首页 > 最新文献

CPT: Pharmacometrics & Systems Pharmacology最新文献

英文 中文
A Step-by-Step Workflow for Performing In Silico Clinical Trials With Nonlinear Mixed Effects Models 一个循序渐进的工作流程执行在硅临床试验与非线性混合效应模型。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-11 DOI: 10.1002/psp4.70122
Javiera Cortés-Ríos, Mindy Magee, Anna Sher, William J. Jusko, Rajat Desikan

In silico clinical trials (ISCT) are computational frameworks that employ mathematical models to generate virtual patients and simulate their responses to new treatments, treatment regimens, or medical devices via simulations mirroring real-world clinical trials. ISCTs are an important component of the model-informed drug development (MIDD) framework for optimizing therapies, treatment personalization, informing regulatory decisions, and accelerating overall drug development by enhancing R&D productivity. However, the emergence of complex models, such as quantitative systems pharmacology (QSP) models, presents significant challenges for their effective implementation. Guidelines for conducting ISCTs have been published to address these challenges, focusing on algorithms and credibility frameworks for generating plausible virtual patients and calibrating virtual populations. However, it is not straightforward to apply existing workflows to models where parameter distributions and correlations are estimated using nonlinear mixed effects (NLME) population fitting approaches, a common practice in the pharmaceutical industry when individual-patient-level data is available. Here, we illustrate a modeling workflow for conducting ISCTs with NLME models, detailing key considerations, methods, and challenges at each step. We demonstrate the practical implementation of this workflow through two examples to showcase its broad applicability: (1) a simple model predicting tumor growth in response to chemotherapy and (2) a more complex mechanistic QSP model of hepatitis B virus infection that captures the physiological mechanisms underlying treatment response with standard-of-care therapies.

计算机临床试验(ISCT)是一种计算框架,它采用数学模型来生成虚拟患者,并通过模拟反映现实世界临床试验来模拟他们对新疗法、治疗方案或医疗设备的反应。isct是模型信息药物开发(MIDD)框架的重要组成部分,用于优化疗法、治疗个性化、为监管决策提供信息,并通过提高研发生产率来加速整体药物开发。然而,复杂模型的出现,如定量系统药理学(QSP)模型,为其有效实施提出了重大挑战。为了应对这些挑战,已经发布了开展isct的指南,重点关注生成可信虚拟患者和校准虚拟人群的算法和可信度框架。然而,将现有的工作流程应用于使用非线性混合效应(NLME)总体拟合方法估计参数分布和相关性的模型并不简单,这是制药行业在个体患者水平数据可用时的常见做法。在这里,我们说明了用NLME模型进行isct的建模工作流程,详细说明了每个步骤的关键考虑因素、方法和挑战。我们通过两个例子演示了该工作流程的实际实施,以展示其广泛的适用性:(1)预测肿瘤对化疗反应的简单模型;(2)更复杂的乙型肝炎病毒感染机制QSP模型,该模型捕获了标准治疗疗法治疗反应的生理机制。
{"title":"A Step-by-Step Workflow for Performing In Silico Clinical Trials With Nonlinear Mixed Effects Models","authors":"Javiera Cortés-Ríos,&nbsp;Mindy Magee,&nbsp;Anna Sher,&nbsp;William J. Jusko,&nbsp;Rajat Desikan","doi":"10.1002/psp4.70122","DOIUrl":"10.1002/psp4.70122","url":null,"abstract":"<p>In silico clinical trials (ISCT) are computational frameworks that employ mathematical models to generate virtual patients and simulate their responses to new treatments, treatment regimens, or medical devices via simulations mirroring real-world clinical trials. ISCTs are an important component of the model-informed drug development (MIDD) framework for optimizing therapies, treatment personalization, informing regulatory decisions, and accelerating overall drug development by enhancing R&amp;D productivity. However, the emergence of complex models, such as quantitative systems pharmacology (QSP) models, presents significant challenges for their effective implementation. Guidelines for conducting ISCTs have been published to address these challenges, focusing on algorithms and credibility frameworks for generating plausible virtual patients and calibrating virtual populations. However, it is not straightforward to apply existing workflows to models where parameter distributions and correlations are estimated using nonlinear mixed effects (NLME) population fitting approaches, a common practice in the pharmaceutical industry when individual-patient-level data is available. Here, we illustrate a modeling workflow for conducting ISCTs with NLME models, detailing key considerations, methods, and challenges at each step. We demonstrate the practical implementation of this workflow through two examples to showcase its broad applicability: (1) a simple model predicting tumor growth in response to chemotherapy and (2) a more complex mechanistic QSP model of hepatitis B virus infection that captures the physiological mechanisms underlying treatment response with standard-of-care therapies.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"1949-1964"},"PeriodicalIF":3.0,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70122","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274114","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
Optimizing Ibrutinib Posology in Chronic Lymphocytic Leukemia Using a Semi-Mechanistic Pharmacometric Framework 利用半机械药理学框架优化依鲁替尼治疗慢性淋巴细胞白血病的疗效。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-11 DOI: 10.1002/psp4.70124
Eman I. K. Ibrahim, Lena E. Friberg

Ibrutinib, a Bruton's tyrosine kinase (Btk) inhibitor, is a key therapy for chronic lymphocytic leukemia (CLL). In clinical practice, adverse events, such as hypertension, frequently necessitate dose reductions or treatment discontinuation. Emerging evidence suggests that reduced doses may retain clinical efficacy while mitigating toxicity. The synergistic ibrutinib–venetoclax combination remains understudied at low doses, particularly for ibrutinib. This study aimed to explore dose optimization strategies, with/without venetoclax, in treatment-naïve (TN) and relapsed/refractory (R/R) CLL using mechanism-based, model-informed approaches to characterize the relationship between systemic ibrutinib exposure and efficacy and safety biomarkers. We leveraged data from phase 1b/2 and 3 studies, including plasma concentrations, leukocyte and lymphocyte counts, lymph node and spleen size measurements, and blood pressure. A previously developed semi-mechanistic population pharmacokinetic-pharmacodynamic (PKPD) framework was re-evaluated, extended by integrating additional biomarkers and identifying differences between TN and R/R patients, and used to simulate alternative dosing strategies. The model successfully captured the temporal dynamics of all biomarkers simultaneously. We quantified a 76% longer phospho-Btk half-life and a 43% shorter peripheral CLL cell half-life in TN versus R/R patients, with no evidence of ibrutinib resistance in TN patients. Dose reductions based on response depth or toxicity preserved comparable response rates and progression-free survival to standard dosing. Ibrutinib de-escalation schedules with venetoclax resulted in a ≤ 5% reduction in peripheral blood measurable residual disease compared to standard dosing at 2 years. This PKPD framework supports dose individualization to improve tolerability without sacrificing treatment outcomes, offering a path toward more personalized, effective CLL management.

