首页 > 最新文献

CPT: Pharmacometrics & Systems Pharmacology最新文献

英文 中文
A Case Study of Model-Informed Drug Development of a Novel PCSK9 Antisense Oligonucleotide. Part 2: Phase 2 to Phase 3 一种新型PCSK9反义寡核苷酸基于模型的药物开发案例研究。第二部分:第二阶段至第三阶段。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-05 DOI: 10.1002/psp4.70076
Jane Knöchel, Catarina Nilsson, Björn Carlsson, Alexis Hofherr, Per Johanson, Tina Rydén-Bergsten, Bengt Hamrén, Dinko Rekić

In this second part of a case study on the practical use of model-informed drug development (MIDD), we describe the clinical development of AZD8233, a novel proprotein convertase subtilisin/kexin type 9 (PCSK9) antisense oligonucleotide, from phase 2b to the start of phase 3. The case study exemplifies the use of MIDD to answer key design questions for the phase 3 program, including the design of a pivotal phase 3 study, a head-to-head study, and a cardiovascular outcome study informed by model-averaging analysis. Extensive phase 3 study simulations assessed the impact of drop-out, readout timing, dose frequency, and analysis method on study outcomes. The final phase 3 design assumed around 1% monthly drop-out (based on other PCSK9 inhibitor trials), used an EMA/FDA-approved analysis method, and set the primary readout at week 16. A simulated study predicted a reduction in low-density lipoprotein cholesterol (LDL-C) by week 16 of −69% with AZD8233 60 mg every 4 weeks. A virtual head-to-head study showed AZD8233 lowered LDL-C by 27% more than an active competitor (inclisiran) at day 270. Predicted cardiovascular relative risk reduction (RRR) for AZD8233 on top of statins ranged from 24% to 49% based on model choice; a model-averaging approach predicted an RRR of 27% assuming 63% LDL-C reduction from a 130 mg/dL baseline. This case study highlights the importance of cross-functional collaboration and other key MIDD enablers to ensure that MIDD extends beyond a simple simulation exercise and is instead considered an integral part of drug development dedicated to quantitative decision making.

在模型信息药物开发(MIDD)实际应用案例研究的第二部分,我们描述了AZD8233的临床开发,这是一种新型蛋白转化酶枯草杆菌素/酮素9型(PCSK9)反义寡核苷酸,从2b期到3期开始。该案例研究举例说明了使用MIDD来回答3期项目的关键设计问题,包括关键3期研究的设计、头对头研究的设计以及通过模型平均分析得出的心血管结果研究。广泛的3期研究模拟评估了退出、读出时间、剂量频率和分析方法对研究结果的影响。最终的3期设计假设每月退出率约为1%(基于其他PCSK9抑制剂试验),使用EMA/ fda批准的分析方法,并在第16周设置主要读数。一项模拟研究预测,每4周服用60毫克AZD8233,到第16周时,低密度脂蛋白胆固醇(LDL-C)降低-69%。一项虚拟面对面研究显示,在第270天,AZD8233比一种活跃的竞争对手(包括伊朗)降低了27%的LDL-C。根据模型选择,AZD8233在他汀类药物的基础上预测心血管相对风险降低(RRR)范围为24%至49%;模型平均方法预测,假设LDL-C从130 mg/dL基线降低63%,RRR为27%。本案例研究强调了跨职能协作和其他关键MIDD推动者的重要性,以确保MIDD超越简单的模拟练习,而是被视为致力于定量决策的药物开发的一个组成部分。
{"title":"A Case Study of Model-Informed Drug Development of a Novel PCSK9 Antisense Oligonucleotide. Part 2: Phase 2 to Phase 3","authors":"Jane Knöchel,&nbsp;Catarina Nilsson,&nbsp;Björn Carlsson,&nbsp;Alexis Hofherr,&nbsp;Per Johanson,&nbsp;Tina Rydén-Bergsten,&nbsp;Bengt Hamrén,&nbsp;Dinko Rekić","doi":"10.1002/psp4.70076","DOIUrl":"10.1002/psp4.70076","url":null,"abstract":"<p>In this second part of a case study on the practical use of model-informed drug development (MIDD), we describe the clinical development of AZD8233, a novel proprotein convertase subtilisin/kexin type 9 (PCSK9) antisense oligonucleotide, from phase 2b to the start of phase 3. The case study exemplifies the use of MIDD to answer key design questions for the phase 3 program, including the design of a pivotal phase 3 study, a head-to-head study, and a cardiovascular outcome study informed by model-averaging analysis. Extensive phase 3 study simulations assessed the impact of drop-out, readout timing, dose frequency, and analysis method on study outcomes. The final phase 3 design assumed around 1% monthly drop-out (based on other PCSK9 inhibitor trials), used an EMA/FDA-approved analysis method, and set the primary readout at week 16. A simulated study predicted a reduction in low-density lipoprotein cholesterol (LDL-C) by week 16 of −69% with AZD8233 60 mg every 4 weeks. A virtual head-to-head study showed AZD8233 lowered LDL-C by 27% more than an active competitor (inclisiran) at day 270. Predicted cardiovascular relative risk reduction (RRR) for AZD8233 on top of statins ranged from 24% to 49% based on model choice; a model-averaging approach predicted an RRR of 27% assuming 63% LDL-C reduction from a 130 mg/dL baseline. This case study highlights the importance of cross-functional collaboration and other key MIDD enablers to ensure that MIDD extends beyond a simple simulation exercise and is instead considered an integral part of drug development dedicated to quantitative decision making.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1556-1561"},"PeriodicalIF":3.0,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70076","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144788476","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Predicting Systemic and Liver Bosentan Exposure Using Physiologically-Based Pharmacokinetic Modeling 使用基于生理的药代动力学模型预测全身和肝脏波生坦暴露。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-05 DOI: 10.1002/psp4.70085
Miao-Chan Huang, Julia Macente, Sofie Heylen, Chen Ning, Kristof De Vos, Neel Deferm, Pieter Annaert

