在非洲患者群体中优化治疗方法的模型启发法。

IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Clinical Pharmacology & Therapeutics Pub Date : 2024-08-20 DOI:10.1002/cpt.3425
Mwila Mulubwa, Kelly Chibale
{"title":"在非洲患者群体中优化治疗方法的模型启发法。","authors":"Mwila Mulubwa,&nbsp;Kelly Chibale","doi":"10.1002/cpt.3425","DOIUrl":null,"url":null,"abstract":"<p>Pharmacokinetics variability arises from individual differences in how drugs are absorbed, distributed, metabolized, and excreted by the body. Pharmacokinetic variations can be influenced by several intrinsic and extrinsic factors, including genetic polymorphisms, weight, sex, age, disease state, or polypharmacy.<span><sup>1</sup></span> Genetic factors such as polymorphisms in drug metabolizing enzymes can account for 20 to 95 percent patient variability in response to individual pharmacotherapy.<span><sup>1</sup></span></p><p>Understanding the genetic diversity in genes encoding for drug-metabolizing enzymes is essential in facilitating the development of more effective therapeutics tailored to diverse populations. Africa exhibits higher genetic diversity than other populations worldwide, a characteristic that extends to variability within genes encoding the cytochrome P450 enzyme system and drug transporter proteins.<span><sup>2</sup></span> It is noteworthy that genetic variability in African populations is not representative of that in the African American population and therefore cannot be assumed to be similar. Variations in gene sequence ultimately translate into decreased, normal, increased, or no enzyme function. This gives rise to metabolic phenotypes such as poor metabolizers, normal metabolizers, and fast metabolizers, which are associated with adverse drug reactions, suboptimal treatment efficacy, and poor treatment outcomes, respectively. This genetic variability has contributed to the high incidences of adverse drug reactions and poor therapeutic outcomes, especially in tuberculosis (TB) and human immunodeficiency virus (HIV) cases reported in Africa.<span><sup>2</sup></span> Hence, variations in pharmacokinetics pose challenges and create disparities in treatment outcomes among the genetically diverse African populations.</p><p>These challenges are compounded by lack of sufficiently powered clinical pharmacokinetics or pharmacogenetics studies to accurately quantify the influence of the prevalent pharmacogenetic variants on drug exposure in most African populations.<span><sup>3</sup></span> In addition, despite Africa accounting for around 15% of the global population, it remains understudied and largely absent from biomedical research. Therefore, it is indispensable to adopt model-informed strategies to address disparities in therapeutic outcomes among African patient populations. These approaches involve the application of data-driven models to guide decision-making and optimize therapeutic interventions for African patient populations. In the absence of African-derived biobanks, this requires the building or refining of existing physiologically based pharmacokinetic (PBPK) models that factor in African genetic variants predicted from artificial intelligence (AI) as illustrated in <b>Figure</b> 1. Here, we propose <i>in vitro</i> and <i>in silico</i> methods that may help explain pharmacokinetic variability and inform dose regimen optimization. By incorporating information such as genetic factors, comorbidities such as TB infection, and treatment response patterns, healthcare providers can tailor therapies to the specific needs of African patients.</p><p>Tuberculosis (TB) is a serious infectious disease that is caused by <i>Mycobacterium tuberculosis</i> (<i>Mtb</i>) and mostly affects not only the lungs but also other organs. Treatment typically involves a combination of antibiotics taken over a long period (4–6 months for drug sensitive TB and 6–20 months for drug-resistant TB) to eliminate the bacteria and prevent the development of drug-resistant strains. The burden of <i>Mtb</i> is high, particularly in low- and middle-income countries of Africa where co-morbidities are prevalent. The burden of noncommunicable diseases (NCDs) in Africa such as diabetes and cardiovascular diseases are a growing concern, accounting for over 30% of the continent's mortality. These NCDs also account for the most common comorbidities in tuberculosis patients. The impact of comorbid conditions on the PK of drugs used to treat <i>Mtb</i> is a crucial area of study, as these interactions can potentially affect treatment outcomes.<span><sup>4</sup></span> Understanding how comorbidities such as hypertension, diabetes and HIV infection can influence the PK is essential for optimizing therapeutic regimens. Given the high burden of <i>Mtb</i> coinfections worldwide, it is important to explore the complex interplay between the <i>Mtb</i> infection and other comorbidities to develop more effective treatment strategies.</p><p>Metabolism of drugs is mostly attributed to liver enzymes particularly the cytochrome P450s. <i>Mtb</i> possesses its own set of enzymes (<b>Table</b> 1) capable of catalyzing biotransformation reactions on xenobiotics, including some antituberculosis drugs. These enzymes enable <i>Mtb</i> to modulate the activity of drugs through various mechanisms, such as deactivation or activation of drugs, ultimately influencing the effectiveness of TB treatment.<span><sup>5, 6</sup></span> The extent of <i>Mtb</i> drug metabolism is dependent on the bacterial load. It was shown in a large cohort of African patients receiving first-line antituberculosis drugs (isoniazid, rifampicin, pyrazinamide, and ethambutol) that <i>Mtb</i> bacterial load persisted to an average amount of 2 log<sub>10</sub> CFU/mL after 12 weeks of treatment from an average baseline of 6 log<sub>10</sub> CFU/mL.