伊鲁替尼是一种布鲁顿酪氨酸激酶(Btk)抑制剂,是治疗慢性淋巴细胞白血病(CLL)的关键药物。在临床实践中,不良事件,如高血压,经常需要减少剂量或停止治疗。新出现的证据表明,减少剂量可以在减轻毒性的同时保持临床疗效。在低剂量情况下,对伊鲁替尼-维托克拉克斯联合增效的研究仍不充分,尤其是对伊鲁替尼。本研究旨在探索使用/不使用venetoclax治疗treatment-naïve (TN)和复发/难治性CLL (R/R)的剂量优化策略,使用基于机制的模型信息方法来表征全身伊鲁替尼暴露与疗效和安全性生物标志物之间的关系。我们利用了1b/2期和3期研究的数据,包括血浆浓度、白细胞和淋巴细胞计数、淋巴结和脾脏大小测量以及血压。先前开发的半机械人群药代动力学-药效学(PKPD)框架被重新评估,通过整合其他生物标志物和识别TN和R/R患者之间的差异进行扩展,并用于模拟替代给药策略。该模型成功地同时捕获了所有生物标志物的时间动态。我们量化了TN患者的磷酸化btk半衰期比R/R患者长76%,外周CLL细胞半衰期比R/R患者短43%,没有证据表明TN患者对伊鲁替尼耐药。基于反应深度或毒性的剂量减少保持了与标准剂量相当的反应率和无进展生存期。与标准剂量相比,2年时依鲁替尼与venetoclax的降级方案导致外周血可测量残留疾病减少≤5%。该PKPD框架支持剂量个性化,在不牺牲治疗结果的情况下提高耐受性,为更个性化、更有效的CLL管理提供了途径。
{"title":"Optimizing Ibrutinib Posology in Chronic Lymphocytic Leukemia Using a Semi-Mechanistic Pharmacometric Framework","authors":"Eman I. K. Ibrahim,&nbsp;Lena E. Friberg","doi":"10.1002/psp4.70124","DOIUrl":"10.1002/psp4.70124","url":null,"abstract":"<p>Ibrutinib, a Bruton's tyrosine kinase (Btk) inhibitor, is a key therapy for chronic lymphocytic leukemia (CLL). In clinical practice, adverse events, such as hypertension, frequently necessitate dose reductions or treatment discontinuation. Emerging evidence suggests that reduced doses may retain clinical efficacy while mitigating toxicity. The synergistic ibrutinib–venetoclax combination remains understudied at low doses, particularly for ibrutinib. This study aimed to explore dose optimization strategies, with/without venetoclax, in treatment-naïve (TN) and relapsed/refractory (R/R) CLL using mechanism-based, model-informed approaches to characterize the relationship between systemic ibrutinib exposure and efficacy and safety biomarkers. We leveraged data from phase 1b/2 and 3 studies, including plasma concentrations, leukocyte and lymphocyte counts, lymph node and spleen size measurements, and blood pressure. A previously developed semi-mechanistic population pharmacokinetic-pharmacodynamic (PKPD) framework was re-evaluated, extended by integrating additional biomarkers and identifying differences between TN and R/R patients, and used to simulate alternative dosing strategies. The model successfully captured the temporal dynamics of all biomarkers simultaneously. We quantified a 76% longer phospho-Btk half-life and a 43% shorter peripheral CLL cell half-life in TN versus R/R patients, with no evidence of ibrutinib resistance in TN patients. Dose reductions based on response depth or toxicity preserved comparable response rates and progression-free survival to standard dosing. Ibrutinib de-escalation schedules with venetoclax resulted in a ≤ 5% reduction in peripheral blood measurable residual disease compared to standard dosing at 2 years. This PKPD framework supports dose individualization to improve tolerability without sacrificing treatment outcomes, offering a path toward more personalized, effective CLL management.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2186-2197"},"PeriodicalIF":3.0,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70124","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274141","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 Unified Whole Lung PBK Model for Inhalational Uptake of Gases and Aerosols in Men 男性吸入气体和气溶胶吸收的统一全肺PBK模型。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-04 DOI: 10.1002/psp4.70117
Norman Nowak, Sylvia E. Escher, Katharina Schwarz

Assessing the risk or benefit of an inhaled substance is challenging due to the variety of forms it can take (gas, vapor, particle, or droplet) and the complex biological processes involved in its uptake and retention. Physiologically based kinetic (PBK) models offer an alternative to in vivo experiments. However, PBK models for inhalational uptake are to date either designed for gases/vapors or airborne particulates, often with only low regional compartmentalization. The here-presented, newly developed model combines both applications. Its mechanisms are an amalgamation of PBK and non-PBK models integrated into a multicompartmental description of the human lung to include the relevant uptake and clearance processes in the different lung regions, of which macrophage-mediated dissolution is novel to PBK modeling. The model was designed to use a minimal number of substance-specific input parameters, which can be derived from in vitro assays or in silico methods. Model predictions for hypothetical substances with varying physicochemical properties were performed, along with rudimentary sensitivity analyses to identify the most important parameters for gases/vapors and particles. This novel PBK model combines all these aspects and provides in silico predictions of systemic and local lung concentrations, reducing uncertainty in risk assessments and supporting drug development. It serves as a valuable tool to translate nominal ambient air doses into effective localized doses within the lung.