Bosentan is the first approved oral medication for pulmonary arterial hypertension, yet the black-box warning on its labeling implies a substantial risk of liver injury associated with bosentan exposure. The risk assessment of bosentan-induced liver injury requires a thorough understanding of the underlying mechanisms, for which there is accumulating evidence. Integrating these mechanisms with clinical liver bosentan concentration would enable a more dynamic and relevant risk assessment. This study designed a workflow of physiologically−based pharmacokinetic (PBPK) model development to capture bosentan's hepatic disposition and predict the (intra)hepatic bosentan exposure. Specifically, clinical plasma and excretion data of bosentan were used to minimize the uncertainty in estimating the hepatic clearance. The model predictions were well overlapped with observations in the systemic circulation and excretion. Furthermore, the model-derived intrinsic hepatic clearance was comparable with the one derived from a clinical study. These results reflected confidence in the model's capability to predict hepatic bosentan exposure. The model-simulated steady-state unbound exposure to bosentan in hepatocytes and liver tissue ranged from 1.65 to 34.1 ng/mL following twice-daily 125-mg oral doses. The ratio of the simulated unbound concentration between the liver matrices and systemic plasma was between 0.80 and 2.93 across the therapeutic dosing regimens. In summary, a bosentan PBPK model was successfully developed with the designed workflow and was able to predict the hepatic disposition of bosentan. The developed model can be applied to generate hepatic bosentan exposure that bridges the toxicological mechanistic findings from in vitro to in vivo, assisting in risk assessment.

波生坦是首个被批准用于治疗肺动脉高压的口服药物,但其标签上的黑框警告意味着与波生坦暴露相关的肝损伤风险很大。波生坦诱导肝损伤的风险评估需要对其潜在机制有透彻的了解,这方面的证据越来越多。将这些机制与临床肝波生坦浓度相结合,将使风险评估更加动态和相关。本研究设计了一个基于生理的药代动力学(PBPK)模型开发工作流程,以捕获波生坦的肝脏配置并预测(体内)肝内波生坦暴露。具体来说,临床血浆和波生坦排泄数据被用来减少估计肝脏清除率的不确定性。模型预测与体循环和排泄的观察结果很好地重叠。此外,模型得出的内在肝脏清除率与临床研究得出的结果相当。这些结果反映了对模型预测肝波生坦暴露能力的信心。模型模拟的肝细胞和肝组织中波生坦的稳态非结合暴露范围为1.65至34.1 ng/mL,每日两次口服剂量为125 mg。肝基质与全身血浆的模拟未结合浓度之比在整个治疗给药方案中介于0.80和2.93之间。总之,根据设计的工作流程成功开发了波生坦PBPK模型,并能够预测波生坦的肝脏处置。开发的模型可以应用于产生肝波生坦暴露,连接从体外到体内的毒理学机制发现,协助风险评估。
{"title":"Predicting Systemic and Liver Bosentan Exposure Using Physiologically-Based Pharmacokinetic Modeling","authors":"Miao-Chan Huang,&nbsp;Julia Macente,&nbsp;Sofie Heylen,&nbsp;Chen Ning,&nbsp;Kristof De Vos,&nbsp;Neel Deferm,&nbsp;Pieter Annaert","doi":"10.1002/psp4.70085","DOIUrl":"10.1002/psp4.70085","url":null,"abstract":"<p>Bosentan is the first approved oral medication for pulmonary arterial hypertension, yet the black-box warning on its labeling implies a substantial risk of liver injury associated with bosentan exposure. The risk assessment of bosentan-induced liver injury requires a thorough understanding of the underlying mechanisms, for which there is accumulating evidence. Integrating these mechanisms with clinical liver bosentan concentration would enable a more dynamic and relevant risk assessment. This study designed a workflow of physiologically−based pharmacokinetic (PBPK) model development to capture bosentan's hepatic disposition and predict the (intra)hepatic bosentan exposure. Specifically, clinical plasma and excretion data of bosentan were used to minimize the uncertainty in estimating the hepatic clearance. The model predictions were well overlapped with observations in the systemic circulation and excretion. Furthermore, the model-derived intrinsic hepatic clearance was comparable with the one derived from a clinical study. These results reflected confidence in the model's capability to predict hepatic bosentan exposure. The model-simulated steady-state unbound exposure to bosentan in hepatocytes and liver tissue ranged from 1.65 to 34.1 ng/mL following twice-daily 125-mg oral doses. The ratio of the simulated unbound concentration between the liver matrices and systemic plasma was between 0.80 and 2.93 across the therapeutic dosing regimens. In summary, a bosentan PBPK model was successfully developed with the designed workflow and was able to predict the hepatic disposition of bosentan. The developed model can be applied to generate hepatic bosentan exposure that bridges the toxicological mechanistic findings from in vitro to in vivo, assisting in risk assessment.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1715-1726"},"PeriodicalIF":3.0,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70085","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144788478","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Pharmacometrics-Informed Trial Simulation Framework for Optimizing Study Designs for Disease-Modifying Treatments in Rare Neurological Disorders 一种基于药物计量学的试验模拟框架,用于优化罕见神经系统疾病改善治疗的研究设计。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-05 DOI: 10.1002/psp4.70082
Yevgen Ryeznik, Ralf-Dieter Hilgers, Nicole Heussen, Emmanuelle Comets, France Mentré, Niels Hendrickx, Mats O. Karlsson, Andrew C. Hooker, Alzahra Hamdan, Xiaomei Chen, Rebecca Schüle, Matthis Synofzik, Oleksandr Sverdlov