<span><sup>7</sup></span> In the case of a drug that is metabolized by <i>Mtb</i>, it is expected for drug clearance to decrease or normalize after a longer duration of treatment. Hence, <i>Mtb</i>-mediated drug metabolism may potentially impact the efficacy and treatment outcomes of tuberculosis or other comorbidities whose drug treatment may be prone to <i>Mtb</i>-mediated metabolism.</p><p>Designing optimum treatment regimens using nonlinear-mixed effects (NLME) modeling and simulation represents an advanced approach to treatment optimization. The NLME modeling approach allows for the incorporation and modeling of <i>Mtb</i>-mediated metabolism or genetic factors as covariates into these mathematical models, enabling the exploration of complex pharmacokinetic scenarios. By leveraging pharmacokinetic data predicted from PBPK models, NLME modeling can characterize the variability in drug exposure and identify factors such as genetic factors, demographics, and disease status such as TB as significant covariates that influence treatment outcomes. Simulation techniques based on NLME models enable the exploration of different dosing strategies, treatment durations, and combination therapies <i>in silico</i>,<span><sup>10</sup></span> providing valuable insights into the optimal design of treatment regimens for specific African patient populations.</p><p>Improving therapeutic outcomes entails administration of the right dose or dose regimen. Model-informed approaches offer a promising avenue for optimizing therapeutics in the African patient population. By building PBPK models using African liver subcellular fractions or AI-predicted prevalent African gene variants (drug metabolizing enzymes) and integrating <i>Mtb</i>-mediated drug metabolism into PBPK models we can predict and capture pharmacokinetic variability. The NLME modeling of PBPK simulated pharmacokinetic profiles enhance the understanding of pharmacogenetic variability, which enables the development of more tailored and precise treatment regimens, minimizing the risk of adverse drug reactions and optimizing therapeutic outcomes among African populations.</p><p>K.C. is the Neville Isdell Chair in African-centric Drug Discovery and Development and thanks Neville Isdell for generously funding the Chair.</p><p>The authors declared no competing interests for this work.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"116 6","pages":"1391-1394"},"PeriodicalIF":6.3000,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3425","citationCount":"0","resultStr":"{\"title\":\"Model-Informed Approaches to Optimizing Therapeutics in the African Patient Populations\",\"authors\":\"Mwila Mulubwa,&nbsp;Kelly Chibale\",\"doi\":\"10.1002/cpt.3425\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Pharmacokinetics variability arises from individual differences in how drugs are absorbed, distributed, metabolized, and excreted by the body. Pharmacokinetic variations can be influenced by several intrinsic and extrinsic factors, including genetic polymorphisms, weight, sex, age, disease state, or polypharmacy.<span><sup>1</sup></span> Genetic factors such as polymorphisms in drug metabolizing enzymes can account for 20 to 95 percent patient variability in response to individual pharmacotherapy.<span><sup>1</sup></span></p><p>Understanding the genetic diversity in genes encoding for drug-metabolizing enzymes is essential in facilitating the development of more effective therapeutics tailored to diverse populations. Africa exhibits higher genetic diversity than other populations worldwide, a characteristic that extends to variability within genes encoding the cytochrome P450 enzyme system and drug transporter proteins.<span><sup>2</sup></span> It is noteworthy that genetic variability in African populations is not representative of that in the African American population and therefore cannot be assumed to be similar. Variations in gene sequence ultimately translate into decreased, normal, increased, or no enzyme function. This gives rise to metabolic phenotypes such as poor metabolizers, normal metabolizers, and fast metabolizers, which are associated with adverse drug reactions, suboptimal treatment efficacy, and poor treatment outcomes, respectively. This genetic variability has contributed to the high incidences of adverse drug reactions and poor therapeutic outcomes, especially in tuberculosis (TB) and human immunodeficiency virus (HIV) cases reported in Africa.<span><sup>2</sup></span> Hence, variations in pharmacokinetics pose challenges and create disparities in treatment outcomes among the genetically diverse African populations.</p><p>These challenges are compounded by lack of sufficiently powered clinical pharmacokinetics or pharmacogenetics studies to accurately quantify the influence of the prevalent pharmacogenetic variants on drug exposure in most African populations.<span><sup>3</sup></span> In addition, despite Africa accounting for around 15% of the global population, it remains understudied and largely absent from biomedical research. Therefore, it is indispensable to adopt model-informed strategies to address disparities in therapeutic outcomes among African patient populations. These approaches involve the application of data-driven models to guide decision-making and optimize therapeutic interventions for African patient populations. In the absence of African-derived biobanks, this requires the building or refining of existing physiologically based pharmacokinetic (PBPK) models that factor in African genetic variants predicted from artificial intelligence (AI) as illustrated in <b>Figure</b> 1. Here, we propose <i>in vitro</i> and <i>in silico</i> methods that may help explain pharmacokinetic variability and inform dose regimen optimization. By incorporating information such as genetic factors, comorbidities such as TB infection, and treatment response patterns, healthcare providers can tailor therapies to the specific needs of African patients.