评估吸入物质的风险或益处是具有挑战性的,因为它可以以多种形式存在(气体、蒸气、颗粒或液滴),并且在其吸收和保留过程中涉及复杂的生物过程。基于生理的动力学(PBK)模型为体内实验提供了另一种选择。然而,迄今为止,用于吸入吸收量的PBK模型要么是为气体/蒸气设计的,要么是为空气中的颗粒物设计的,通常只有较低的区域划分。这里介绍的新开发的模型结合了这两个应用程序。其机制是PBK和非PBK模型的融合,整合到人肺的多室描述中,包括不同肺区域的相关摄取和清除过程,其中巨噬细胞介导的溶解是PBK模型的新内容。该模型被设计为使用最小数量的物质特异性输入参数,这些参数可以从体外测定或计算机方法中获得。对具有不同物理化学性质的假设物质进行了模型预测,并进行了基本的灵敏度分析,以确定气体/蒸气和颗粒的最重要参数。这种新颖的PBK模型结合了所有这些方面,并提供了全身和局部肺浓度的计算机预测,减少了风险评估的不确定性,并支持药物开发。它是一种有价值的工具,可将名义环境空气剂量转化为肺内有效的局部剂量。
{"title":"A Unified Whole Lung PBK Model for Inhalational Uptake of Gases and Aerosols in Men","authors":"Norman Nowak,&nbsp;Sylvia E. Escher,&nbsp;Katharina Schwarz","doi":"10.1002/psp4.70117","DOIUrl":"10.1002/psp4.70117","url":null,"abstract":"<p>Assessing the risk or benefit of an inhaled substance is challenging due to the variety of forms it can take (gas, vapor, particle, or droplet) and the complex biological processes involved in its uptake and retention. Physiologically based kinetic (PBK) models offer an alternative to in vivo experiments. However, PBK models for inhalational uptake are to date either designed for gases/vapors or airborne particulates, often with only low regional compartmentalization. The here-presented, newly developed model combines both applications. Its mechanisms are an amalgamation of PBK and non-PBK models integrated into a multicompartmental description of the human lung to include the relevant uptake and clearance processes in the different lung regions, of which macrophage-mediated dissolution is novel to PBK modeling. The model was designed to use a minimal number of substance-specific input parameters, which can be derived from in vitro assays or in silico methods. Model predictions for hypothetical substances with varying physicochemical properties were performed, along with rudimentary sensitivity analyses to identify the most important parameters for gases/vapors and particles. This novel PBK model combines all these aspects and provides in silico predictions of systemic and local lung concentrations, reducing uncertainty in risk assessments and supporting drug development. It serves as a valuable tool to translate nominal ambient air doses into effective localized doses within the lung.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2173-2185"},"PeriodicalIF":3.0,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70117","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145225070","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
Population Pharmacokinetic and Pharmacodynamic Modeling for the Prediction of the Extended Amlitelimab Phase 3 Dosing Regimen in Atopic Dermatitis 延长Amlitelimab 3期给药方案治疗特应性皮炎的人群药代动力学和药效学模型预测。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-01 DOI: 10.1002/psp4.70121
Gilles Tiraboschi, Kim Papp, Thomas Bieber, Stephan Weidinger, Lisa Beck, Chih-Hung Lee, John T. O'Malley, Karl Yen, Charlotte Bernigaud, David Fabre, Fabrice Hurbin

Amlitelimab is a fully human, nondepleting, anti-OX40 ligand monoclonal antibody being investigated for the treatment of moderate-to-severe atopic dermatitis (AD) in adults and adolescents. Population pharmacokinetic (PopPK) and pharmacokinetic/pharmacodynamic-Eczema Area and Severity Index (PopPK/PD-EASI) models were used to inform dosing regimen selection for amlitelimab phase 3 trials. The PopPK model was developed using phase 1 (healthy volunteers) and phase 2 (participants with AD) trial data, including individual exposure variables from the STREAM-AD phase 2b trial following subcutaneous amlitelimab doses ranging from 62.5 to 250 mg given every 4 weeks (Q4W). The PopPK model was used to compute exposures for an extended dosing regimen of 250 mg Q12W (with 500 mg loading dose [+LD]). The PopPK/PD-EASI model was developed from phase 2 trials to predict treatment responses (EASI values) with selected dosing scenarios. Finally, the dose for individuals with lower body weight (i.e., < 40 kg) was determined. Utilizing the PopPK model, the amlitelimab 250 mg Q12W + LD computed exposures were between the exposures of 62.5 mg Q4W and 250 mg Q4W + LD efficacious doses in the STREAM-AD trial. Using the PopPK/PD-EASI model, the simulated efficacy for dosing scenarios of 250 mg Q12W + LD regimen from initiation or 250 mg Q4W + LD for 24 weeks followed by Q12W to Week 60 was similar to continuous 250 mg Q4W. Simulations identified that a twofold dose reduction would allow participants < 40 kg to achieve amlitelimab exposures within the range observed in participants ≥ 40 kg on 250 mg Q4W or Q12W. These results support evaluation of a Q12W dosing regimen for adults and adolescents in phase 3 trials.

Amlitelimab是一种全人源、非消耗性、抗ox40配体单克隆抗体,正在研究用于治疗成人和青少年中重度特应性皮炎(AD)。人群药代动力学(PopPK)和药代动力学/药效学-湿疹面积和严重程度指数(PopPK/PD-EASI)模型用于amlitelimab 3期试验的给药方案选择。PopPK模型是使用1期(健康志愿者)和2期(AD患者)试验数据开发的,包括来自流式-AD 2b期试验的个体暴露变量,每4周(Q4W)皮下给药amlitelimab剂量范围为62.5至250mg。PopPK模型用于计算250 mg Q12W延长给药方案(500 mg负荷剂量[+LD])的暴露量。PopPK/PD-EASI模型是根据2期试验开发的,用于预测选定给药方案下的治疗反应(EASI值)。最后,对于体重较轻的个体(即
{"title":"Population Pharmacokinetic and Pharmacodynamic Modeling for the Prediction of the Extended Amlitelimab Phase 3 Dosing Regimen in Atopic Dermatitis","authors":"Gilles Tiraboschi,&nbsp;Kim Papp,&nbsp;Thomas Bieber,&nbsp;Stephan Weidinger,&nbsp;Lisa Beck,&nbsp;Chih-Hung Lee,&nbsp;John T. O'Malley,&nbsp;Karl Yen,&nbsp;Charlotte Bernigaud,&nbsp;David Fabre,&nbsp;Fabrice Hurbin","doi":"10.1002/psp4.70121","DOIUrl":"10.1002/psp4.70121","url":null,"abstract":"<p>Amlitelimab is a fully human, nondepleting, anti-OX40 ligand monoclonal antibody being investigated for the treatment of moderate-to-severe atopic dermatitis (AD) in adults and adolescents. Population pharmacokinetic (PopPK) and pharmacokinetic/pharmacodynamic-Eczema Area and Severity Index (PopPK/PD-EASI) models were used to inform dosing regimen selection for amlitelimab phase 3 trials. The PopPK model was developed using phase 1 (healthy volunteers) and phase 2 (participants with AD) trial data, including individual exposure variables from the STREAM-AD phase 2b trial following subcutaneous amlitelimab doses ranging from 62.5 to 250 mg given every 4 weeks (Q4W). The PopPK model was used to compute exposures for an extended dosing regimen of 250 mg Q12W (with 500 mg loading dose [+LD]). The PopPK/PD-EASI model was developed from phase 2 trials to predict treatment responses (EASI values) with selected dosing scenarios. Finally, the dose for individuals with lower body weight (i.e., &lt; 40 kg) was determined. Utilizing the PopPK model, the amlitelimab 250 mg Q12W + LD computed exposures were between the exposures of 62.5 mg Q4W and 250 mg Q4W + LD efficacious doses in the STREAM-AD trial. Using the PopPK/PD-EASI model, the simulated efficacy for dosing scenarios of 250 mg Q12W + LD regimen from initiation or 250 mg Q4W + LD for 24 weeks followed by Q12W to Week 60 was similar to continuous 250 mg Q4W. Simulations identified that a twofold dose reduction would allow participants &lt; 40 kg to achieve amlitelimab exposures within the range observed in participants ≥ 40 kg on 250 mg Q4W or Q12W. These results support evaluation of a Q12W dosing regimen for adults and adolescents in phase 3 trials.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2161-2172"},"PeriodicalIF":3.0,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70121","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145205708","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
Population Pharmacokinetic Analysis of Datopotamab Deruxtecan (Dato-DXd), a TROP2-Directed Antibody-Drug Conjugate, in Patients With Advanced Solid Tumors 一种trop2导向抗体-药物偶联物Dato-DXd在晚期实体肿瘤患者中的群体药代动力学分析
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-01 DOI: 10.1002/psp4.70118
Ying Hong, Sophie Peigné, Υuzhuo Pan, Sofia Friberg Hietala, Anna McLaughlin, Naoyuki Tajima, Deise Uema, Hong Zebger-Gong, Zoey Tang, Diansong Zhou, Malaz Abutarif, Τushar Garimella

