Clinical Trial Data-Driven Risk Assessment of Drug-Drug Interactions: A Rapid and Accurate Decision-Making Tool.

IF 4.6 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Clinical Pharmacokinetics Pub Date : 2024-08-01 Epub Date: 2024-08-05 DOI:10.1007/s40262-024-01404-0
Tong Yuan, Fulin Bi, Kuan Hu, Yuqi Zhu, Yan Lin, Jin Yang
{"title":"Clinical Trial Data-Driven Risk Assessment of Drug-Drug Interactions: A Rapid and Accurate Decision-Making Tool.","authors":"Tong Yuan, Fulin Bi, Kuan Hu, Yuqi Zhu, Yan Lin, Jin Yang","doi":"10.1007/s40262-024-01404-0","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>In clinical practice, the vast array of potential drug combinations necessitates swift and accurate assessments of pharmacokinetic drug-drug interactions (DDIs), along with recommendations for adjustments. Current methodologies for clinical DDI evaluations primarily rely on basic extrapolations from clinical trial data. However, these methods are limited in accuracy owing to their lack of a comprehensive consideration of various critical factors, including the inhibitory potency, dosage, and type of the inhibitor, as well as the metabolic fraction and intestinal availability of the substrate.</p><p><strong>Objective: </strong>This study aims to propose an efficient and accurate clinical pharmacokinetic-mediated DDI assessment tool, which comprehensively considers the effects of inhibitory potency and dosage of inhibitors, intestinal availability and fraction metabolized of substrates on DDI outcomes.</p><p><strong>Methods: </strong>This study focuses on DDIs caused by cytochrome P450 3A4 enzyme inhibition, utilizing extensive clinical trial data to establish a methodology to calculate the metabolic fraction and intestinal availability for substrates, as well as the concentration and inhibitory potency for inhibitors ( <math><msub><mi>K</mi> <mtext>i</mtext></msub> </math> or <math> <mrow><msub><mi>k</mi> <mtext>inact</mtext></msub> <mo>/</mo> <msub><mi>K</mi> <mtext>I</mtext></msub> </mrow> </math> ). These parameters were then used to predict the outcomes of DDIs involving 33 substrates and 20 inhibitors. We also defined the risk index for substrates and the potency index for inhibitors to establish a clinical DDI risk scale. The training set for parameter calculation consisted of 73 clinical trials. The validation set comprised 89 clinical DDI trials involving 53 drugs. which was used to evaluate the reliability of in vivo values of <math><msub><mtext>K</mtext> <mtext>i</mtext></msub> </math> and <math> <mrow><msub><mi>k</mi> <mtext>inact</mtext></msub> <mo>/</mo> <msub><mi>K</mi> <mtext>I</mtext></msub> </mrow> </math> , the accuracy of DDI predictions, and the false-negative rate of risk scale.</p><p><strong>Results: </strong>First, the reliability of the in vivo <math><msub><mi>K</mi> <mtext>i</mtext></msub> </math> and <math> <mrow><msub><mi>k</mi> <mtext>inact</mtext></msub> <mo>/</mo> <msub><mi>K</mi> <mtext>I</mtext></msub> </mrow> </math> values calculated in this study was assessed using a basic static model. Compared with values obtained from other methods, this study values showed a lower geometric mean fold error and root mean square error. Additionally, incorporating these values into the physiologically based pharmacokinetic-DDI model facilitated a good fitting of the C-t curves when the substrate's metabolic enzymes are inhibited. Second, area under the curve ratio predictions of studied drugs were within a 1.5 × margin of error in 81% of cases compared with clinical observations in the validation set. Last, the clinical DDI risk scale developed in this study predicted the actual risks in the validation set with only a 5.6% incidence of serious false negatives.</p><p><strong>Conclusions: </strong>This study offers a rapid and accurate approach for assessing the risk of pharmacokinetic-mediated DDIs in clinical practice, providing a foundation for rational combination drug use and dosage adjustments.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":null,"pages":null},"PeriodicalIF":4.6000,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacokinetics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s40262-024-01404-0","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/8/5 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: In clinical practice, the vast array of potential drug combinations necessitates swift and accurate assessments of pharmacokinetic drug-drug interactions (DDIs), along with recommendations for adjustments. Current methodologies for clinical DDI evaluations primarily rely on basic extrapolations from clinical trial data. However, these methods are limited in accuracy owing to their lack of a comprehensive consideration of various critical factors, including the inhibitory potency, dosage, and type of the inhibitor, as well as the metabolic fraction and intestinal availability of the substrate.

