Population Pharmacokinetics of Abrocitinib in Healthy Individuals and Patients with Psoriasis or Atopic Dermatitis

Jessica Wojciechowski, Bimal K Malhotra, Xiaoxing Wang, Luke Fostvedt, Hernan Valdez, Timothy Nicholas, Jessica Wojciechowski, Bimal K Malhotra, Xiaoxing Wang, Luke Fostvedt, Hernan Valdez, Timothy Nicholas

Abstract

Background and objective: Abrocitinib is a Janus kinase 1 inhibitor in development for the treatment of atopic dermatitis (AD). This work characterized orally administered abrocitinib population pharmacokinetics in healthy individuals, patients with psoriasis, and patients with AD and the effects of covariates on abrocitinib exposure.

Methods: Abrocitinib concentration measurements (n = 6206) from 995 individuals from 11 clinical trials (seven phase I, two phase II, and two phase III) were analyzed, and a non-linear mixed-effects model was developed. Simulations of abrocitinib dose proportionality and steady-state accumulation of maximal plasma drug concentration (Cmax) and area under the curve (AUC) were conducted using the final model.

Results: A two-compartment model with parallel zero- and first-order absorption, time-dependent bioavailability, and time- and dose-dependent clearance best described abrocitinib pharmacokinetics. Abrocitinib coadministration with rifampin resulted in lower exposure, whereas Asian/other race coadministration with fluconazole and fluvoxamine, inflammatory skin conditions (psoriasis/AD), and hepatic impairment resulted in higher exposure. After differences in body weight are accounted for, Asian participants demonstrated a 1.43- and 1.48-fold increase in Cmax and AUC, respectively. The overall distribution of exposures (Cmax and AUC) was similar in adolescents and adults after accounting for differences in total body weight.

Conclusions: A population pharmacokinetics model was developed for abrocitinib that can be used to predict abrocitinib steady-state exposure in the presence of drug-drug interaction effects or intrinsic patient factors. Key covariates in the study population accounting for variability in abrocitinib exposures are Asian race and adolescent age, although these factors are not clinically meaningful.

Clinical trial numbers: NCT01835197, NCT02163161, NCT02201524, NCT02780167, NCT03349060, NCT03575871, NCT03634345, NCT03637790, NCT03626415, NCT03386279, NCT03937258.

