دورية أكاديمية

Creation of Novel Sensitive Probe Substrate and Moderate Inhibitor Models for a Comprehensive Prediction of CYP2C8 Interactions for Tucatinib.

التفاصيل البيبلوغرافية
العنوان: Creation of Novel Sensitive Probe Substrate and Moderate Inhibitor Models for a Comprehensive Prediction of CYP2C8 Interactions for Tucatinib.
المؤلفون: Templeton IE; Certara, UK Ltd. (Simcyp Division), Sheffield, UK., Rowland-Yeo K; Certara, UK Ltd. (Simcyp Division), Sheffield, UK., Jones HM; Certara, UK Ltd. (Simcyp Division), Sheffield, UK., Endres CJ; Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington, USA., Topletz-Erickson AR; Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington, USA., Sun H; Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington, USA., Lee AJ; Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington, USA.
المصدر: Clinical pharmacology and therapeutics [Clin Pharmacol Ther] 2024 Feb; Vol. 115 (2), pp. 299-308. Date of Electronic Publication: 2023 Dec 08.
نوع المنشور: Journal Article; Research Support, Non-U.S. Gov't
اللغة: English
بيانات الدورية: Publisher: Wiley Country of Publication: United States NLM ID: 0372741 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1532-6535 (Electronic) Linking ISSN: 00099236 NLM ISO Abbreviation: Clin Pharmacol Ther Subsets: MEDLINE
أسماء مطبوعة: Publication: 2015- : Hoboken, NJ : Wiley
Original Publication: St. Louis : C.V. Mosby
مواضيع طبية MeSH: Gemfibrozil*/pharmacokinetics , Cytochrome P-450 CYP2C8 Inhibitors* , Oxazoles* , Pyridines* , Quinazolines*, Humans ; Cytochrome P-450 CYP3A ; Cytochrome P-450 CYP2C8 ; Cytochrome P-450 CYP2C9 ; Drug Interactions ; Models, Biological ; Cytochrome P-450 CYP3A Inhibitors
مستخلص: A physiologically-based pharmacokinetic (PBPK) model was developed to simulate plasma concentrations of tucatinib (TUKYSA®) after single-dose or multiple-dose administration of 300 mg b.i.d. orally. This PBPK model was subsequently applied to support evaluation of drug-drug interaction (DDI) risk as a perpetrator resulting from tucatinib inhibition of CYP3A4, CYP2C8, CYP2C9, P-gp, or MATE1/2-K. The PBPK model was also applied to support evaluation of DDI risk as a victim resulting from co-administration with CYP3A4 or CYP2C8 inhibitors, or a CYP3A4 inducer. After refinement with clinical DDI data, the final PBPK model was able to recover the clinically observed single and multiple-dose plasma concentrations for tucatinib when tucatinib was administered as a single agent in healthy subjects. In addition, the final model was able to recover clinically observed plasma concentrations of tucatinib when administered in combination with itraconazole, rifampin, or gemfibrozil as well as clinically observed plasma concentrations of probe substrates of CYP3A4, CYP2C8, CYP2C9, P-gp, or MATE1/2-K. The PBPK model was then applied to prospectively predict the potential perpetrator or victim DDIs with other substrates, inducers, or inhibitors. To simulate a potential interaction with a moderate CYP2C8 inhibitor, two novel PBPK models representing a moderate CYP2C8 inhibitor and a sensitive CYP2C8 substrate were developed based on the existing PBPK models for gemfibrozil and rosiglitazone, respectively. The simulated population geometric mean area under the curve ratio of tucatinib with a moderate CYP2C8 inhibitor ranged from 1.98- to 3.08-fold, and based on these results, no dose modifications were proposed for moderate CYP2C8 inhibitors for the tucatinib label.
(© 2023 Seagen Inc. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
References: US Food and Drug Administration. TUKYSA (tucatinib) Label <https://www.accessdata.fda.gov/drugsatfda&#95;docs/label/2023/213411s004lbl.pdf> (2023). Accessed January 19 2023.
Sun, H. et al. Elimination of tucatinib, a small molecule kinase inhibitor of HER2, is primarily governed by CYP2C8 enantioselective oxidation of gem-dimethyl. Cancer Chemother. Pharmacol. 89, 737-750 (2022).
Topletz-Erickson, A.R. et al. The pharmacokinetics and safety of tucatinib in volunteers with hepatic impairment. Clin. Pharmacokinet. 61, 1761-1770 (2022).
Topletz-Erickson, A. et al. Evaluation of safety and clinically relevant drug-drug interactions with tucatinib in healthy volunteers. Clin. Pharmacokinet. 61, 1417-1426 (2022).
Topletz-Erickson, A.R. et al. Tucatinib inhibits renal transporters OCT2 and MATE without impacting renal function in healthy subjects. J. Clin. Pharmacol. 61, 461-471 (2021).