</p><p>Tuberculosis (TB) is a serious infectious disease that is caused by <i>Mycobacterium tuberculosis</i> (<i>Mtb</i>) and mostly affects not only the lungs but also other organs. Treatment typically involves a combination of antibiotics taken over a long period (4–6 months for drug sensitive TB and 6–20 months for drug-resistant TB) to eliminate the bacteria and prevent the development of drug-resistant strains. The burden of <i>Mtb</i> is high, particularly in low- and middle-income countries of Africa where co-morbidities are prevalent. The burden of noncommunicable diseases (NCDs) in Africa such as diabetes and cardiovascular diseases are a growing concern, accounting for over 30% of the continent's mortality. These NCDs also account for the most common comorbidities in tuberculosis patients. The impact of comorbid conditions on the PK of drugs used to treat <i>Mtb</i> is a crucial area of study, as these interactions can potentially affect treatment outcomes.<span><sup>4</sup></span> Understanding how comorbidities such as hypertension, diabetes and HIV infection can influence the PK is essential for optimizing therapeutic regimens. Given the high burden of <i>Mtb</i> coinfections worldwide, it is important to explore the complex interplay between the <i>Mtb</i> infection and other comorbidities to develop more effective treatment strategies.</p><p>Metabolism of drugs is mostly attributed to liver enzymes particularly the cytochrome P450s. <i>Mtb</i> possesses its own set of enzymes (<b>Table</b> 1) capable of catalyzing biotransformation reactions on xenobiotics, including some antituberculosis drugs. These enzymes enable <i>Mtb</i> to modulate the activity of drugs through various mechanisms, such as deactivation or activation of drugs, ultimately influencing the effectiveness of TB treatment.<span><sup>5, 6</sup></span> The extent of <i>Mtb</i> drug metabolism is dependent on the bacterial load. It was shown in a large cohort of African patients receiving first-line antituberculosis drugs (isoniazid, rifampicin, pyrazinamide, and ethambutol) that <i>Mtb</i> bacterial load persisted to an average amount of 2 log<sub>10</sub> CFU/mL after 12 weeks of treatment from an average baseline of 6 log<sub>10</sub> CFU/mL.<span><sup>7</sup></span> In the case of a drug that is metabolized by <i>Mtb</i>, it is expected for drug clearance to decrease or normalize after a longer duration of treatment. Hence, <i>Mtb</i>-mediated drug metabolism may potentially impact the efficacy and treatment outcomes of tuberculosis or other comorbidities whose drug treatment may be prone to <i>Mtb</i>-mediated metabolism.</p><p>Designing optimum treatment regimens using nonlinear-mixed effects (NLME) modeling and simulation represents an advanced approach to treatment optimization. The NLME modeling approach allows for the incorporation and modeling of <i>Mtb</i>-mediated metabolism or genetic factors as covariates into these mathematical models, enabling the exploration of complex pharmacokinetic scenarios. By leveraging pharmacokinetic data predicted from PBPK models, NLME modeling can characterize the variability in drug exposure and identify factors such as genetic factors, demographics, and disease status such as TB as significant covariates that influence treatment outcomes. Simulation techniques based on NLME models enable the exploration of different dosing strategies, treatment durations, and combination therapies <i>in silico</i>,<span><sup>10</sup></span> providing valuable insights into the optimal design of treatment regimens for specific African patient populations.</p><p>Improving therapeutic outcomes entails administration of the right dose or dose regimen. Model-informed approaches offer a promising avenue for optimizing therapeutics in the African patient population. By building PBPK models using African liver subcellular fractions or AI-predicted prevalent African gene variants (drug metabolizing enzymes) and integrating <i>Mtb</i>-mediated drug metabolism into PBPK models we can predict and capture pharmacokinetic variability. The NLME modeling of PBPK simulated pharmacokinetic profiles enhance the understanding of pharmacogenetic variability, which enables the development of more tailored and precise treatment regimens, minimizing the risk of adverse drug reactions and optimizing therapeutic outcomes among African populations.</p><p>K.C. is the Neville Isdell Chair in African-centric Drug Discovery and Development and thanks Neville Isdell for generously funding the Chair.</p><p>The authors declared no competing interests for this work.</p>\",\"PeriodicalId\":153,\"journal\":{\"name\":\"Clinical Pharmacology & Therapeutics\",\"volume\":\"116 6\",\"pages\":\"1391-1394\"},\"PeriodicalIF\":6.3000,\"publicationDate\":\"2024-08-20\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3425\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical Pharmacology & Therapeutics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3425\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3425","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