The development of new treatments for rare neurological diseases (RNDs) may be very challenging due to limited natural history data, lack of relevant biomarkers and clinical endpoints, small and heterogeneous patient populations, and other complexities. A systematic approach is needed for comparing various design and analysis strategies to identify “optimal” approaches for a clinical trial in a chosen RND with the given resource constraints. For this purpose, we propose a pharmacometrics-informed clinical scenario evaluation framework (CSE-PMx), which includes some important research hallmarks relevant to RND clinical trials: a disease progression model for simulating individual longitudinal outcomes, the choice of a suitable randomization method for trial design, and an option to perform subsequent statistical analysis with randomization tests. We illustrate the utility of CSE-PMx for an exemplary randomized trial to compare the disease-modifying effect of an experimental treatment versus control in patients with Autosomal-Recessive Spastic Ataxia Charlevoix Saguenay (ARSACS). In the considered example, our simulation evidence suggests that a nonlinear mixed-effects model (NLMEM) with a population-based likelihood ratio test analysis is valid, robust, and more powerful than some conventional methods such as two-sample t-test, analysis of covariance (ANCOVA), or a mixed model with repeated measurements (MMRM). Our proposed framework is very flexible and generalizable to clinical research in other rare disease indications.

由于自然病史数据有限,缺乏相关生物标志物和临床终点,患者群体小且异质性,以及其他复杂性,罕见神经疾病(rnd)新疗法的开发可能非常具有挑战性。需要一种系统的方法来比较各种设计和分析策略,以确定在给定资源限制下选定RND进行临床试验的“最佳”方法。为此,我们提出了一个基于药物计量学的临床情景评估框架(CSE-PMx),其中包括与RND临床试验相关的一些重要研究特征:模拟个体纵向结果的疾病进展模型,试验设计的合适随机化方法的选择,以及通过随机化试验进行后续统计分析的选项。我们在一项典型的随机试验中说明了CSE-PMx的效用,以比较常染色体隐性痉挛性共济失调(ARSACS)患者的实验性治疗与对照组的疾病改善效果。在考虑的示例中,我们的模拟证据表明,具有基于总体的似然比检验分析的非线性混合效应模型(NLMEM)是有效的,稳健的,并且比一些传统方法(如双样本t检验,协方差分析(ANCOVA)或具有重复测量的混合模型(MMRM))更强大。我们提出的框架非常灵活,可推广到其他罕见病适应症的临床研究。
{"title":"A Pharmacometrics-Informed Trial Simulation Framework for Optimizing Study Designs for Disease-Modifying Treatments in Rare Neurological Disorders","authors":"Yevgen Ryeznik,&nbsp;Ralf-Dieter Hilgers,&nbsp;Nicole Heussen,&nbsp;Emmanuelle Comets,&nbsp;France Mentré,&nbsp;Niels Hendrickx,&nbsp;Mats O. Karlsson,&nbsp;Andrew C. Hooker,&nbsp;Alzahra Hamdan,&nbsp;Xiaomei Chen,&nbsp;Rebecca Schüle,&nbsp;Matthis Synofzik,&nbsp;Oleksandr Sverdlov","doi":"10.1002/psp4.70082","DOIUrl":"10.1002/psp4.70082","url":null,"abstract":"<p>The development of new treatments for rare neurological diseases (RNDs) may be very challenging due to limited natural history data, lack of relevant biomarkers and clinical endpoints, small and heterogeneous patient populations, and other complexities. A systematic approach is needed for comparing various design and analysis strategies to identify “optimal” approaches for a clinical trial in a chosen RND with the given resource constraints. For this purpose, we propose a pharmacometrics-informed clinical scenario evaluation framework (CSE-PMx), which includes some important research hallmarks relevant to RND clinical trials: a disease progression model for simulating individual longitudinal outcomes, the choice of a suitable randomization method for trial design, and an option to perform subsequent statistical analysis with randomization tests. We illustrate the utility of CSE-PMx for an exemplary randomized trial to compare the disease-modifying effect of an experimental treatment versus control in patients with Autosomal-Recessive Spastic Ataxia Charlevoix Saguenay (ARSACS). In the considered example, our simulation evidence suggests that a nonlinear mixed-effects model (NLMEM) with a population-based likelihood ratio test analysis is valid, robust, and more powerful than some conventional methods such as two-sample <i>t</i>-test, analysis of covariance (ANCOVA), or a mixed model with repeated measurements (MMRM). Our proposed framework is very flexible and generalizable to clinical research in other rare disease indications.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1705-1714"},"PeriodicalIF":3.0,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70082","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144788477","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Model-Informed Precision Dosing of Infliximab in Korean Inflammatory Bowel Disease Patients: External Validation of Population Pharmacokinetic Models 韩国炎症性肠病患者英夫利昔单抗基于模型的精确剂量:群体药代动力学模型的外部验证。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-04 DOI: 10.1002/psp4.70089
Yoonjin Kim, Seung Hwan Baek, In-Jin Jang, Jae-Yong Chung

Underexposure to infliximab often leads to loss of response in patients with inflammatory bowel disease (IBD). Model-informed precision dosing (MIPD) offers a superior approach to maintaining target infliximab concentrations compared to empirical dosage adjustment. This study aims to externally validate the population pharmacokinetic (PK) models implemented in TDMx, an online MIPD dashboard system, for adult and pediatric Korean IBD patients before clinical use. This retrospective study included 199 IBD patients (142 adults, 57 children) treated with intravenous infliximab at Seoul National University Hospital (Seoul, Republic of Korea) from 2019 to 2023. Three adult and seven pediatric models were evaluated based on accuracy, precision, goodness of fit plots, prediction-corrected visual predictive checks, and normalized prediction distribution errors. For adults, the Passot model showed the best fit (mean percentage error (MPE) 26.4%, mean absolute error (MAE) 1.1 mg/L, relative root-mean square error (rRMSE) 159.0%), whereas all pediatric models were unsuitable for clinical use (MPE 30.4%–143.4%, MAE 1.4–2.6 mg/L, rRMSE 96.3%–564.0%). Predictive performance was compared between datasets with or without accurate information on antibodies-toward-infliximab (ATI), as well as with and without previous concentrations. Assuming all patients were ATI positive improved predictive performance, likely due to the inherent positive bias of the population PK models. Incorporating previous concentrations improved predictions for adult models, achieving acceptable accuracy and precision (Passot model: MPE 17.5%, MAE 1.8 mg/L, rRMSE 80.3% with one concentration). However, pediatric models remained clinically unacceptable, highlighting the need to develop models specifically tailored for this population.

