Evaluation of the Effect of Abrocitinib on Drug Transporters by Integrated Use of Probe Drugs and Endogenous Biomarkers

Manoli Vourvahis, Wonkyung Byon, Cheng Chang, Vu Le, Annette Diehl, Daniela Graham, Sakambari Tripathy, Nancy Raha, Lina Luo, Sumathy Mathialagan, Martin Dowty, A David Rodrigues, Bimal Malhotra, Manoli Vourvahis, Wonkyung Byon, Cheng Chang, Vu Le, Annette Diehl, Daniela Graham, Sakambari Tripathy, Nancy Raha, Lina Luo, Sumathy Mathialagan, Martin Dowty, A David Rodrigues, Bimal Malhotra

Abstract

Abrocitinib is an oral Janus kinase 1 (JAK1) inhibitor currently approved in the United Kingdom for the treatment of moderate-to-severe atopic dermatitis (AD). As patients with AD may use medications to manage comorbidities, abrocitinib could be used concomitantly with hepatic and/or renal transporter substrates. Therefore, we assessed the potential effect of abrocitinib on probe drugs and endogenous biomarker substrates for the drug transporters of interest. In vitro studies indicated that, among the transporters tested, abrocitinib has the potential to inhibit the activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporter 3 (OAT3), organic cation transporter 1 (OCT1), and multidrug and toxin extrusion protein 1 and 2K (MATE1/2K). Therefore, subsequent phase I, two-way crossover, open-label studies in healthy participants were performed to assess the impact of abrocitinib on the pharmacokinetics of the transporter probe substrates dabigatran etexilate (P-gp), rosuvastatin (BCRP and OAT3), and metformin (OCT2 and MATE1/2K), as well as endogenous biomarkers for MATE1/2K (N1 -methylnicotinamide (NMN)) and OCT1 (isobutyryl-L -carnitine (IBC)). Co-administration with abrocitinib was shown to increase the plasma exposure of dabigatran by ~ 50%. In comparison, the plasma exposure and renal clearance of rosuvastatin and metformin were not altered with abrocitinib co-administration. Similarly, abrocitinib did not affect the exposure of NMN or IBC. An increase in dabigatran exposure suggests that abrocitinib inhibits P-gp activity. By contrast, a lack of impact on plasma exposure and/or renal clearance of rosuvastatin, metformin, NMN, or IBC suggests that BCRP, OAT3, OCT1, and MATE1/2K activity are unaffected by abrocitinib.

Trial registration: ClinicalTrials.gov NCT03742336 NCT03806101 NCT03796182.

Conflict of interest statement

All authors are employees and stockholders of Pfizer Inc.

© 2022 Pfizer Inc. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Treatment schedule for clinical pharmacokinetic studies. All studies were randomized, with a two‐way crossover design. In each study, participants were randomized 1:1 into 1 of 2 treatment sequences, in which treatment A was followed by treatment B or vice versa, with a washout period between treatments.
Figure 2
Figure 2
Mean (+ SD) plasma concentration‐time curves for (a) dabigatran, (b) rosuvastatin, and (c) metformin alone and with abrocitinib, with semi‐log scale plasma concentration‐time curves inset in top right corners. QD, once‐daily.
Figure 3
Figure 3
Mean (+ SD) plasma concentration‐time curves for (a) NMN and (b) IBC.a Panel (c) shows a scatterplot of individual ratios for metformin CLr vs. NMN CLr.b CLr, renal clearance; IBC, isobutyryl‐l‐carnitine; NMN, N1‐methylnicotinamide; QD, once daily. aLog plots were not generated as NMN and IBC are endogenous compounds; bOnly 10 of the 12 study participants are included in this figure and contributed to the Pearson correlation coefficient calculation due to missing metformin CLr or NMN CLr values for metformin + abrocitinib treatment period.