在一大批接受一线抗结核药物(异烟肼、利福平、吡嗪酰胺和乙胺丁醇)治疗的非洲患者中显示,Mtb细菌负荷从平均基线6 log10 CFU/mL持续到治疗12周后的平均2 log10 CFU/mL。因此,Mtb 介导的药物代谢可能会对结核病或其他合并症的疗效和治疗结果产生潜在影响,而结核病或其他合并症的药物治疗可能易受 Mtb 介导的代谢影响。非线性混合效应建模方法允许将 Mtb 介导的代谢或遗传因素作为协变量纳入这些数学模型并进行建模,从而能够探索复杂的药代动力学方案。通过利用 PBPK 模型预测的药代动力学数据,NLME 模型可以描述药物暴露的变异性,并确定遗传因素、人口统计学和结核病等疾病状态等因素是影响治疗结果的重要协变量。基于 NLME 模型的仿真技术可以对不同的给药策略、治疗持续时间和联合疗法进行硅学探索,10 为特定非洲患者群体的最佳治疗方案设计提供有价值的见解。以模型为依据的方法为优化非洲患者群体的治疗方案提供了一条前景广阔的途径。通过使用非洲肝脏亚细胞组分或 AI 预测的非洲流行基因变异(药物代谢酶)建立 PBPK 模型,并将 Mtb 介导的药物代谢纳入 PBPK 模型,我们可以预测并捕捉药代动力学变异。PBPK模拟药代动力学特征的NLME建模增强了对药物基因变异性的理解,从而能够开发出更有针对性和更精确的治疗方案,最大限度地降低药物不良反应的风险,优化非洲人群的治疗效果。K.C.是以非洲为中心的药物发现和开发领域的Neville Isdell讲席教授,感谢Neville Isdell对该讲席教授的慷慨资助。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Model-Informed Approaches to Optimizing Therapeutics in the African Patient Populations

