PBPK modeling to predict the pharmacokinetics of venlafaxine and its active metabolite in different CYP2D6 genotypes and drug–drug interactions with clarithromycin and paroxetine

IF 6.9 3区 医学 Q1 CHEMISTRY, MEDICINAL Archives of Pharmacal Research Pub Date : 2024-04-25 DOI:10.1007/s12272-024-01495-0
Chang-Keun Cho, Pureum Kang, Choon-Gon Jang, Seok-Yong Lee, Yun Jeong Lee, Jung-Woo Bae, Chang-Ik Choi
{"title":"PBPK modeling to predict the pharmacokinetics of venlafaxine and its active metabolite in different CYP2D6 genotypes and drug–drug interactions with clarithromycin and paroxetine","authors":"Chang-Keun Cho,&nbsp;Pureum Kang,&nbsp;Choon-Gon Jang,&nbsp;Seok-Yong Lee,&nbsp;Yun Jeong Lee,&nbsp;Jung-Woo Bae,&nbsp;Chang-Ik Choi","doi":"10.1007/s12272-024-01495-0","DOIUrl":null,"url":null,"abstract":"<div><p>Venlafaxine, a serotonin-norepinephrine reuptake inhibitor (SNRI), is indicated for the treatment of major depressive disorder, social anxiety disorder, generalized anxiety disorder, and panic disorder. Venlafaxine is metabolized to the active metabolite desvenlafaxine mainly by CYP2D6. Genetic polymorphism of <i>CYP2D6</i> and coadministration with other medications can significantly affect the pharmacokinetics and/or pharmacodynamics of venlafaxine and its active metabolite. This study aimed to establish the PBPK models of venlafaxine and its active metabolite related to <i>CYP2D6</i> genetic polymorphism and to predict drug–drug interactions (DDIs) with clarithromycin and paroxetine in different <i>CYP2D6</i> genotypes. Clinical pharmacogenomic data for venlafaxine and desvenlafaxine were collected to build the PBPK model. Physicochemical and absorption, distribution, metabolism, and excretion (ADME) characteristics of respective compounds were obtained from previously reported data, predicted by the PK-Sim<sup>®</sup> software, or optimized to capture the plasma concentration–time profiles. Model evaluation was performed by comparing the predicted pharmacokinetic parameters and plasma concentration–time profiles to the observed data. Predicted plasma concentration–time profiles of venlafaxine and its active metabolite were visually similar to the observed profiles and all predicted AUC and C<sub>max</sub> values for respective compounds were included in the twofold error range of observed values in non-genotyped populations and different <i>CYP2D6</i> genotypes. When clarithromycin or clarithromycin plus paroxetine was concomitantly administered, predicted plasma concentration–time profiles of venlafaxine properly captured the observed profiles in two different <i>CYP2D6</i> genotypes and all predicted DDI ratios for AUC and C<sub>max</sub> were included within the acceptance range. Consequently, the present model successfully captured the pharmacokinetic alterations of venlafaxine and its active metabolite according to <i>CYP2D6</i> genetic polymorphism as well as the DDIs between venlafaxine and two CYP inhibitors. The present model can be used to predict the pharmacokinetics of venlafaxine and its active metabolite considering different races, ages, coadministered drugs, and CYP2D6 activity of individuals and it can contribute to individualized pharmacotherapy of venlafaxine.</p></div>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":"47 5","pages":"481 - 504"},"PeriodicalIF":6.9000,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Archives of Pharmacal Research","FirstCategoryId":"3","ListUrlMain":"https://link.springer.com/article/10.1007/s12272-024-01495-0","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MEDICINAL","Score":null,"Total":0}
引用次数: 0