英夫利昔单抗暴露不足经常导致炎症性肠病(IBD)患者的反应丧失。与经验剂量调整相比,模型信息精确给药(MIPD)提供了一种更好的方法来维持目标英夫利昔单抗浓度。本研究旨在外部验证在线MIPD仪表板系统TDMx中实现的韩国成人和儿童IBD患者临床使用前的群体药代动力学(PK)模型。这项回顾性研究包括2019年至2023年在首尔国立大学医院(韩国首尔)接受静脉注射英夫利昔单抗治疗的199例IBD患者(142名成人,57名儿童)。根据准确性、精密度、拟合优度、预测校正后的视觉预测检查和归一化预测分布误差对3个成人模型和7个儿童模型进行评估。对于成人,Passot模型最适合(平均百分比误差(MPE) 26.4%,平均绝对误差(MAE) 1.1 mg/L,相对均方根误差(rRMSE) 159.0%),而所有儿童模型都不适合临床使用(MPE 30.4% ~ 143.4%, MAE 1.4 ~ 2.6 mg/L, rRMSE 96.3% ~ 564.0%)。预测性能在具有或不具有针对英夫利昔单抗(ATI)抗体的准确信息的数据集之间以及具有和不具有先前浓度的数据集之间进行比较。假设所有患者均为ATI阳性,可提高预测性能,这可能是由于群体PK模型固有的正偏倚。结合以前的浓度改进了成人模型的预测,达到了可接受的准确度和精度(Passot模型:MPE 17.5%, MAE 1.8 mg/L, rRMSE 80.3%,一个浓度)。然而,儿科模型在临床上仍然是不可接受的,这突出了开发专门针对这一人群的模型的必要性。
{"title":"Model-Informed Precision Dosing of Infliximab in Korean Inflammatory Bowel Disease Patients: External Validation of Population Pharmacokinetic Models","authors":"Yoonjin Kim,&nbsp;Seung Hwan Baek,&nbsp;In-Jin Jang,&nbsp;Jae-Yong Chung","doi":"10.1002/psp4.70089","DOIUrl":"10.1002/psp4.70089","url":null,"abstract":"<p>Underexposure to infliximab often leads to loss of response in patients with inflammatory bowel disease (IBD). Model-informed precision dosing (MIPD) offers a superior approach to maintaining target infliximab concentrations compared to empirical dosage adjustment. This study aims to externally validate the population pharmacokinetic (PK) models implemented in TDMx, an online MIPD dashboard system, for adult and pediatric Korean IBD patients before clinical use. This retrospective study included 199 IBD patients (142 adults, 57 children) treated with intravenous infliximab at Seoul National University Hospital (Seoul, Republic of Korea) from 2019 to 2023. Three adult and seven pediatric models were evaluated based on accuracy, precision, goodness of fit plots, prediction-corrected visual predictive checks, and normalized prediction distribution errors. For adults, the Passot model showed the best fit (mean percentage error (MPE) 26.4%, mean absolute error (MAE) 1.1 mg/L, relative root-mean square error (rRMSE) 159.0%), whereas all pediatric models were unsuitable for clinical use (MPE 30.4%–143.4%, MAE 1.4–2.6 mg/L, rRMSE 96.3%–564.0%). Predictive performance was compared between datasets with or without accurate information on antibodies-toward-infliximab (ATI), as well as with and without previous concentrations. Assuming all patients were ATI positive improved predictive performance, likely due to the inherent positive bias of the population PK models. Incorporating previous concentrations improved predictions for adult models, achieving acceptable accuracy and precision (Passot model: MPE 17.5%, MAE 1.8 mg/L, rRMSE 80.3% with one concentration). However, pediatric models remained clinically unacceptable, highlighting the need to develop models specifically tailored for this population.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1682-1694"},"PeriodicalIF":3.0,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70089","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144774862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transforming Pediatric Rare Disease Drug Development: Enhancing Clinical Trials and Regulatory Evidence With Virtual Patients 转变儿科罕见病药物开发:加强虚拟患者的临床试验和监管证据。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-04 DOI: 10.1002/psp4.70096
Fianne Sips, Marco Virgolin, Giuseppe Pasculli, Federico Reali, Alessio Paris, Annette Janus, Yann Godfrin, Daniel Röshammar, Luca Marchetti, Jane Knöchel

Drug development in pediatric rare diseases is complicated by practical and ethical constraints on clinical trial design, stemming from small, highly heterogeneous, and vulnerable patient populations. Virtual patients (VPs) created with machine-learning (ML), mechanistically driven computational approaches, or hybrids thereof, have the potential to expedite and maximize the impact of trials. We discuss the potential of VPs to transform the efficiency and impact of clinical trials in pediatric rare diseases, based on adult and pediatric examples.