Datopotamab deruxtecan (Dato-DXd) is a trophoblast cell surface antigen 2 (TROP2)-directed antibody-drug conjugate (ADC) developed for the treatment of advanced or metastatic solid tumors. Dato-DXd demonstrated promising antitumor activity and a manageable safety profile in the first-in-human Phase 1 study TROPION-PanTumor01. The present work established population pharmacokinetic models for Dato-DXd and DXd in patients with advanced solid tumors, using data from three clinical studies. The final Dato-DXd model was a two-compartment model with both linear (CLlinDatoDXd) and nonlinear elimination. CLlinDatoDXd was the major elimination pathway for Dato-DXd doses ≥ 4 mg/kg, whereas nonlinear clearance was necessary to capture the concentration-dependent clearance at lower doses. Body weight was added as a mechanistic covariate with a fixed allometric exponent of 0.75 (estimated value: 0.80) on clearance and estimated exponents on volumes of distribution. The final model for DXd was a one-compartment model with linear clearance (CLDXd). The release of DXd was equal to the total elimination rate of Dato-DXd and was found to be time-dependent within and between cycles, as previously observed for other ADCs. For a typical 66 kg male patient, CLlinDatoDXd was 0.386 L/day, central volume of distribution VcDatoDXd was 3.06 L, CLDXd was 2.66 L/day, and VcDXd was 25.1 L. Covariate analysis identified body weight as the most influential covariate on Dato-DXd and DXd exposure. These findings support the weight-based dosing strategy and indicate no dose adjustments are warranted based on the covariates evaluated.

Datopotamab deruxtecan (Dato-DXd)是一种滋养细胞表面抗原2 (TROP2)导向的抗体-药物偶联物(ADC),用于治疗晚期或转移性实体瘤。Dato-DXd在首次人体i期研究TROPION-PanTumor01中显示出有希望的抗肿瘤活性和可管理的安全性。本工作利用三个临床研究的数据,建立了Dato-DXd和DXd在晚期实体瘤患者中的群体药代动力学模型。最终的Dato-DXd模型是一个具有线性(CLlinDatoDXd)和非线性消除的两室模型。CLlinDatoDXd是Dato-DXd剂量≥4 mg/kg时的主要消除途径,而在较低剂量下,非线性清除是捕获浓度依赖性清除所必需的。体重作为一个机制协变量加入,在间隙上的异速生长指数为0.75(估计值为0.80),在分布体积上的估计指数为0.75。DXd的最终模型是线性间隙的单室模型(CLDXd)。DXd的释放量等于Dato-DXd的总消除率,并且发现在周期内和周期之间具有时间依赖性,与先前观察到的其他adc相同。典型66 kg男性患者CLlinDatoDXd为0.386 L/day,中心分布容积VcDatoDXd为3.06 L, CLDXd为2.66 L/day, VcDXd为25.1 L。协变量分析发现,体重是影响达多-DXd和DXd暴露的最大协变量。这些发现支持基于体重的给药策略,并表明不需要根据评估的协变量进行剂量调整。
{"title":"Population Pharmacokinetic Analysis of Datopotamab Deruxtecan (Dato-DXd), a TROP2-Directed Antibody-Drug Conjugate, in Patients With Advanced Solid Tumors","authors":"Ying Hong,&nbsp;Sophie Peigné,&nbsp;Υuzhuo Pan,&nbsp;Sofia Friberg Hietala,&nbsp;Anna McLaughlin,&nbsp;Naoyuki Tajima,&nbsp;Deise Uema,&nbsp;Hong Zebger-Gong,&nbsp;Zoey Tang,&nbsp;Diansong Zhou,&nbsp;Malaz Abutarif,&nbsp;Τushar Garimella","doi":"10.1002/psp4.70118","DOIUrl":"10.1002/psp4.70118","url":null,"abstract":"<p>Datopotamab deruxtecan (Dato-DXd) is a trophoblast cell surface antigen 2 (TROP2)-directed antibody-drug conjugate (ADC) developed for the treatment of advanced or metastatic solid tumors. Dato-DXd demonstrated promising antitumor activity and a manageable safety profile in the first-in-human Phase 1 study TROPION-PanTumor01. The present work established population pharmacokinetic models for Dato-DXd and DXd in patients with advanced solid tumors, using data from three clinical studies. The final Dato-DXd model was a two-compartment model with both linear (CL<sub>linDatoDXd</sub>) and nonlinear elimination. CL<sub>linDatoDXd</sub> was the major elimination pathway for Dato-DXd doses ≥ 4 mg/kg, whereas nonlinear clearance was necessary to capture the concentration-dependent clearance at lower doses. Body weight was added as a mechanistic covariate with a fixed allometric exponent of 0.75 (estimated value: 0.80) on clearance and estimated exponents on volumes of distribution. The final model for DXd was a one-compartment model with linear clearance (CL<sub>DXd</sub>). The release of DXd was equal to the total elimination rate of Dato-DXd and was found to be time-dependent within and between cycles, as previously observed for other ADCs. For a typical 66 kg male patient, CL<sub>linDatoDXd</sub> was 0.386 L/day, central volume of distribution <i>V</i><sub>cDatoDXd</sub> was 3.06 L, CL<sub>DXd</sub> was 2.66 L/day, and <i>V</i><sub>cDXd</sub> was 25.1 L. Covariate analysis identified body weight as the most influential covariate on Dato-DXd and DXd exposure. These findings support the weight-based dosing strategy and indicate no dose adjustments are warranted based on the covariates evaluated.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2149-2160"},"PeriodicalIF":3.0,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70118","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145205678","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
Using Virtual Patients to Evaluate Dosing Strategies for Bispecifics With a Bell-Shaped Efficacy Curve 使用虚拟患者评估双种药的钟形疗效曲线的给药策略。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-09-30 DOI: 10.1002/psp4.70105
Jana L. Gevertz, Irina Kareva