Abstract

Venlafaxine, a serotonin-norepinephrine reuptake inhibitor (SNRI), is indicated for the treatment of major depressive disorder, social anxiety disorder, generalized anxiety disorder, and panic disorder. Venlafaxine is metabolized to the active metabolite desvenlafaxine mainly by CYP2D6. Genetic polymorphism of CYP2D6 and coadministration with other medications can significantly affect the pharmacokinetics and/or pharmacodynamics of venlafaxine and its active metabolite. This study aimed to establish the PBPK models of venlafaxine and its active metabolite related to CYP2D6 genetic polymorphism and to predict drug–drug interactions (DDIs) with clarithromycin and paroxetine in different CYP2D6 genotypes. Clinical pharmacogenomic data for venlafaxine and desvenlafaxine were collected to build the PBPK model. Physicochemical and absorption, distribution, metabolism, and excretion (ADME) characteristics of respective compounds were obtained from previously reported data, predicted by the PK-Sim® software, or optimized to capture the plasma concentration–time profiles. Model evaluation was performed by comparing the predicted pharmacokinetic parameters and plasma concentration–time profiles to the observed data. Predicted plasma concentration–time profiles of venlafaxine and its active metabolite were visually similar to the observed profiles and all predicted AUC and Cmax values for respective compounds were included in the twofold error range of observed values in non-genotyped populations and different CYP2D6 genotypes. When clarithromycin or clarithromycin plus paroxetine was concomitantly administered, predicted plasma concentration–time profiles of venlafaxine properly captured the observed profiles in two different CYP2D6 genotypes and all predicted DDI ratios for AUC and Cmax were included within the acceptance range. Consequently, the present model successfully captured the pharmacokinetic alterations of venlafaxine and its active metabolite according to CYP2D6 genetic polymorphism as well as the DDIs between venlafaxine and two CYP inhibitors. The present model can be used to predict the pharmacokinetics of venlafaxine and its active metabolite considering different races, ages, coadministered drugs, and CYP2D6 activity of individuals and it can contribute to individualized pharmacotherapy of venlafaxine.

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
通过 PBPK 模型预测文拉法辛及其活性代谢物在不同 CYP2D6 基因型中的药代动力学,以及与克拉霉素和帕罗西汀的药物相互作用。
文拉法辛是一种血清素-去甲肾上腺素再摄取抑制剂(SNRI),适用于治疗重度抑郁症、社交焦虑症、广泛性焦虑症和恐慌症。文拉法辛主要通过 CYP2D6 代谢为活性代谢物去文拉法辛。CYP2D6 的基因多态性以及与其他药物合用会显著影响文拉法辛及其活性代谢物的药代动力学和/或药效学。本研究旨在建立文拉法辛及其活性代谢物与CYP2D6基因多态性相关的PBPK模型,并预测不同CYP2D6基因型的患者与克拉霉素和帕罗西汀的药物相互作用(DDI)。为建立 PBPK 模型,收集了文拉法辛和去文拉法辛的临床药理基因组学数据。各化合物的理化特性以及吸收、分布、代谢和排泄(ADME)特性均来自于先前报告的数据、PK-Sim® 软件预测的数据或为捕捉血浆浓度-时间曲线而优化的数据。通过将预测的药代动力学参数和血浆浓度-时间曲线与观察到的数据进行比较,对模型进行评估。预测的文拉法辛及其活性代谢物的血浆浓度-时间曲线与观察到的曲线直观相似,而且在非基因分型人群和不同 CYP2D6 基因分型中,各自化合物的所有预测 AUC 值和 Cmax 值都在观察值的两倍误差范围内。当同时服用克拉霉素或克拉霉素加帕罗西汀时,预测的文拉法辛血浆浓度-时间曲线正确捕捉到了两种不同 CYP2D6 基因型人群的观察曲线,并且所有预测的 AUC 和 Cmax 的 DDI 比率都包含在接受范围内。因此,本模型成功地捕捉到了文拉法辛及其活性代谢物随 CYP2D6 基因多态性而发生的药代动力学变化,以及文拉法辛与两种 CYP 抑制剂之间的 DDI。本模型可用于预测文拉法辛及其活性代谢物的药代动力学,同时考虑到不同种族、年龄、共用药物和个体的 CYP2D6 活性,有助于文拉法辛的个体化药物治疗。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
13.40
自引率
9.00%
发文量
48
审稿时长
3.3 months
期刊介绍: Archives of Pharmacal Research is the official journal of the Pharmaceutical Society of Korea and has been published since 1976. Archives of Pharmacal Research is an interdisciplinary journal devoted to the publication of original scientific research papers and reviews in the fields of drug discovery, drug development, and drug actions with a view to providing fundamental and novel information on drugs and drug candidates.
期刊最新文献
Saponins as potential novel NLRP3 inflammasome inhibitors for inflammatory disorders. Modulating versatile pathways using a cleavable PEG shell and EGFR-targeted nanoparticles to deliver CRISPR-Cas9 and docetaxel for triple-negative breast cancer inhibition. Ginsenoside Rg3 activates the immune function of CD8+ T cells via circFOXP1-miR-4477a-PD-L1 axis to induce ferroptosis in gallbladder cancer. Potential effects of a human milk oligosaccharide 6'-sialyllactose on angiotensin II-induced aortic aneurysm via p90RSK/TGF-β/SMAD2 signaling pathway. Akt-activated GSK3β inhibitory peptide effectively blocks tau hyperphosphorylation.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1