儿童罕见病的药物开发由于临床试验设计的实际和伦理限制而变得复杂,这些限制源于小的、高度异质的和脆弱的患者群体。使用机器学习(ML)、机械驱动的计算方法或其混合方法创建的虚拟患者(vp)有可能加速并最大化试验的影响。我们以成人和儿童为例,讨论了副总裁在改变儿科罕见病临床试验效率和影响方面的潜力。
{"title":"Transforming Pediatric Rare Disease Drug Development: Enhancing Clinical Trials and Regulatory Evidence With Virtual Patients","authors":"Fianne Sips,&nbsp;Marco Virgolin,&nbsp;Giuseppe Pasculli,&nbsp;Federico Reali,&nbsp;Alessio Paris,&nbsp;Annette Janus,&nbsp;Yann Godfrin,&nbsp;Daniel Röshammar,&nbsp;Luca Marchetti,&nbsp;Jane Knöchel","doi":"10.1002/psp4.70096","DOIUrl":"10.1002/psp4.70096","url":null,"abstract":"<p>Drug development in pediatric rare diseases is complicated by practical and ethical constraints on clinical trial design, stemming from small, highly heterogeneous, and vulnerable patient populations. Virtual patients (VPs) created with machine-learning (ML), mechanistically driven computational approaches, or hybrids thereof, have the potential to expedite and maximize the impact of trials. We discuss the potential of VPs to transform the efficiency and impact of clinical trials in pediatric rare diseases, based on adult and pediatric examples.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 11","pages":"1739-1742"},"PeriodicalIF":3.0,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70096","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144774863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Assessing Cytochrome P450 Drug Interaction Risk for Dordaviprone Using Physiologically Based Pharmacokinetic Modeling 利用基于生理的药代动力学模型评估Dordaviprone患者的细胞色素P450药物相互作用风险。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-04 DOI: 10.1002/psp4.70093
Swati Jaiswal, Nikunjkumar K. Patel, Hannah M. Jones, Savannah McFeely, Shamia L. Faison, Tim Tippin, Odin Naderer

A physiologically based pharmacokinetic (PBPK) model was developed and verified for dordaviprone, a small molecule with antitumor effects in glioma patients. The model was applied to assess the drug–drug interaction (DDI) potential of dordaviprone as a victim of CYP3A4 inhibitors and inducers, and as a perpetrator of CYP3A4, CYP2C8, CYP2D6 inhibition. A combination of in vitro and clinical data was used to develop a minimal distribution PBPK model with a single adjusting compartment and mechanistic absorption using the Simcyp Population-Based Simulator (V21). Simulated maximum concentration (Cmax) and area under the concentration time curve (AUC) of the 3 clinical studies used to verify the PBPK model were within 1.4-fold of observed exposures. The simulated increase in dordaviprone AUC and Cmax (4.6- and 1.7-fold) following administration of multiple doses of itraconazole was consistent with the observed values (4.4- and 1.9-fold). All PBPK-simulated changes in dordaviprone plasma exposure when administered with CYP3A4 moderate (erythromycin, fluconazole) and weak (cimetidine) inhibitors, and moderate (efavirenz) and strong (rifampicin) inducers were consistent with their CYP3A4 potency classification (AUC ratio = 2.68, 2.48, 1.42, 0.35, and 0.17, respectively). The simulated AUC and Cmax of probe substrates for CYP3A4 (midazolam), CYP2C8 (repaglinide) and CYP2D6 (desipramine) after coadministration with 625 mg dordaviprone were the same as those in the absence of dordaviprone (ratio = 1.0) and remained unchanged after a sensitivity analysis using 10-fold more potent inhibition constants. Due to changes in dordaviprone plasma exposure when co-administered with CYP3A4 inhibitors, dordaviprone dose adjustments may be necessary; CYP3A4 inducers should be avoided.