Conflict of interest statement

Jessica Wojciechowski, Bimal K. Malhotra, Xiaoxing Wang, Luke Fostvedt, Hernan Valdez, and Timothy Nicholas are employees and shareholders of Pfizer Inc.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Schematic representation of the final abrocitinib population pharmacokinetic model. AK1 and FK1 are the amount and fraction of the total dose absorbed by first-order processes, respectively. F1 and F2 are the fractions of the total bioavailable (Fi) dose that enters the depot/absorption compartment (for first-order absorption, F1) and the central compartment (for zero-order absorption, F2), respectively. The rate of absorption is dictated by the first-order rate constant (ka) and the zero-order absorption process is dictated by the zero-order absorption rate (k0). Absorption delays, ALAG1 for first-order absorption lag and ALAG2 for zero-order absorption lag, assume the zero-order process starts at the same time as the first-order process (i.e., where ALAG1 = ALAG2 and the processes occur in parallel) or after the first-order process has commenced (i.e., where ALAG2 = ALAG1 + ALAG2 and the processes occur sequentially). For a two-compartment model, CL is clearance from the central compartment (L/h), Vc is the volume of central compartment (L), Q is the inter-compartmental clearance (L/h), and Vp is the volume of peripheral compartment (L). Fabs fraction absorbed
Fig. 2
Fig. 2
Prediction-corrected visual predictive check. The prediction-corrected observed data against time after dose (a) and time after first dose (c) are represented by blue circles and dashed black lines (median, 5th and 95th percentiles). The prediction-corrected simulated abrocitinib concentration (200 mg) based on the index population (n = 1000 simulations) are represented by the red lines and red shaded ribbons (median and 95% prediction interval of the median, respectively) and the blue lines and blue shaded ribbons (median and 95% prediction intervals of the 5th and 95th percentiles, respectively). The black dashed lines represent the proportion of observed BLQ concentrations over time after dose (b) and time after first dose (d), and the green solid line and green shaded ribbons are the median and 90% prediction intervals, respectively, of simulated BLQ concentrations (n = 1000) based on the index population. Yellow indicators in the x-axis represent the time bins for summarizing the data. BLQ below limit of quantification
Fig. 3
Fig. 3
Ratios of steady-state Cmax (a) and 24-h AUC (b) after abrocitinib 200 mg QD for given covariates. For each covariate scenario on the left y-axis, concentration-time profiles for 1000 trials of 30 randomly assigned participants administered 200 mg QD were simulated using the full model and summarized by Cmax (a) or 24-h AUC (b) at steady state. The geometric mean ratio of Cmax or 24-h AUC compared with the reference scenario (healthy, White, adult males, 70 kg, fasted status, phase III tablet) was calculated for each trial. The gray-colored density distributions represent the geometric mean ratios across all trials; red numbers are the proportion of trials with ratios of < 0.8 (left) or > 1.25 (right). Black numbers on the right y-axis are the median (5th and 95th percentiles) of ratios for the covariate scenario. The blue shaded region is the range of geometric mean ratios from 0.8 to 1.25, and the black vertical dashed line is a geometric mean ratio of 1. Reference low and high body weights are the 5th and 95th percentiles of the analysis population. AUC area under the plasma concentration-time curve, Cmax maximal concentration, DDI drug–drug interaction, QD once daily
Fig. 4
Fig. 4
Evaluation of race on abrocitinib 200 mg QD steady-state Cmax (a) and 24-h AUC (b) for all individuals. Red circles represent the distribution of 200 mg once-daily steady-state Cmax (a) and AUC (b) based on the EBE for all participants in the analysis population (irrespective of what dose they received), and blue box and whisker plots depict newly simulated participants (n = 200) based on the final population PK model. The model provides an appropriate depiction of the observed differences between the race categories. AUC area under the plasma concentration-time curve, Cmax maximal concentration, EBE empirical Bayes estimates, PK pharmacokinetics, QD once daily
Fig. 5
Fig. 5
Weight-based simulations on abrocitinib 200 mg QD steady-state Cmax (a) and 24-h AUC (b) in adolescents. Red circles represent the EBE of steady-state Cmax (a) and AUC (b) for participants from the two phase III studies who received abrocitinib 200 mg, and blue box and whisker plots depict new simulated populations (n = 200) based on the final population PK model, with representative weights observed in the two phase III studies (adolescent and adult categories), test weights (25, 30, 35, or 40 kg), or reference weight (70 kg). AUC area under the plasma concentration-time curve, Cmax maximal concentration, EBE empirical Bayes estimates, PK pharmacokinetics, QD once daily