Objective: This study aims to propose an efficient and accurate clinical pharmacokinetic-mediated DDI assessment tool, which comprehensively considers the effects of inhibitory potency and dosage of inhibitors, intestinal availability and fraction metabolized of substrates on DDI outcomes.

Methods: This study focuses on DDIs caused by cytochrome P450 3A4 enzyme inhibition, utilizing extensive clinical trial data to establish a methodology to calculate the metabolic fraction and intestinal availability for substrates, as well as the concentration and inhibitory potency for inhibitors ( K i or k inact / K I ). These parameters were then used to predict the outcomes of DDIs involving 33 substrates and 20 inhibitors. We also defined the risk index for substrates and the potency index for inhibitors to establish a clinical DDI risk scale. The training set for parameter calculation consisted of 73 clinical trials. The validation set comprised 89 clinical DDI trials involving 53 drugs. which was used to evaluate the reliability of in vivo values of K i and k inact / K I , the accuracy of DDI predictions, and the false-negative rate of risk scale.

Results: First, the reliability of the in vivo K i and k inact / K I values calculated in this study was assessed using a basic static model. Compared with values obtained from other methods, this study values showed a lower geometric mean fold error and root mean square error. Additionally, incorporating these values into the physiologically based pharmacokinetic-DDI model facilitated a good fitting of the C-t curves when the substrate's metabolic enzymes are inhibited. Second, area under the curve ratio predictions of studied drugs were within a 1.5 × margin of error in 81% of cases compared with clinical observations in the validation set. Last, the clinical DDI risk scale developed in this study predicted the actual risks in the validation set with only a 5.6% incidence of serious false negatives.

Conclusions: This study offers a rapid and accurate approach for assessing the risk of pharmacokinetic-mediated DDIs in clinical practice, providing a foundation for rational combination drug use and dosage adjustments.

Abstract Image

临床试验数据驱动的药物相互作用风险评估:快速准确的决策工具。
背景:在临床实践中,由于潜在的药物组合种类繁多,因此有必要对药物动力学上的药物相互作用(DDIs)进行迅速而准确的评估,并提出调整建议。目前临床 DDI 评估的方法主要依赖于临床试验数据的基本推断。然而,这些方法的准确性有限,因为它们没有全面考虑各种关键因素,包括抑制剂的抑制效力、剂量和类型,以及底物的代谢率和肠道可用性:本研究旨在提出一种高效、准确的临床药代动力学介导的 DDI 评估工具,该工具可综合考虑抑制剂的抑制效力和剂量、底物的肠道可利用性和代谢率对 DDI 结果的影响:本研究的重点是细胞色素 P450 3A4 酶抑制引起的 DDI,利用大量临床试验数据建立了一种方法来计算底物的代谢率和肠道利用率,以及抑制剂的浓度和抑制效力(K i 或 k inact / K I)。然后利用这些参数来预测涉及 33 种底物和 20 种抑制剂的 DDI 结果。我们还定义了底物的风险指数和抑制剂的效力指数,以建立临床 DDI 风险量表。参数计算的训练集包括 73 项临床试验。验证集由涉及 53 种药物的 89 项临床 DDI 试验组成,用于评估 K i 和 k inact / K I 体内值的可靠性、DDI 预测的准确性以及风险量表的假阴性率:首先,使用基本静态模型评估了本研究计算的体内 K i 和 k inact / K I 值的可靠性。与其他方法得出的数值相比,本研究的数值显示出较低的几何平均折叠误差和均方根误差。此外,当底物的代谢酶受到抑制时,将这些值纳入基于生理学的药代动力学-DDI 模型有助于很好地拟合 C-t 曲线。其次,与验证集的临床观察结果相比,所研究药物的曲线下面积比预测值有 81% 的误差在 1.5 × 误差范围内。最后,本研究开发的临床 DDI 风险量表预测了验证集中的实际风险,而严重假阴性的发生率仅为 5.6%:本研究为评估临床实践中药动学介导的 DDI 风险提供了一种快速、准确的方法,为合理使用联合用药和调整剂量奠定了基础。
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 求助全文
来源期刊
CiteScore
8.80
自引率
4.40%
发文量
86
审稿时长
6-12 weeks
期刊介绍: Clinical Pharmacokinetics promotes the continuing development of clinical pharmacokinetics and pharmacodynamics for the improvement of drug therapy, and for furthering postgraduate education in clinical pharmacology and therapeutics. Pharmacokinetics, the study of drug disposition in the body, is an integral part of drug development and rational use. Knowledge and application of pharmacokinetic principles leads to accelerated drug development, cost effective drug use and a reduced frequency of adverse effects and drug interactions.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信