References

    1. Silverberg, J.I. et al. Patient burden and quality of life in atopic dermatitis in US adults: a population‐based cross‐sectional study. Ann. Allergy Asthma Immunol. 121, 340–347 (2018).
    1. Brunner, P.M. , Guttman‐Yassky, E. & Leung, D.Y. The immunology of atopic dermatitis and its reversibility with broad‐spectrum and targeted therapies. J. Allergy Clin. Immunol. 139, S65–S76 (2017).
    1. Japan's MHLW Approves Pfizer's Cibinqo® (abrocitinib) for Adults and Adolescents with Moderate to Severe Atopic Dermatitis . News release. Published September 30, 2021. PFE) today announced,and older with inadequate response. Accessed April 26, 2022.
    1. UK's MHRA Grants Marketing Authorisation for Pfizer's Cibinqo® (abrocitinib) for Adults and Adolescents with Moderate‐to‐Severe Atopic Dermatitis . News release. Published September 9, 2021. PFE) today announced,AD) in adults and adolescents. Accessed April 26, 2022.
    1. European Commission Approves Pfizer's Cibinqo® (abrocitinib) for the Treatment of Adults with Moderate‐to‐Severe Atopic Dermatitis . News release. Published December 10, 2021. . Accessed April 26, 2022.
    1. FDA Approves Pfizer’s Cibinqo® (abrocitinib) for Adults with Moderate‐to‐Severe Atopic Dermatitis . News release. Published January 14, 2022. . Accessed April 26, 2022.
    1. Bieber, T. et al. Abrocitinib versus placebo or dupilumab for atopic dermatitis. N. Engl. J. Med. 384, 1101–1112 (2021).
    1. Nezamololama, N. , Crowley, E.L. , Gooderham, M.J. & Papp, K. Abrocitinib: a potential treatment for moderate‐to‐severe atopic dermatitis. Expert Opin. Investig. Drugs 29, 911–917 (2020).
    1. Crowley, E.L. , Nezamololama, N. , Papp, K. & Gooderham, M.J. Abrocitinib for the treatment of atopic dermatitis. Expert Rev. Clin. Immunol. 16, 955–962 (2020).
    1. Silverberg, J.I. Comorbidities and the impact of atopic dermatitis. Ann. Allergy Asthma Immunol. 123, 144–151 (2019).
    1. Liang, Y. , Li, S. & Chen, L. The physiological role of drug transporters. Protein Cell 6, 334–350 (2015).
    1. US Department of Health and Human Services . In vitro drug interaction studies — Cytochrome P450 enzyme‐ and transporter‐mediated drug interactions guidance for industry. Published January 2020. . Accessed February 14, 2022.
    1. Giacomini, K.M. et al. International Transporter Consortium commentary on clinically important transporter polymorphisms. Clin. Pharmacol. Ther. 94, 23–26 (2013).
    1. Tornio, A. , Filppula, A.M. , Niemi, M. & Backman, J.T. Clinical studies on drug‐drug interactions involving metabolism and transport: methodology, pitfalls, and interpretation. Clin. Pharmacol. Ther. 105, 1345–1361 (2019).
    1. Mariappan, T.T. , Shen, H. & Marathe, P. Endogenous biomarkers to assess drug‐drug interactions by drug transporters and enzymes. Curr. Drug Metab. 18, 757–768 (2017).
    1. Luo, L. et al. A multiplexed HILIC‐MS/HRMS assay for the assessment of transporter inhibition biomarkers in phase i clinical trials: isobutyryl‐carnitine as an organic cation transporter (OCT1) biomarker. Anal. Chem. 92, 9745–9754 (2020).
    1. Chu, X. et al. Clinical probes and endogenous biomarkers as substrates for transporter drug‐drug interaction evaluation: perspectives from the international transporter consortium. Clin. Pharmacol. Ther. 104, 836–864 (2018).
    1. Miyake, T. et al. Identification of appropriate endogenous biomarker for risk assessment of multidrug and toxin extrusion protein‐mediated drug‐drug interactions in healthy volunteers. Clin. Pharmacol. Ther. 109, 507–516 (2021).
    1. Mori, D. et al. Dose‐dependent inhibition of OATP1B by rifampicin in healthy volunteers: comprehensive evaluation of candidate biomarkers and OATP1B probe drugs. Clin. Pharmacol. Ther. 107, 1004–1013 (2020).
    1. Luo, L. et al. LC‐MS/MS assay for N(1)‐methylnicotinamide in humans, an endogenous probe for renal transporters. Bioanalysis 10, 673–689 (2018).
    1. Müller, F. et al. N(1)‐methylnicotinamide as an endogenous probe for drug interactions by renal cation transporters: studies on the metformin‐trimethoprim interaction. Eur. J. Clin. Pharmacol. 71, 85–94 (2015).
    1. Jensen, O. , Matthaei, J. , Klemp, H.G. , Meyer, M.J. , Brockmöller, J. & Tzvetkov, M.V. Isobutyrylcarnitine as a biomarker of OCT1 activity and interspecies differences in its membrane transport. Front. Pharmacol. 12, 674559 (2021).
    1. Matthaei, J. et al. Effects of genetic polymorphism in CYP2D6, CYP2C19, and the organic cation transporter OCT1 on amitriptyline pharmacokinetics in healthy volunteers and depressive disorder patients. Front. Pharmacol. 12, 688950 (2021).