Yeo, K.R., Kenny, J.R. & Rostami-Hodjegan, A. Application of in vitro-in vivo extrapolation (IVIVE) and physiologically based pharmacokinetic (PBPK) modelling to investigate the impact of the CYP2C8 polymorphism on rosiglitazone exposure. Eur. J. Clin. Pharmacol. 69, 1311-1320 (2013).
Baldwin, S.J., Clarke, S.E. & Chenery, R.J. Characterization of the cytochrome P450 enzymes involved in the in vitro metabolism of rosiglitazone. Br. J. Clin. Pharmacol. 48, 424-432 (1999).
US Food and Drug Administration. Drug Development and Drug Interactions. Table of Substrates, Inhibitors and Inducers. <https://www.fda.gov/drugs/drug-interactions-labeling/drug-development-and-drug-interactions-table-substrates-inhibitors-and-inducers>. Accessed August 24, 2022.
Wagner, C. et al. Predicting the effect of cytochrome P450 inhibitors on substrate drugs: analysis of physiologically based pharmacokinetic modeling submissions to the US Food and Drug Administration. Clin. Pharmacokinet. 54, 117-127 (2015).
Kilford, P.J., Gertz, M., Houston, J.B. & Galetin, A. Hepatocellular binding of drugs: correction for unbound fraction in hepatocyte incubations using microsomal binding or drug lipophilicity data. Drug Metab. Dispos. 36, 1194-1197 (2008).
VandenBrink, B.M., Foti, R.S., Rock, D.A., Wienkers, L.C. & Wahlstrom, J.L. Evaluation of CYP2C8 inhibition in vitro: utility of montelukast as a selective CYP2C8 probe substrate. Drug Metab. Dispos. 39, 1546-1554 (2011).
Greupink, R., Schreurs, M., Benne, M.S., Huisman, M.T. & Russel, F.G. Semi-mechanistic physiologically-based pharmacokinetic modeling of clinical glibenclamide pharmacokinetics and drug-drug-interactions. Eur. J. Pharm. Sci. 49, 819-828 (2013).
Hsu, V. et al. Towards quantitation of the effects of renal impairment and probenecid inhibition on kidney uptake and efflux transporters, using physiologically based pharmacokinetic modelling and simulations. Clin. Pharmacokinet. 53, 283-293 (2014).
Li, R., Barton, H.A. & Varma, M.V. Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved. Clin. Pharmacokinet. 53, 659-678 (2014).
Varma, M.V., Lai, Y., Feng, B., Litchfield, J., Goosen, T.C. & Bergman, A. Physiologically based modeling of pravastatin transporter-mediated hepatobiliary disposition and drug-drug interactions. Pharm. Res. 29, 2860-2873 (2012).
Zamek-Gliszczynski, M.J. et al. ITC recommendations for transporter kinetic parameter estimation and translational modeling of transport-mediated PK and DDIs in humans. Clin. Pharmacol. Ther. 94, 64-79 (2013).
Shebley, M., Fu, W., Badri, P., Bow, D. & Fischer, V. Physiologically based pharmacokinetic modeling suggests limited drug-drug interaction between Clopidogrel and Dasabuvir. Clin. Pharmacol. Ther. 102, 679-687 (2017).
Tornio, A. et al. Glucuronidation converts clopidogrel to a strong time-dependent inhibitor of CYP2C8: a phase II metabolite as a perpetrator of drug-drug interactions. Clin. Pharmacol. Ther. 96, 498-507 (2014).
Varma, M.V.S., Bi, Y.A., Lazzaro, S. & West, M. Clopidogrel as a perpetrator of drug-drug interactions: a challenge for quantitative predictions? Clin. Pharmacol. Ther. 105, 1295-1299 (2019).
Gan, J. et al. Repaglinide-gemfibrozil drug interaction: inhibition of repaglinide glucuronidation as a potential additional contributing mechanism. Br. J. Clin. Pharmacol. 70, 870-880 (2010).
معلومات مُعتمدة: Seagen Inc.
المشرفين على المادة: 234248D0HH (tucatinib)
Q8X02027X3 (Gemfibrozil)
0 (Cytochrome P-450 CYP2C8 Inhibitors)
EC 1.14.14.1 (Cytochrome P-450 CYP3A)
EC 1.14.14.1 (Cytochrome P-450 CYP2C8)
EC 1.14.13.- (Cytochrome P-450 CYP2C9)
0 (Cytochrome P-450 CYP3A Inhibitors)
EC 1.14.14.1 (CYP2C8 protein, human)
0 (Oxazoles)
0 (Pyridines)
0 (Quinazolines)
تواريخ الأحداث: Date Created: 20231116 Date Completed: 20240123 Latest Revision: 20240411
رمز التحديث: 20240412
DOI: 10.1002/cpt.3104
PMID: 37971208
قاعدة البيانات: MEDLINE
الوصف
تدمد:1532-6535
DOI:10.1002/cpt.3104