References

    1. Vazquez ML, Kaila N, Strohbach JW, Trzupek JD, Brown MF, Flanagan ME, et al. Identification of N-{cis-3-[Methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino]cyclobutyl}propane-1-sulfo namide (PF-04965842): a selective JAK1 clinical candidate for the treatment of autoimmune diseases. J Med Chem. 2018;61(3):1130–1152. doi: 10.1021/acs.jmedchem.7b01598.
    1. Gooderham MJ, Forman SB, Bissonnette R, Beebe JS, Zhang W, Banfield C, et al. Efficacy and safety of oral Janus kinase 1 inhibitor abrocitinib for patients with atopic dermatitis: a phase 2 randomized clinical trial. JAMA Dermatol. 2019;155(12):1371–1379. doi: 10.1001/jamadermatol.2019.2855.
    1. Silverberg JI, Simpson EL, Thyssen JP, Gooderham M, Chan G, Feeney C, et al. Efficacy and safety of abrocitinib in patients with moderate-to-severe atopic dermatitis: a randomized clinical trial. JAMA Dermatol. 2020;156(8):863–873. doi: 10.1001/jamadermatol.2020.1406.
    1. Simpson EL, Sinclair R, Forman S, Wollenberg A, Aschoff R, Cork M, et al. Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet. 2020;396(10246):255–266. doi: 10.1016/S0140-6736(20)30732-7.
    1. European Medicines Agency. SmPC for Cibinqo 2021. . Accessed 5 Nov 2021.
    1. Pharmaceuticals and Medical Device Agency (PMDA) H.P. List of Approved Products in Reiwa 3rd-year October 27, 2021. . Accessed 5 Nov 2021.
    1. Schmieder GJ, Draelos ZD, Pariser DM, Banfield C, Cox L, Hodge M, et al. Efficacy and safety of the Janus kinase 1 inhibitor PF-04965842 in patients with moderate-to-severe psoriasis: phase II, randomized, double-blind, placebo-controlled study. Br J Dermatol. 2018;179(1):54–62. doi: 10.1111/bjd.16004.
    1. Peeva E, Hodge MR, Kieras E, Vazquez ML, Goteti K, Tarabar SG, et al. Evaluation of a Janus kinase 1 inhibitor, PF-04965842, in healthy subjects: a phase 1, randomized, placebo-controlled, dose-escalation study. Br J Clin Pharmacol. 2018;84(8):1776–1788. doi: 10.1111/bcp.13612.
    1. Dowty M, Yang X, Lin J, Bauman J, Doran A, Goosen T, et al. P190—the effect of CYP2C9 and CYP2C19 genotype on the pharmacokinetics of PF 04965842, a JAK1 inhibitor in clinical development. Drug Metab Pharmacokinet. 2020;35(1 Suppl):S80. doi: 10.1016/j.dmpk.2020.04.191.
    1. Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development-part 2: introduction to pharmacokinetic modeling methods. CPT Pharmacomet Syst Pharmacol. 2013;2:e38. doi: 10.1038/psp.2013.14.
    1. Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development. CPT Pharmacomet Syst Pharmacol. 2012;1(9):e6. doi: 10.1038/psp.2012.4.
    1. Beal SS, Boeckmann L, Sheiner LB. NONMEM User’s Guides. (1989–2009). Gaithersburg, MD: ICON Development Solutions; 2009. Corpus ID: 65385267. . Accessed 20 Jan 2021.
    1. R Core Team. R: A Language and Environment for Statistical Computing. Vienna: R Foundation for Statistical Computing; 2019. Available at: . Accessed 20 Jan 2021.
    1. Beal SL. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn. 2001;28(5):481–504. doi: 10.1023/A:1012299115260.
    1. Kim K, Johnson JA, Derendorf H. Differences in drug pharmacokinetics between East Asians and Caucasians and the role of genetic polymorphisms. J Clin Pharmacol. 2004;44(10):1083–1105. doi: 10.1177/0091270004268128.
    1. Krekels EHJ, Rower JE, Constance JE, Knibbe CAJ, Sherwin CMT. Hepatic drug metabolism in pediatric patients. In: Xie W, editor. Drug metabolism in diseases. Boston: Academic Press; 2017. pp. 181–206.
    1. de Wildt SN, Tibboel D, Leeder JS. Drug metabolism for the paediatrician. Arch Dis Child. 2014;99(12):1137–1142. doi: 10.1136/archdischild-2013-305212.
    1. Kaye JL. Review of paediatric gastrointestinal physiology data relevant to oral drug delivery. Int J Clin Pharm. 2011;33(1):20–24. doi: 10.1007/s11096-010-9455-0.
    1. Morgan ET. Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clin Pharmacol Ther. 2009;85(4):434–438. doi: 10.1038/clpt.2008.302.
    1. Tanino T, Komada A, Ueda K, Bando T, Nojiri Y, Ueda Y, et al. Pharmacokinetics and differential regulation of cytochrome P450 enzymes in type 1 allergic mice. Drug Metab Dispos. 2016;44(12):1950–1957. doi: 10.1124/dmd.116.072462.
    1. Ma G, Xie R, Strober B, Langley R, Ito K, Krishnaswami S, et al. Pharmacokinetic characteristics of tofacitinib in adult patients with moderate to severe chronic plaque psoriasis. Clin Pharmacol Drug Dev. 2018;7(6):587–596. doi: 10.1002/cpdd.471.
    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(4):528–539. doi: 10.1002/jcph.1550.
    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(8):977–988. doi: 10.1007/s40262-017-0605-6.

Source: PubMed

3
Tilaa