Pharmacokinetics variability arises from individual differences in how drugs are absorbed, distributed, metabolized, and excreted by the body. Pharmacokinetic variations can be influenced by several intrinsic and extrinsic factors, including genetic polymorphisms, weight, sex, age, disease state, or polypharmacy.1 Genetic factors such as polymorphisms in drug metabolizing enzymes can account for 20 to 95 percent patient variability in response to individual pharmacotherapy.1

Understanding the genetic diversity in genes encoding for drug-metabolizing enzymes is essential in facilitating the development of more effective therapeutics tailored to diverse populations. Africa exhibits higher genetic diversity than other populations worldwide, a characteristic that extends to variability within genes encoding the cytochrome P450 enzyme system and drug transporter proteins.2 It is noteworthy that genetic variability in African populations is not representative of that in the African American population and therefore cannot be assumed to be similar. Variations in gene sequence ultimately translate into decreased, normal, increased, or no enzyme function. This gives rise to metabolic phenotypes such as poor metabolizers, normal metabolizers, and fast metabolizers, which are associated with adverse drug reactions, suboptimal treatment efficacy, and poor treatment outcomes, respectively. This genetic variability has contributed to the high incidences of adverse drug reactions and poor therapeutic outcomes, especially in tuberculosis (TB) and human immunodeficiency virus (HIV) cases reported in Africa.2 Hence, variations in pharmacokinetics pose challenges and create disparities in treatment outcomes among the genetically diverse African populations.

These challenges are compounded by lack of sufficiently powered clinical pharmacokinetics or pharmacogenetics studies to accurately quantify the influence of the prevalent pharmacogenetic variants on drug exposure in most African populations.3 In addition, despite Africa accounting for around 15% of the global population, it remains understudied and largely absent from biomedical research. Therefore, it is indispensable to adopt model-informed strategies to address disparities in therapeutic outcomes among African patient populations. These approaches involve the application of data-driven models to guide decision-making and optimize therapeutic interventions for African patient populations. In the absence of African-derived biobanks, this requires the building or refining of existing physiologically based pharmacokinetic (PBPK) models that factor in African genetic variants predicted from artificial intelligence (AI) as illustrated in Figure 1. Here, we propose in vitro and in silico methods that may help explain pharmacokinetic variability and inform dose regimen optimization. By incorporating information such as genetic factors, comorbidities such as TB infection, and treatment response patterns, healthcare providers can tailor therapies to the specific needs of African patients.

Tuberculosis (TB) is a serious infectious disease that is caused by Mycobacterium tuberculosis (Mtb) and mostly affects not only the lungs but also other organs. Treatment typically involves a combination of antibiotics taken over a long period (4–6 months for drug sensitive TB and 6–20 months for drug-resistant TB) to eliminate the bacteria and prevent the development of drug-resistant strains. The burden of Mtb is high, particularly in low- and middle-income countries of Africa where co-morbidities are prevalent. The burden of noncommunicable diseases (NCDs) in Africa such as diabetes and cardiovascular diseases are a growing concern, accounting for over 30% of the continent's mortality. These NCDs also account for the most common comorbidities in tuberculosis patients. The impact of comorbid conditions on the PK of drugs used to treat Mtb is a crucial area of study, as these interactions can potentially affect treatment outcomes.4 Understanding how comorbidities such as hypertension, diabetes and HIV infection can influence the PK is essential for optimizing therapeutic regimens. Given the high burden of Mtb coinfections worldwide, it is important to explore the complex interplay between the Mtb infection and other comorbidities to develop more effective treatment strategies.