Bispecific antibodies can be broadly divided into two categories: those that are pharmacologically active as either dimers bound to a single target or as trimers bound to both targets, and those that are only active as trimers. Dose selection of trimer-based bispecifics poses a unique challenge, as toxicity increases with dose, but efficacy does not. Instead, trimer-driven bispecifics have a bell-shaped efficacy curve, for which both under- and over-dosing can cause a decrease in efficacy. To address the challenge of dose selection for trimer-based bispecifics, we develop a semi-mechanistic pharmacokinetic/pharmacodynamic model of one such bispecific, teclistamab. By introducing variability in key patient-specific parameters, we find that the currently selected phase II recommended dose of 1.5 mg/kg administered subcutaneously weekly falls within the calculated optimal range for maximizing concentration of the pharmacologically active trimer for a broad population. We next explore different strategies for patient stratification based on pre-treatment levels of measurable biomarkers. We discover that significantly more variability across subpopulations is predicted when the drug is administered every 2 weeks as compared to weekly administration, and that higher doses generally result in more interpatient variability. Further, the pharmacologically active trimers are predicted to be maximized at different doses for different subpopulations. These findings underscore the potential for model-supported patient stratification based on measurable biomarkers, offering a middle ground between population-level approaches and fully personalized medicine.

双特异性抗体大致可分为两类:一类是作为二聚体结合单一靶标或作为三聚体结合两个靶标具有药理活性的抗体,另一类是仅作为三聚体具有活性的抗体。基于三聚体的双特异性药物的剂量选择提出了一个独特的挑战,因为毒性随剂量增加而增加,但效力不会增加。相反,三聚体驱动的双特异性药物具有钟形疗效曲线,剂量不足和过量都会导致疗效下降。为了解决基于三聚体的双特异性药物剂量选择的挑战,我们开发了一种半机械药代动力学/药效学模型,用于一种这样的双特异性,teclistamab。通过引入关键患者特异性参数的可变性,我们发现目前选择的II期推荐剂量为每周1.5 mg/kg皮下给药,在计算的最佳范围内,可以使广泛人群的药理活性三聚体浓度最大化。接下来,我们将根据治疗前可测量的生物标志物水平探索不同的患者分层策略。我们发现,与每周给药相比,每两周给药一次可以预测亚人群之间的差异更大,而且更高的剂量通常会导致更大的患者间差异。此外,预测在不同的亚群中,不同剂量的药理学活性三聚体达到最大。这些发现强调了基于可测量的生物标志物的模型支持的患者分层的潜力,在人口水平的方法和完全个性化的医疗之间提供了一个中间地带。
{"title":"Using Virtual Patients to Evaluate Dosing Strategies for Bispecifics With a Bell-Shaped Efficacy Curve","authors":"Jana L. Gevertz,&nbsp;Irina Kareva","doi":"10.1002/psp4.70105","DOIUrl":"10.1002/psp4.70105","url":null,"abstract":"<p>Bispecific antibodies can be broadly divided into two categories: those that are pharmacologically active as either dimers bound to a single target or as trimers bound to both targets, and those that are only active as trimers. Dose selection of trimer-based bispecifics poses a unique challenge, as toxicity increases with dose, but efficacy does not. Instead, trimer-driven bispecifics have a bell-shaped efficacy curve, for which both under- and over-dosing can cause a decrease in efficacy. To address the challenge of dose selection for trimer-based bispecifics, we develop a semi-mechanistic pharmacokinetic/pharmacodynamic model of one such bispecific, teclistamab. By introducing variability in key patient-specific parameters, we find that the currently selected phase II recommended dose of 1.5 mg/kg administered subcutaneously weekly falls within the calculated optimal range for maximizing concentration of the pharmacologically active trimer for a broad population. We next explore different strategies for patient stratification based on pre-treatment levels of measurable biomarkers. We discover that significantly more variability across subpopulations is predicted when the drug is administered every 2 weeks as compared to weekly administration, and that higher doses generally result in more interpatient variability. Further, the pharmacologically active trimers are predicted to be maximized at different doses for different subpopulations. These findings underscore the potential for model-supported patient stratification based on measurable biomarkers, offering a middle ground between population-level approaches and fully personalized medicine.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2139-2148"},"PeriodicalIF":3.0,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70105","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145191391","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 Model-Based Meta-Analysis of Pembrolizumab Effects on Patient-Reported Quality of Life: Advancing Patient-Centered Oncology Drug Development. Pembrolizumab对患者报告的生活质量影响的基于模型的meta分析:推进以患者为中心的肿瘤药物开发
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-09-30 DOI: 10.1002/psp4.70106
Yiqin Zou, Yimeng Sun, Sudhamshu Ravva, Lynne I Wagner, Jiawei Zhou

Pembrolizumab is an immune checkpoint inhibitor that has been approved for more than 20 different indications and has shown great survival benefits in various types of cancer. However, the reported benefits of pembrolizumab in patients' quality of life (QoL) have been inconsistent across different studies and different types of cancer. As oncology drug development increasingly emphasizes patient-centered care, patient-reported outcomes (PROs), particularly patient-reported QoL, are recognized as important clinical endpoints. To characterize the effects of pembrolizumab on patient-reported QoL, we conducted a model-based meta-analysis (MBMA) of published clinical trials evaluating pembrolizumab across different types of cancer. The longitudinal EORTC QLQ-C30 GHS/QOL data were extracted in our analysis as QoL scores. A population model was developed to characterize the longitudinal QoL trajectories and quantify both treatment toxicity and efficacy. Out of more than 300 screened studies, only 20 reported longitudinal EORTC QLQ-C30 QoL data. Among these, 8 studies reported no between-group differences in QoL outcomes between pembrolizumab and control arms. However, our modeling revealed that pembrolizumab was associated with greater toxicity but improved long-term QoL. Notably, our approach identified treatment effects on QoL that were not detected by traditional statistical analyses in the original publications. In summary, our study demonstrates that MBMA combined with population modeling enables more accurate evaluation of longitudinal PROs data, overcoming the limitations of conventional methods. This approach offers a robust framework for integrating patient-centered outcomes into oncology drug development and supports the broader use of PROs data in regulatory and clinical decision-making.