建立并验证了脑胶质瘤患者中具有抗肿瘤作用的小分子dordaviprone的生理药代动力学(PBPK)模型。该模型用于评估dordaviprone作为CYP3A4抑制剂和诱诱剂的受害者,以及作为CYP3A4、CYP2C8、CYP2D6抑制的犯罪者的药物-药物相互作用(DDI)潜力。结合体外和临床数据,使用Simcyp基于人群的模拟器(V21)建立最小分布PBPK模型,该模型具有单个调节室和机械吸收。用于验证PBPK模型的3项临床研究的模拟最大浓度(Cmax)和浓度时间曲线下面积(AUC)均在观察暴露量的1.4倍以内。在给予多剂量伊曲康唑后,模拟的dordav易感AUC和Cmax的增加(4.6倍和1.7倍)与观察值(4.4倍和1.9倍)一致。当给予CYP3A4中度(红霉素、氟康唑)和弱(西咪替丁)抑制剂,以及中度(依非韦伦)和强(利福平)诱变剂时,所有pbpk模拟的多达韦易感血浆暴露的变化与其CYP3A4效价分类一致(AUC比分别为2.68、2.48、1.42、0.35和0.17)。与625 mg dordaviprone共给药后,CYP3A4(咪达唑仑)、CYP2C8(瑞格列奈)和CYP2D6(地西帕明)的模拟AUC和Cmax与不含dordaviprone(比值= 1.0)时的AUC和Cmax相同,在使用10倍有效抑制常数进行敏感性分析后保持不变。由于与CYP3A4抑制剂联合给药时多达韦易感性血浆暴露的变化,多达韦易感性剂量调整可能是必要的;应避免使用CYP3A4诱导剂。
{"title":"Assessing Cytochrome P450 Drug Interaction Risk for Dordaviprone Using Physiologically Based Pharmacokinetic Modeling","authors":"Swati Jaiswal,&nbsp;Nikunjkumar K. Patel,&nbsp;Hannah M. Jones,&nbsp;Savannah McFeely,&nbsp;Shamia L. Faison,&nbsp;Tim Tippin,&nbsp;Odin Naderer","doi":"10.1002/psp4.70093","DOIUrl":"10.1002/psp4.70093","url":null,"abstract":"<p>A physiologically based pharmacokinetic (PBPK) model was developed and verified for dordaviprone, a small molecule with antitumor effects in glioma patients. The model was applied to assess the drug–drug interaction (DDI) potential of dordaviprone as a victim of CYP3A4 inhibitors and inducers, and as a perpetrator of CYP3A4, CYP2C8, CYP2D6 inhibition. A combination of in vitro and clinical data was used to develop a minimal distribution PBPK model with a single adjusting compartment and mechanistic absorption using the Simcyp Population-Based Simulator (V21). Simulated maximum concentration (<i>C</i>max) and area under the concentration time curve (AUC) of the 3 clinical studies used to verify the PBPK model were within 1.4-fold of observed exposures. The simulated increase in dordaviprone AUC and Cmax (4.6- and 1.7-fold) following administration of multiple doses of itraconazole was consistent with the observed values (4.4- and 1.9-fold). All PBPK-simulated changes in dordaviprone plasma exposure when administered with CYP3A4 moderate (erythromycin, fluconazole) and weak (cimetidine) inhibitors, and moderate (efavirenz) and strong (rifampicin) inducers were consistent with their CYP3A4 potency classification (AUC ratio = 2.68, 2.48, 1.42, 0.35, and 0.17, respectively). The simulated AUC and Cmax of probe substrates for CYP3A4 (midazolam), CYP2C8 (repaglinide) and CYP2D6 (desipramine) after coadministration with 625 mg dordaviprone were the same as those in the absence of dordaviprone (ratio = 1.0) and remained unchanged after a sensitivity analysis using 10-fold more potent inhibition constants. Due to changes in dordaviprone plasma exposure when co-administered with CYP3A4 inhibitors, dordaviprone dose adjustments may be necessary; CYP3A4 inducers should be avoided.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1695-1704"},"PeriodicalIF":3.0,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70093","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144774861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pharmacometric Model-Based Sample Size Allocation for a Region of Interest in a Multi-Regional Phase 2 Trial: A Case Study of an Anti-Psoriatic Drug 基于药物计量学模型的多区域2期试验中感兴趣区域的样本量分配:一种抗银屑病药物的案例研究
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-01 DOI: 10.1002/psp4.70090
Xiao Zhang, Yubo Xiao, Junyi Wu, Scott Marshall, Xuan Zhou

Phase 2 trials have historically focused on characterizing the dose-exposure-response relationship in relatively homogeneous patient populations before proceeding to confirmatory trials. However, with the rise of multi-regional Phase 2 trials, it is important to strike a balance between this goal and the requirement to make sure that the optimal doses are chosen for patients from various geographic areas. This study uses a dose-ranging trial for an anti-psoriatic drug, featuring a typical design with a total sample size of N = 175, to highlight key considerations regarding sample size in multi-regional exploratory studies. The allocation of sample size to a region of interest (Region X) was evaluated using both a conventional statistical approach and a pharmacometric model-based (PMx) approach, predicated on the assumption of a minimum treatment improvement in Region X. Further evaluation was performed to assess the probability of reaching reliable conclusions regarding clinically relevant inter-regional differences in treatment response. The statistical approach, relying solely on end-of-trial observations from a single dose group, exhibited a maximum power of less than 40% in detecting treatment differences across regions when Region X accounts for 50% of the total sample size. In contrast, the PMx approach, employing data from multiple dose groups across trial duration, demonstrated that 26% of the total sample size yielded over 80% power to identify the inter-regional difference. The PMx approach has also been shown to offer a more efficient characterization of the clinical relevance of inter-regional differences, and has potential to improve decision-making in development progression by integrating prior knowledge.

在进行确证性试验之前,2期试验历来侧重于在相对均匀的患者群体中描述剂量-暴露-反应关系。然而,随着多地区二期试验的兴起,在这一目标和确保为不同地理区域的患者选择最佳剂量的要求之间取得平衡是很重要的。本研究采用一种抗银屑病药物的剂量范围试验,采用典型设计,总样本量为N = 175,以突出多区域探索性研究中样本量的关键考虑因素。使用传统统计方法和基于药物计量模型(PMx)的方法对感兴趣的区域(X区域)的样本量分配进行评估,该方法假设X区域的治疗改善最小,并进行进一步评估,以评估就临床相关的区域间治疗反应差异得出可靠结论的可能性。当X地区占总样本量的50%时,统计方法仅依赖于单一剂量组的试验结束观察结果,在检测不同地区治疗差异方面显示出不到40%的最大功率。相比之下,PMx方法在整个试验期间采用来自多个剂量组的数据,表明总样本量的26%产生了超过80%的能力来识别区域间差异。PMx方法也被证明可以更有效地描述区域间差异的临床相关性,并有可能通过整合先验知识来改善发展进程中的决策。
{"title":"Pharmacometric Model-Based Sample Size Allocation for a Region of Interest in a Multi-Regional Phase 2 Trial: A Case Study of an Anti-Psoriatic Drug","authors":"Xiao Zhang,&nbsp;Yubo Xiao,&nbsp;Junyi Wu,&nbsp;Scott Marshall,&nbsp;Xuan Zhou","doi":"10.1002/psp4.70090","DOIUrl":"10.1002/psp4.70090","url":null,"abstract":"<p>Phase 2 trials have historically focused on characterizing the dose-exposure-response relationship in relatively homogeneous patient populations before proceeding to confirmatory trials. However, with the rise of multi-regional Phase 2 trials, it is important to strike a balance between this goal and the requirement to make sure that the optimal doses are chosen for patients from various geographic areas. This study uses a dose-ranging trial for an anti-psoriatic drug, featuring a typical design with a total sample size of <i>N</i> = 175, to highlight key considerations regarding sample size in multi-regional exploratory studies. The allocation of sample size to a region of interest (Region X) was evaluated using both a conventional statistical approach and a pharmacometric model-based (PMx) approach, predicated on the assumption of a minimum treatment improvement in Region X. Further evaluation was performed to assess the probability of reaching reliable conclusions regarding clinically relevant inter-regional differences in treatment response. The statistical approach, relying solely on end-of-trial observations from a single dose group, exhibited a maximum power of less than 40% in detecting treatment differences across regions when Region X accounts for 50% of the total sample size. In contrast, the PMx approach, employing data from multiple dose groups across trial duration, demonstrated that 26% of the total sample size yielded over 80% power to identify the inter-regional difference. The PMx approach has also been shown to offer a more efficient characterization of the clinical relevance of inter-regional differences, and has potential to improve decision-making in development progression by integrating prior knowledge.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1673-1681"},"PeriodicalIF":3.0,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70090","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144764744","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Moving From Point-Based Analysis to Systems-Based Modeling: Knowledge Integration to Address Antimicrobial Resistance 从基于点的分析到基于系统的建模:解决抗菌素耐药性的知识整合。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-08-01 DOI: 10.1002/psp4.70092
Bhavatharini Arun, Gauri G. Rao