Metabolism of drugs is mostly attributed to liver enzymes particularly the cytochrome P450s. Mtb possesses its own set of enzymes (Table 1) capable of catalyzing biotransformation reactions on xenobiotics, including some antituberculosis drugs. These enzymes enable Mtb to modulate the activity of drugs through various mechanisms, such as deactivation or activation of drugs, ultimately influencing the effectiveness of TB treatment.5, 6 The extent of Mtb drug metabolism is dependent on the bacterial load. It was shown in a large cohort of African patients receiving first-line antituberculosis drugs (isoniazid, rifampicin, pyrazinamide, and ethambutol) that Mtb bacterial load persisted to an average amount of 2 log10 CFU/mL after 12 weeks of treatment from an average baseline of 6 log10 CFU/mL.7 In the case of a drug that is metabolized by Mtb, it is expected for drug clearance to decrease or normalize after a longer duration of treatment. Hence, Mtb-mediated drug metabolism may potentially impact the efficacy and treatment outcomes of tuberculosis or other comorbidities whose drug treatment may be prone to Mtb-mediated metabolism.

Designing optimum treatment regimens using nonlinear-mixed effects (NLME) modeling and simulation represents an advanced approach to treatment optimization. The NLME modeling approach allows for the incorporation and modeling of Mtb-mediated metabolism or genetic factors as covariates into these mathematical models, enabling the exploration of complex pharmacokinetic scenarios. By leveraging pharmacokinetic data predicted from PBPK models, NLME modeling can characterize the variability in drug exposure and identify factors such as genetic factors, demographics, and disease status such as TB as significant covariates that influence treatment outcomes. Simulation techniques based on NLME models enable the exploration of different dosing strategies, treatment durations, and combination therapies in silico,10 providing valuable insights into the optimal design of treatment regimens for specific African patient populations.

Improving therapeutic outcomes entails administration of the right dose or dose regimen. Model-informed approaches offer a promising avenue for optimizing therapeutics in the African patient population. By building PBPK models using African liver subcellular fractions or AI-predicted prevalent African gene variants (drug metabolizing enzymes) and integrating Mtb-mediated drug metabolism into PBPK models we can predict and capture pharmacokinetic variability. The NLME modeling of PBPK simulated pharmacokinetic profiles enhance the understanding of pharmacogenetic variability, which enables the development of more tailored and precise treatment regimens, minimizing the risk of adverse drug reactions and optimizing therapeutic outcomes among African populations.

K.C. is the Neville Isdell Chair in African-centric Drug Discovery and Development and thanks Neville Isdell for generously funding the Chair.

The authors declared no competing interests for this work.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
12.70
自引率
7.50%
发文量
290
审稿时长
2 months
期刊介绍: Clinical Pharmacology & Therapeutics (CPT) is the authoritative cross-disciplinary journal in experimental and clinical medicine devoted to publishing advances in the nature, action, efficacy, and evaluation of therapeutics. CPT welcomes original Articles in the emerging areas of translational, predictive and personalized medicine; new therapeutic modalities including gene and cell therapies; pharmacogenomics, proteomics and metabolomics; bioinformation and applied systems biology complementing areas of pharmacokinetics and pharmacodynamics, human investigation and clinical trials, pharmacovigilence, pharmacoepidemiology, pharmacometrics, and population pharmacology.
期刊最新文献
A Phase I Trial of the Pharmacokinetic Interaction Between Cannabidiol and Tacrolimus. Oxygenation Effects of Antihypertensive Agents in Intensive Care: A Prospective Comparative Study of Nicardipine and Urapidil. Discovering Severe Adverse Reactions From Pharmacokinetic Drug-Drug Interactions Through Literature Analysis and Electronic Health Record Verification. French-Speaking Network of Pharmacogenetics (RNPGx) Recommendations for Clinical Use of Mavacamten. Clinical and Physiologically Based Pharmacokinetic Model Evaluations of Adagrasib Drug-Drug Interactions.
×
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