Pembrolizumab是一种免疫检查点抑制剂,已被批准用于20多种不同的适应症,并在各种类型的癌症中显示出巨大的生存益处。然而,在不同的研究和不同类型的癌症中,报告的派姆单抗对患者生活质量(QoL)的益处并不一致。随着肿瘤药物开发越来越强调以患者为中心的护理,患者报告的预后(pro),特别是患者报告的生活质量(QoL),被认为是重要的临床终点。为了描述派姆单抗对患者报告的生活质量的影响,我们对已发表的临床试验进行了基于模型的荟萃分析(MBMA),评估派姆单抗对不同类型癌症的影响。在我们的分析中提取纵向EORTC QLQ-C30 GHS/QOL数据作为QOL评分。建立了一个群体模型来描述纵向生活质量轨迹,并量化治疗毒性和疗效。在300多个筛选研究中,只有20个报告了纵向EORTC QLQ-C30生活质量数据。其中,8项研究报告派姆单抗组与对照组的生活质量结果无组间差异。然而,我们的模型显示pembrolizumab与更大的毒性相关,但改善了长期生活质量。值得注意的是,我们的方法确定了原始出版物中传统统计分析未检测到的治疗对生活质量的影响。综上所述,本研究表明MBMA结合种群模型可以更准确地评估纵向PROs数据,克服了传统方法的局限性。该方法为将以患者为中心的结果整合到肿瘤药物开发中提供了一个强大的框架,并支持在监管和临床决策中更广泛地使用PROs数据。
{"title":"A Model-Based Meta-Analysis of Pembrolizumab Effects on Patient-Reported Quality of Life: Advancing Patient-Centered Oncology Drug Development.","authors":"Yiqin Zou, Yimeng Sun, Sudhamshu Ravva, Lynne I Wagner, Jiawei Zhou","doi":"10.1002/psp4.70106","DOIUrl":"https://doi.org/10.1002/psp4.70106","url":null,"abstract":"<p><p>Pembrolizumab is an immune checkpoint inhibitor that has been approved for more than 20 different indications and has shown great survival benefits in various types of cancer. However, the reported benefits of pembrolizumab in patients' quality of life (QoL) have been inconsistent across different studies and different types of cancer. As oncology drug development increasingly emphasizes patient-centered care, patient-reported outcomes (PROs), particularly patient-reported QoL, are recognized as important clinical endpoints. To characterize the effects of pembrolizumab on patient-reported QoL, we conducted a model-based meta-analysis (MBMA) of published clinical trials evaluating pembrolizumab across different types of cancer. The longitudinal EORTC QLQ-C30 GHS/QOL data were extracted in our analysis as QoL scores. A population model was developed to characterize the longitudinal QoL trajectories and quantify both treatment toxicity and efficacy. Out of more than 300 screened studies, only 20 reported longitudinal EORTC QLQ-C30 QoL data. Among these, 8 studies reported no between-group differences in QoL outcomes between pembrolizumab and control arms. However, our modeling revealed that pembrolizumab was associated with greater toxicity but improved long-term QoL. Notably, our approach identified treatment effects on QoL that were not detected by traditional statistical analyses in the original publications. In summary, our study demonstrates that MBMA combined with population modeling enables more accurate evaluation of longitudinal PROs data, overcoming the limitations of conventional methods. This approach offers a robust framework for integrating patient-centered outcomes into oncology drug development and supports the broader use of PROs data in regulatory and clinical decision-making.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198648","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PD-1-Cis IL-2R Agonism Determines the Predicted Pharmacological Dose Range for the Immunocytokine Eciskafusp Alfa (PD1-IL2v) PD-1-Cis IL-2R激动作用决定免疫细胞因子Eciskafusp α (PD1-IL2v)的预测药理学剂量范围。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-09-27 DOI: 10.1002/psp4.70112
Lucy G. Hutchinson, Thomas D. Lewin, Laura Lauener, Meret Martin-Facklam, Merlind Muecke, Volker Teichgraeber, Laura Codarri Deak

Binding in cis-configuration to the PD-1 receptor (PD-1) and IL-2βγ receptor (IL-2Rβγ) has been shown to lead to differentiation of CD8 T cells to better effectors, which is anticipated to drive efficacy of the immune-targeted cytokine eciskafusp alfa, or PD1-IL2v. Here we present a geometrically driven mathematical formulation informed by in vitro and early clinical data which enables prediction of doses at which cis-binding is at its highest, and explains observed differences in concentration-time profiles for patients who had recent exposure to other anti-PD-1 molecules compared with those who had not. Furthermore, binding in cis-configuration is expected to follow a “bell-shaped” relationship with drug concentration such that high concentrations may lead to reduced benefit/risk ratio compared with concentrations around the peak of the bell-shape. Model simulations identify patient cohorts for whom the upper limit of the pharmacological dosing range may be defined by either undesirable off-tumor target engagement or a decrease in on-tumor cis-binding.