Optimizing antibiotic therapy requires a holistic bench-to-bedside approach with interdisciplinary collaboration between pharmacologists, clinicians, microbiologists, and computational scientists. Novel experimental models provide insights into drug-pathogen interactions within complex host environments, while multiomics data provide details of the molecular mechanisms shaping bacterial responses. Pharmacometrics and machine learning can be used to integrate these insights into in silico models. This perspective highlights how these approaches—when used effectively and often together to build a systems-level view—can inform drug development and improve clinical decision-making, ensuring the right drug is given to each patient at the right time, at the right dose, and for the right duration.

优化抗生素治疗需要药理学家、临床医生、微生物学家和计算科学家之间的跨学科合作,从实验室到床边的整体方法。新的实验模型提供了复杂宿主环境中药物-病原体相互作用的见解,而多组学数据提供了形成细菌反应的分子机制的细节。药物计量学和机器学习可用于将这些见解整合到计算机模型中。这一观点强调了这些方法——当有效使用并经常一起构建系统级视图时——如何为药物开发提供信息并改善临床决策,确保在正确的时间、正确的剂量和正确的持续时间给每个患者正确的药物。
{"title":"Moving From Point-Based Analysis to Systems-Based Modeling: Knowledge Integration to Address Antimicrobial Resistance","authors":"Bhavatharini Arun,&nbsp;Gauri G. Rao","doi":"10.1002/psp4.70092","DOIUrl":"10.1002/psp4.70092","url":null,"abstract":"<p>Optimizing antibiotic therapy requires a holistic bench-to-bedside approach with interdisciplinary collaboration between pharmacologists, clinicians, microbiologists, and computational scientists. Novel experimental models provide insights into drug-pathogen interactions within complex host environments, while multiomics data provide details of the molecular mechanisms shaping bacterial responses. Pharmacometrics and machine learning can be used to integrate these insights into in silico models. This perspective highlights how these approaches—when used effectively and often together to build a systems-level view—can inform drug development and improve clinical decision-making, ensuring the right drug is given to each patient at the right time, at the right dose, and for the right duration.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1535-1539"},"PeriodicalIF":3.0,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70092","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144764743","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Basel Modeling and Simulation Seminar: 20 Editions of Fostering Local Exchange in Pharmacometrics 巴塞尔建模和模拟研讨会:促进药物计量学本地交流的20个版本。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-07-31 DOI: 10.1002/psp4.70091
Tamara van Donge, João A. Abrantes, Kai Grosch, Gilbert Koch, Meemansa Sood, Britta Steffens, Andreas Krause

This year marks the 20th edition of the Basel Modeling and Simulation (M&S) Seminar, an initiative rooted in a commitment to promoting the exchange of the latest advancements in pharmacometrics and related disciplines in the region of Basel, Switzerland. This article provides insight into the history of this event, its operations to the present date, and a glimpse at the future.

今年是第20届巴塞尔建模与仿真(M&S)研讨会,这是一项旨在促进瑞士巴塞尔地区药物计量学和相关学科最新进展交流的倡议。本文将深入介绍这一事件的历史、迄今为止的运作情况,以及对未来的展望。
{"title":"The Basel Modeling and Simulation Seminar: 20 Editions of Fostering Local Exchange in Pharmacometrics","authors":"Tamara van Donge,&nbsp;João A. Abrantes,&nbsp;Kai Grosch,&nbsp;Gilbert Koch,&nbsp;Meemansa Sood,&nbsp;Britta Steffens,&nbsp;Andreas Krause","doi":"10.1002/psp4.70091","DOIUrl":"10.1002/psp4.70091","url":null,"abstract":"<p>This year marks the 20th edition of the Basel Modeling and Simulation (M&amp;S) Seminar, an initiative rooted in a commitment to promoting the exchange of the latest advancements in pharmacometrics and related disciplines in the region of Basel, Switzerland. This article provides insight into the history of this event, its operations to the present date, and a glimpse at the future.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1531-1534"},"PeriodicalIF":3.0,"publicationDate":"2025-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70091","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144752637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PBPK Modeling to Predict Clinical Drug–Drug Interaction and Impact of Hepatic Impairment for an ADC With the Payload Auristatin F-Hydroxypropylamide PBPK模型预测临床药物-药物相互作用和ADC与有效载荷Auristatin f -羟丙酰胺肝损害的影响。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-07-31 DOI: 10.1002/psp4.70088
Niyanta Kumar, Vaishali Dixit, Howard Burt, Katherine L. Gill, Hannah M. Jones, Natalie Keirstead, Dian Su, Dorin Toader, Timothy B. Lowinger