    1. Stangier, J. & Clemens, A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin. Appl. Thromb. Hemost. 15(Suppl. 1), 9s–16s (2009).
    1. Boehringer Ingelheim Pharmaceuticals Inc. Pradaxa® (Dabigatran Etexilate Mesylate) Capsules for Oral Use (Initial U.S., Ridgefield, CT, 2014).
    1. AstraZeneca . Crestor (Rosuvastatin Calcium) Tablets (AstraZeneca, Osaka, Japan, 2015).
    1. Windass, A.S. , Lowes, S. , Wang, Y. & Brown, C.D. The contribution of organic anion transporters OAT1 and OAT3 to the renal uptake of rosuvastatin. J. Pharmacol. Exp. Ther. 322, 1221–1227 (2007).
    1. Stage, T.B. , Brøsen, K. & Christensen, M.M. A comprehensive review of drug‐drug interactions with metformin. Clin. Pharmacokinet. 54, 811–824 (2015).
    1. Kimura, N. et al. Metformin is a superior substrate for renal organic cation transporter OCT2 rather than hepatic OCT1. Drug Metab. Pharmacokinet. 20, 379–386 (2005).
    1. Bristol‐Meyers Squibb Company . Glucophage (metformin hydrochloride) Tablets, for Oral Use. Glucophage XR (metformin hydrochloride) Extended‐Release Tablets, for Oral Use (Bristol‐Meyers Squibb Company, Princeton, NJ, 2018).
    1. Zamek‐Gliszczynski, M.J. et al. ITC commentary on metformin clinical drug‐drug interaction study design that enables an efficacy‐ and safety‐based dose adjustment decision. Clin. Pharmacol. Ther. 104, 781–784 (2018).
    1. European Medicines Agency EMA . Guideline on the investigation of drug interactions. Published June 21, 2012. . Accessed April 16, 2021.
    1. Keskitalo, J.E. , Zolk, O. , Fromm, M.F. , Kurkinen, K.J. , Neuvonen, P.J. & Niemi, M. ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin. Clin. Pharmacol. Ther. 86, 197–203 (2009).
    1. McTaggart, F. et al. Preclinical and clinical pharmacology of Rosuvastatin, a new 3‐hydroxy‐3‐methylglutaryl coenzyme A reductase inhibitor. Am. J. Cardiol. 87, 28b–32b (2001).
    1. Wiebe, S.T. et al. Validation of a drug transporter probe cocktail using the prototypical inhibitors rifampin, probenecid, verapamil, and cimetidine. Clin. Pharmacokinet. 59, 1627–1639 (2020).
    1. Zhou, T. , Arya, V. & Zhang, L. Comparing various in vitro prediction methods to assess the potential of a drug to Inhibit P‐glycoprotein (P‐gp) transporter in vivo. J. Clin. Pharmacol. 59, 1049–1060 (2019).
    1. Costales, C. et al. Quantitative prediction of breast cancer resistant protein mediated drug‐drug interactions using physiologically‐based pharmacokinetic modeling. CPT Pharmacometrics Syst. Pharmacol. 10, 1018–1031 (2021).
    1. Kim, H.I. et al. Fine mapping and functional analysis reveal a role of SLC22A1 in acylcarnitine transport. Am. J. Hum. Genet. 101, 489–502 (2017).
    1. Ramsay, R.R. , Gandour, R.D. & van der Leij, F.R. Molecular enzymology of carnitine transfer and transport. Biochim. Biophys. Acta 1546, 21–43 (2001).
    1. Shen, H. , Yao, M. , Sinz, M. , Marathe, P. , Rodrigues, A.D. & Zhu, M. Renal excretion of dabigatran: the potential role of Multidrug and Toxin Extrusion (MATE) proteins. Mol. Pharm. 16, 4065–4076 (2019).
    1. Stangier, J. , Rathgen, K. , Stähle, H. , Gansser, D. & Roth, W. The pharmacokinetics, pharmacodynamics and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in healthy male subjects. Br. J. Clin. Pharmacol. 64, 292–303 (2007).
    1. Beveridge, T. , Nüesch, E. & Ohnhaus, E.E. Absolute bioavailability of digoxin tablets. Arzneimittelforschung 28, 701–703 (1978).
    1. Manninen, V. , Reissell, P. & Ojala, K. Maximal bioavailability of digoxin from tablets and oral solution in steady state. Acta Med. Scand. 199, 487–489 (1976).
    1. Binnion, P.F. A comparison of the bioavailability of digoxin in capsule, tablet, and solution taken orally with intravenous digoxin. J. Clin. Pharmacol. 16, 461–467 (1976).
    1. US Food and Drug Administration . In vitro metabolism and transporter‐mediated drug‐drug interaction studies. Guidance for industry. Published October 2017. . Accessed April 16, 2021.
    1. Vaidyanathan, J. , Yoshida, K. , Arya, V. & Zhang, L. Comparing various in vitro prediction criteria to assess the potential of a new molecular entity to inhibit organic anion transporting polypeptide 1B1. J. Clin. Pharmacol. 56(Suppl. 7), S59–S72 (2016).
    1. Paine, M.F. , Ludington, S.S. , Chen, M.L. , Stewart, P.W. , Huang, S.M. & Watkins, P.B. Do men and women differ in proximal small intestinal CYP3A or P‐glycoprotein expression? Drug Metab. Dispos. 33, 426–433 (2005).
    1. Fernandez, C. , Buyse, M. , German‐Fattal, M. & Gimenez, F. Influence of the pro‐inflammatory cytokines on P‐glycoprotein expression and functionality. J. Pharm. Pharm. Sci. 7, 359–371 (2004).

Source: PubMed

3
Suscribir