以顺式结构结合PD-1受体(PD-1)和IL-2 βγ $$ beta gamma $$受体(IL-2R βγ $$ beta gamma $$)已被证明可导致CD8 T细胞分化为更好的效应器,这有望推动免疫靶向细胞因子eciskafusp alfa或PD1-IL2v的功效。在这里,我们提出了一个几何驱动的数学公式,根据体外和早期临床数据,可以预测顺式结合最高的剂量,并解释了最近暴露于其他抗pd -1分子的患者与未暴露于其他抗pd -1分子的患者在浓度-时间谱上的观察差异。此外,顺式结构的结合预计与药物浓度呈“钟形”关系,即与钟形峰值附近的浓度相比,高浓度可能导致收益/风险比降低。模型模拟确定了药理学剂量范围上限可能由不希望的肿瘤外靶点接合或肿瘤上顺式结合减少来定义的患者队列。
{"title":"PD-1-Cis IL-2R Agonism Determines the Predicted Pharmacological Dose Range for the Immunocytokine Eciskafusp Alfa (PD1-IL2v)","authors":"Lucy G. Hutchinson,&nbsp;Thomas D. Lewin,&nbsp;Laura Lauener,&nbsp;Meret Martin-Facklam,&nbsp;Merlind Muecke,&nbsp;Volker Teichgraeber,&nbsp;Laura Codarri Deak","doi":"10.1002/psp4.70112","DOIUrl":"10.1002/psp4.70112","url":null,"abstract":"<p>Binding in cis-configuration to the PD-1 receptor (PD-1) and IL-2<span></span><math>\u0000 <semantics>\u0000 <mrow>\u0000 <mi>βγ</mi>\u0000 </mrow>\u0000 </semantics></math> receptor (IL-2R<span></span><math>\u0000 <semantics>\u0000 <mrow>\u0000 <mi>βγ</mi>\u0000 </mrow>\u0000 </semantics></math>) has been shown to lead to differentiation of CD8 T cells to better effectors, which is anticipated to drive efficacy of the immune-targeted cytokine eciskafusp alfa, or PD1-IL2v. Here we present a geometrically driven mathematical formulation informed by in vitro and early clinical data which enables prediction of doses at which cis-binding is at its highest, and explains observed differences in concentration-time profiles for patients who had recent exposure to other anti-PD-1 molecules compared with those who had not. Furthermore, binding in cis-configuration is expected to follow a “bell-shaped” relationship with drug concentration such that high concentrations may lead to reduced benefit/risk ratio compared with concentrations around the peak of the bell-shape. Model simulations identify patient cohorts for whom the upper limit of the pharmacological dosing range may be defined by either undesirable off-tumor target engagement or a decrease in on-tumor cis-binding.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2128-2138"},"PeriodicalIF":3.0,"publicationDate":"2025-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70112","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145181798","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
Evaluation and Mitigation of Time-Dependent Confounding Effects in Conventional Exposure-Response Analyses for Oncology Drugs 肿瘤药物常规暴露-反应分析中时间依赖性混杂效应的评估和缓解。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-09-25 DOI: 10.1002/psp4.70119
Xuefen Yin, Ye Xiong, Youwei Bi, Xin Wei, Hong Zhao, Elimika Pfuma Fletcher, Rajanikanth Madabushi, Amal Ayyoub, Hao Zhu, Stephan Schmidt, Jiang Liu

Conventional exposure–response (E–R) analyses, such as logistic regression and time-to-event analysis using summary exposure metrics, are often conducted in oncology using data from the pivotal trial(s) with a single dose level to support dosing decisions. However, these E–R analyses, affected by multiple confounding factors, may mischaracterize true E–R relationships, potentially limiting their utility in dosing decisions. This study investigates potential mischaracterization in such analyses influenced by the following time-dependent confounding factors: exposure accumulation, dose modification patterns, and event onset time. We used a simulation-based approach to evaluate two E–R scenarios: ER1, where event time generated with a Weibull distribution is not affected by drug exposure, and ER2, where the response is driven by drug exposure via a joint PK-tumor size model. Our analyses indicate that when using time-dependent exposure metrics (e.g., average concentration till event/censoring), exposure accumulation tends to induce an inverse E–R trend, while dose modifications (interruptions/reductions) likely induce a positive E–R trend. Simulations suggest that employing static exposure metrics (e.g., first-cycle or steady-state) minimizes these biases. Additionally, adopting an Emax model aligned with the underground truth in ER2 in the E–R analyses could reduce bias. When significant dose modifications are present, including relevant data from a dose-range study and employing modified methods for time-dependent exposure derivation may help mitigate bias. Overall, using multiple exposure metrics (including static ones) to assess E–R consistency and interpreting the potential causal effects with totality of evidence (including dose–response results) should be implemented to better inform dosing decisions.

传统的暴露-反应(E-R)分析,如使用总结暴露指标的逻辑回归和时间-事件分析,通常在肿瘤学中使用单一剂量水平的关键试验数据来支持剂量决策。然而,这些E-R分析受到多种混杂因素的影响,可能会错误地描述真正的E-R关系,从而可能限制其在剂量决策中的效用。本研究调查了受以下时间相关混杂因素影响的此类分析中可能存在的错误描述:暴露积累、剂量调整模式和事件发生时间。我们使用基于模拟的方法来评估两种E-R情景:ER1,其中威布尔分布产生的事件时间不受药物暴露的影响,ER2,其中反应由药物暴露驱动,通过联合kp -肿瘤大小模型。我们的分析表明,当使用与时间相关的暴露指标(例如,事件/审查前的平均浓度)时,暴露积累倾向于诱导反向E-R趋势,而剂量变化(中断/减少)可能诱导正E-R趋势。模拟表明,采用静态暴露度量(例如,第一周期或稳态)可以最大限度地减少这些偏差。此外,在E-R分析中采用与ER2地下真相一致的Emax模型可以减少偏差。当存在显著的剂量变化时,包括来自剂量范围研究的相关数据并采用经修改的时间依赖性暴露推导方法可能有助于减轻偏倚。总体而言,应使用多种暴露指标(包括静态暴露指标)来评估E-R一致性,并利用全部证据(包括剂量-反应结果)解释潜在的因果效应,以便更好地为剂量决策提供信息。
{"title":"Evaluation and Mitigation of Time-Dependent Confounding Effects in Conventional Exposure-Response Analyses for Oncology Drugs","authors":"Xuefen Yin,&nbsp;Ye Xiong,&nbsp;Youwei Bi,&nbsp;Xin Wei,&nbsp;Hong Zhao,&nbsp;Elimika Pfuma Fletcher,&nbsp;Rajanikanth Madabushi,&nbsp;Amal Ayyoub,&nbsp;Hao Zhu,&nbsp;Stephan Schmidt,&nbsp;Jiang Liu","doi":"10.1002/psp4.70119","DOIUrl":"10.1002/psp4.70119","url":null,"abstract":"<p>Conventional exposure–response (E–R) analyses, such as logistic regression and time-to-event analysis using summary exposure metrics, are often conducted in oncology using data from the pivotal trial(s) with a single dose level to support dosing decisions. However, these E–R analyses, affected by multiple confounding factors, may mischaracterize true E–R relationships, potentially limiting their utility in dosing decisions. This study investigates potential mischaracterization in such analyses influenced by the following time-dependent confounding factors: exposure accumulation, dose modification patterns, and event onset time. We used a simulation-based approach to evaluate two E–R scenarios: ER1, where event time generated with a Weibull distribution is not affected by drug exposure, and ER2, where the response is driven by drug exposure via a joint PK-tumor size model. Our analyses indicate that when using time-dependent exposure metrics (e.g., average concentration till event/censoring), exposure accumulation tends to induce an inverse E–R trend, while dose modifications (interruptions/reductions) likely induce a positive E–R trend. Simulations suggest that employing static exposure metrics (e.g., first-cycle or steady-state) minimizes these biases. Additionally, adopting an <i>E</i>max model aligned with the underground truth in ER2 in the E–R analyses could reduce bias. When significant dose modifications are present, including relevant data from a dose-range study and employing modified methods for time-dependent exposure derivation may help mitigate bias. Overall, using multiple exposure metrics (including static ones) to assess E–R consistency and interpreting the potential causal effects with totality of evidence (including dose–response results) should be implemented to better inform dosing decisions.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2118-2127"},"PeriodicalIF":3.0,"publicationDate":"2025-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70119","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145148023","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 Drug Development of Subcutaneous Nivolumab: Comparison of Pharmacokinetic Analysis Methodologies Using Clinical Trial Simulation 基于模型的皮下纳武单抗药物开发:使用临床试验模拟的药代动力学分析方法的比较。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-09-24 DOI: 10.1002/psp4.70120
Yue Zhao, Heather Vezina, Zheyi Hu, Anna Kondic, Li Zhu, Amit Roy

