 2020;107:639‐649.
    1. Mohamed MF, Gopalakrishnan S, Teixeira HD, Othman AA. Exposure‐response analyses for upadacitinib efficacy in subjects with atopic dermatitis‐analyses of phase 2b study to support selection of phase 3 doses. J Clin Pharmacol. 2021;61:628‐635.
    1. Nader A, Stodtmann S, Friedel A, Mohamed MF, Othman AA. Pharmacokinetics of upadacitinib in healthy subjects and subjects with rheumatoid arthritis, Crohn’s disease, ulcerative colitis, or atopic dermatitis: population analyses of phase 1 and 2 clinical trials. J Clin Pharmacol. 2020;60:528‐539.
    1. Mohamed MF, Minocha M, Trueman S, et al. Characterization of the effect of upadacitinib on the pharmacokinetics of bupropion, a sensitive cytochrome P450 2B6 probe substrate. Clin Pharmacol Drug Dev. 2021;10:299‐306.
    1. Chan Kwong AHXP, Calvier EAM, Fabre D, Gattacceca F, Khier S. Prior information for population pharmacokinetic and pharmacokinetic/pharmacodynamic analysis: overview and guidance with a focus on the NONMEM PRIOR subroutine. J Pharmacokinet Pharmacodyn. 2020;47:431‐446.
    1. Nader A, Mohamed M‐EF, Winzenborg I, et al. Exposure‐response analyses of upadacitinib efficacy and safety in phase II and III studies to support benefit‐risk assessment in rheumatoid arthritis. Clin Pharmacol Ther. 2020;107:994‐1003.
    1. Puig L, Strohal R, Husni ME, et al. Cardiometabolic profile, clinical features, quality of life and treatment outcomes in patients with moderate‐to‐severe psoriasis and psoriatic arthritis. J Dermatol Treat. 2015;26:7‐15.
    1. Veale DJ, Fearon U. What makes psoriatic and rheumatoid arthritis so different? RMD Open. 2015;1:e000025.
    1. Glintborg B, Østergaard M, Dreyer L, et al. Treatment response, drug survival, and predictors thereof in 764 patients with psoriatic arthritis treated with anti‐tumor necrosis factor α therapy: results from the nationwide Danish DANBIO registry. Arthritis Rheum. 2011;63:382‐390.

Source: PubMed

3
Abonner