Upifitamab rilsodotin—an antibody drug conjugate (ADC)—comprises a NaPi2b-targeted antibody conjugated to an auristatin-based payload (auristatin F-hydroxypropylamide [AF-HPA]). AF-HPA is metabolized by cytochrome P450 3A4 (CYP3A4) and, to a lower extent, by CYP3A5 and demonstrates both reversible and time-dependent inhibition of CYP3A4. AF-HPA is also a P-glycoprotein (P-gp) substrate. A PBPK model was developed using a mixed “bottom-up” and “top-down” modeling approach with a combination of in vitro, nonclinical, and clinical ADME/PK data. The model recapitulated the clinical PK of conjugated and unconjugated AF-HPA. Simulations were used to predict the potential of unconjugated AF-HPA to be a victim or perpetrator of clinical drug–drug interactions (DDI) and predict the impact of hepatic impairment on the exposure to unconjugated AF-HPA. Simulations suggested negligible potential for clinical DDI between unconjugated AF-HPA and CYP3A substrates. Simulations also showed ~30% increase in unconjugated AF-HPA exposure following an IV dose of 36 mg/m2 in the presence of itraconazole, an inhibitor of both CYP3A4 and P-gp. A negligible change in the exposure to unconjugated AF-HPA was predicted in patients with mild hepatic impairment, which aligned with observed clinical data. The model predicted a ~1.5-fold increase in unconjugated AF-HPA AUC and negligible change in the Cmax in patients with moderate and severe hepatic impairment. Finally, this PBPK model may be applied (with modification to the conjugated drug sub-model parameters) to predict DDI and hepatic impairment potential for other ADCs with the same linker and payload.

Upifitamab rilsodotin是一种抗体药物偶联物(ADC),由一种靶向napi2b的抗体偶联到一种基于auristatin的有效载荷(auristatin F-hydroxypropylamide [AF-HPA])。AF-HPA被细胞色素P450 3A4 (CYP3A4)代谢,并在较低程度上被CYP3A5代谢,并表现出对CYP3A4的可逆和时间依赖性抑制。AF-HPA也是p -糖蛋白(P-gp)底物。采用混合的“自下而上”和“自上而下”建模方法,结合体外、非临床和临床ADME/PK数据,建立了PBPK模型。模型重现了偶联和未偶联AF-HPA的临床PK。模拟用于预测非偶联AF-HPA成为临床药物-药物相互作用(DDI)的受害者或犯罪者的潜力,并预测暴露于非偶联AF-HPA对肝脏损害的影响。模拟表明,未偶联的AF-HPA和CYP3A底物之间的临床DDI潜力可以忽略不计。模拟还显示,在伊曲康唑(一种CYP3A4和P-gp抑制剂)存在的情况下,静脉注射36mg /m2剂量后,非偶联AF-HPA暴露增加约30%。在轻度肝功能损害患者中,未偶联AF-HPA暴露的变化可忽略不计,这与观察到的临床数据一致。该模型预测,在中重度肝功能损害患者中,非共轭AF-HPA AUC增加约1.5倍,Cmax变化可忽略不计。最后,该PBPK模型可以应用(修改了共轭药物子模型参数)来预测具有相同连接物和有效载荷的其他adc的DDI和肝损害潜力。
{"title":"PBPK Modeling to Predict Clinical Drug–Drug Interaction and Impact of Hepatic Impairment for an ADC With the Payload Auristatin F-Hydroxypropylamide","authors":"Niyanta Kumar,&nbsp;Vaishali Dixit,&nbsp;Howard Burt,&nbsp;Katherine L. Gill,&nbsp;Hannah M. Jones,&nbsp;Natalie Keirstead,&nbsp;Dian Su,&nbsp;Dorin Toader,&nbsp;Timothy B. Lowinger","doi":"10.1002/psp4.70088","DOIUrl":"10.1002/psp4.70088","url":null,"abstract":"<p>Upifitamab rilsodotin—an antibody drug conjugate (ADC)—comprises a NaPi2b-targeted antibody conjugated to an auristatin-based payload (auristatin F-hydroxypropylamide [AF-HPA]). AF-HPA is metabolized by cytochrome P450 3A4 (CYP3A4) and, to a lower extent, by CYP3A5 and demonstrates both reversible and time-dependent inhibition of CYP3A4. AF-HPA is also a P-glycoprotein (P-gp) substrate. A PBPK model was developed using a mixed “bottom-up” and “top-down” modeling approach with a combination of in vitro, nonclinical, and clinical ADME/PK data. The model recapitulated the clinical PK of conjugated and unconjugated AF-HPA. Simulations were used to predict the potential of unconjugated AF-HPA to be a victim or perpetrator of clinical drug–drug interactions (DDI) and predict the impact of hepatic impairment on the exposure to unconjugated AF-HPA. Simulations suggested negligible potential for clinical DDI between unconjugated AF-HPA and CYP3A substrates. Simulations also showed ~30% increase in unconjugated AF-HPA exposure following an IV dose of 36 mg/m<sup>2</sup> in the presence of itraconazole, an inhibitor of both CYP3A4 and P-gp. A negligible change in the exposure to unconjugated AF-HPA was predicted in patients with mild hepatic impairment, which aligned with observed clinical data. The model predicted a ~1.5-fold increase in unconjugated AF-HPA AUC and negligible change in the <i>C</i><sub>max</sub> in patients with moderate and severe hepatic impairment. Finally, this PBPK model may be applied (with modification to the conjugated drug sub-model parameters) to predict DDI and hepatic impairment potential for other ADCs with the same linker and payload.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 10","pages":"1661-1672"},"PeriodicalIF":3.0,"publicationDate":"2025-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70088","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144759398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
CPT: Pharmacometrics & Systems Pharmacology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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