A subcutaneous formulation of nivolumab was evaluated in the phase III study CheckMate 67T (NCT04810078). The co-primary pharmacokinetic exposure endpoints, time-averaged serum concentration over the first 28 days (Cavgd28), and steady-state trough concentration (Cminss) were determined through population pharmacokinetic analysis as compared to conventional non-compartmental analysis (NCA). We proposed a model-based approach to determine subcutaneous and intravenous nivolumab exposures in CheckMate 67T, leveraging extensive prior pharmacokinetic data across various tumor types. The robustness of this model-informed drug development (MIDD) approach was assessed via clinical trial simulations. Concentration–time profiles in randomly sampled patients with renal cell carcinoma receiving second-line systemic therapy were simulated for subcutaneous/intravenous nivolumab. Population pharmacokinetic parameters, sampled from the joint parameter uncertainty distribution, were applied in simulations based on the CheckMate 67T design. Two population pharmacokinetic approaches—the PRIOR subroutine ($PRIOR) in NONMEM and a pooled analysis with historical nivolumab pharmacokinetic data—were used to analyze the simulated data. Results were compared to NCA using intensive and conventional sampling schemes. Analyses showed that model-based analysis provided more accurate area under the curve estimates than NCA. Model-predicted exposure measures, including Cavgd28, maximum serum concentration after the first dose, and minimum serum concentration at day 28, were also consistent across both population pharmacokinetic approaches, with minimal differences in geometric means. In conclusion, both the $PRIOR and pooled population pharmacokinetic methods yielded more accurate results compared to conventional NCA. The MIDD approach was validated as a robust and feasible method to support non-inferiority assessment based on clinical trial simulations.

nivolumab的皮下制剂在CheckMate 67T (NCT04810078)的III期研究中进行了评估。与传统的非区室分析(NCA)相比,通过群体药代动力学分析确定了共主要药代动力学暴露终点、前28天的时间平均血清浓度(Cavgd28)和稳态谷浓度(Cminss)。我们提出了一种基于模型的方法来确定CheckMate 67T的皮下和静脉纳武单抗暴露,利用各种肿瘤类型的广泛的先前药代动力学数据。通过临床试验模拟评估了这种基于模型的药物开发(MIDD)方法的稳健性。随机抽样接受二线全身治疗的肾细胞癌患者的浓度-时间分布模拟皮下/静脉纳武单抗。基于CheckMate 67T设计,从联合参数不确定性分布中抽取种群药代动力学参数进行模拟。使用两种群体药代动力学方法- NONMEM中的PRIOR子程序($PRIOR)和具有历史纳武单抗药代动力学数据的汇总分析来分析模拟数据。结果比较了NCA使用密集和常规抽样方案。分析表明,基于模型的分析提供了比NCA更准确的曲线下面积估计。模型预测的暴露测量,包括Cavgd28,第一次给药后的最大血清浓度和第28天的最低血清浓度,在两种人群药代动力学方法中也是一致的,几何平均差异很小。总之,与传统的NCA相比,$PRIOR和集合人群药代动力学方法产生了更准确的结果。MIDD方法被证实是一种稳健可行的方法,可以支持基于临床试验模拟的非劣效性评估。
{"title":"Model-Informed Drug Development of Subcutaneous Nivolumab: Comparison of Pharmacokinetic Analysis Methodologies Using Clinical Trial Simulation","authors":"Yue Zhao,&nbsp;Heather Vezina,&nbsp;Zheyi Hu,&nbsp;Anna Kondic,&nbsp;Li Zhu,&nbsp;Amit Roy","doi":"10.1002/psp4.70120","DOIUrl":"10.1002/psp4.70120","url":null,"abstract":"<p>A subcutaneous formulation of nivolumab was evaluated in the phase III study CheckMate 67T (NCT04810078). The co-primary pharmacokinetic exposure endpoints, time-averaged serum concentration over the first 28 days (<i>C</i><sub>avgd28</sub>), and steady-state trough concentration (<i>C</i><sub>minss</sub>) were determined through population pharmacokinetic analysis as compared to conventional non-compartmental analysis (NCA). We proposed a model-based approach to determine subcutaneous and intravenous nivolumab exposures in CheckMate 67T, leveraging extensive prior pharmacokinetic data across various tumor types. The robustness of this model-informed drug development (MIDD) approach was assessed via clinical trial simulations. Concentration–time profiles in randomly sampled patients with renal cell carcinoma receiving second-line systemic therapy were simulated for subcutaneous/intravenous nivolumab. Population pharmacokinetic parameters, sampled from the joint parameter uncertainty distribution, were applied in simulations based on the CheckMate 67T design. Two population pharmacokinetic approaches—the PRIOR subroutine ($PRIOR) in NONMEM and a pooled analysis with historical nivolumab pharmacokinetic data—were used to analyze the simulated data. Results were compared to NCA using intensive and conventional sampling schemes. Analyses showed that model-based analysis provided more accurate area under the curve estimates than NCA. Model-predicted exposure measures, including <i>C</i><sub>avgd28</sub>, maximum serum concentration after the first dose, and minimum serum concentration at day 28, were also consistent across both population pharmacokinetic approaches, with minimal differences in geometric means. In conclusion, both the $PRIOR and pooled population pharmacokinetic methods yielded more accurate results compared to conventional NCA. The MIDD approach was validated as a robust and feasible method to support non-inferiority assessment based on clinical trial simulations.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 12","pages":"2107-2117"},"PeriodicalIF":3.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70120","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145129873","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