首页 > 最新文献

CPT: Pharmacometrics & Systems Pharmacology最新文献

英文 中文
Characterizing Apixaban Pharmacokinetics Through Physiologically-Based Pharmacokinetic Modeling: Critical Role of Biliary Secretion and Enterohepatic Circulation in Humans 通过基于生理的药代动力学建模表征阿哌沙班的药代动力学:人类胆道分泌和肠肝循环的关键作用。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-02 DOI: 10.1002/psp4.70163
Toshiaki Tsuchitani, Wen Kou, Masatoshi Tomi, Yuichi Sugiyama

Apixaban, a factor Xa inhibitor, is a direct oral anticoagulant with a well-balanced elimination; it is eliminated evenly via feces, urine (with no active secretion), and as metabolites after oral administration. The common understanding is that biliary secretion and enterohepatic circulation (EHC) of apixaban are limited in humans, and that fecal excretion may be attributable to intestinal secretion. However, a decrease in apixaban blood concentration with activated charcoal coadministration in humans suggests possible involvement of EHC. This study aimed to evaluate the contribution of biliary excretion, EHC, and intestinal secretion to apixaban pharmacokinetics (PK) using a physiologically-based pharmacokinetic (PBPK) model. A top-down analysis was performed using blood concentration and mass balance data from healthy volunteers. Model parameters were optimized using the Cluster-Gauss Newton method (CGNM), followed by the bootstrap method. The model accurately described observed data and indicated moderate to high biliary secretion relative to metabolic clearance. Simulated biliary secretion into the duodenum well predicted the biliary secretion data in humans (< 1% of dose at 8 h post-dose). Virtual knockout of EHC resulted in a shortened half-life from 8.7 to 2.9 h, and 17% and 55% decrease in area under the concentration curve (AUC) and fecal excretion after intravenous dosing, respectively, confirming the significant contribution of biliary excretion and EHC. The model also accurately described apixaban PK with activated charcoal coadministration at 2 or 6 h post-dose. Although further experimental validation (e.g., sandwich-cultured hepatocytes) would strengthen these findings, our study demonstrates that biliary secretion and EHC play a substantial role in apixaban elimination and disposition in humans.

阿哌沙班是一种Xa因子抑制剂,是一种直接口服抗凝剂,具有良好的消除平衡;经粪、尿(无活性分泌)及口服后作为代谢物均匀排出。普遍的理解是阿哌沙班在人体内的胆道分泌和肠肝循环(EHC)是有限的,粪便排泄可能归因于肠道分泌。然而,人类与活性炭共给药时阿哌沙班血药浓度的降低提示可能涉及EHC。本研究旨在通过基于生理的药代动力学(PBPK)模型评估胆道排泄、EHC和肠道分泌对阿哌沙班药代动力学(PK)的贡献。使用健康志愿者的血液浓度和物质平衡数据进行了自上而下的分析。采用聚类-高斯牛顿法(CGNM)对模型参数进行优化,然后采用自举法对模型参数进行优化。该模型准确地描述了观察到的数据,并表明相对于代谢清除率,胆分泌中度至高。模拟胆汁分泌进入十二指肠,很好地预测了人类的胆汁分泌数据(
{"title":"Characterizing Apixaban Pharmacokinetics Through Physiologically-Based Pharmacokinetic Modeling: Critical Role of Biliary Secretion and Enterohepatic Circulation in Humans","authors":"Toshiaki Tsuchitani,&nbsp;Wen Kou,&nbsp;Masatoshi Tomi,&nbsp;Yuichi Sugiyama","doi":"10.1002/psp4.70163","DOIUrl":"10.1002/psp4.70163","url":null,"abstract":"<p>Apixaban, a factor Xa inhibitor, is a direct oral anticoagulant with a well-balanced elimination; it is eliminated evenly via feces, urine (with no active secretion), and as metabolites after oral administration. The common understanding is that biliary secretion and enterohepatic circulation (EHC) of apixaban are limited in humans, and that fecal excretion may be attributable to intestinal secretion. However, a decrease in apixaban blood concentration with activated charcoal coadministration in humans suggests possible involvement of EHC. This study aimed to evaluate the contribution of biliary excretion, EHC, and intestinal secretion to apixaban pharmacokinetics (PK) using a physiologically-based pharmacokinetic (PBPK) model. A top-down analysis was performed using blood concentration and mass balance data from healthy volunteers. Model parameters were optimized using the Cluster-Gauss Newton method (CGNM), followed by the bootstrap method. The model accurately described observed data and indicated moderate to high biliary secretion relative to metabolic clearance. Simulated biliary secretion into the duodenum well predicted the biliary secretion data in humans (&lt; 1% of dose at 8 h post-dose). Virtual knockout of EHC resulted in a shortened half-life from 8.7 to 2.9 h, and 17% and 55% decrease in area under the concentration curve (AUC) and fecal excretion after intravenous dosing, respectively, confirming the significant contribution of biliary excretion and EHC. The model also accurately described apixaban PK with activated charcoal coadministration at 2 or 6 h post-dose. Although further experimental validation (e.g., sandwich-cultured hepatocytes) would strengthen these findings, our study demonstrates that biliary secretion and EHC play a substantial role in apixaban elimination and disposition in humans.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70163","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660522","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 Nonparametric Population Pharmacokinetic Model of Selumetinib in Pediatric Patients Diagnosed With Neurofibromatosis-I or Plexiform Neurofibromas Selumetinib在诊断为神经纤维瘤病i型或丛状神经纤维瘤的儿童患者中的非参数群体药代动力学模型
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-30 DOI: 10.1002/psp4.70156
Zoltán Köllő, Janka Kovács, Michael N. Neely, Barna Vásárhelyi, Edit Brückner, Attila J. Szabo, Miklós Garami, Gellért Balázs Karvaly

A twice-daily administration of oral selumetinib (SLT) in the fasted state is the only approved pharmaceutical option for treating inoperable neurofibromatosis type I (NF-1) and plexiform neurofibromas (PN). In children, exposure to SLT is highly variable, and fasting presents a substantial burden. Therapeutic drug monitoring and pharmacokinetic modeling can support individualized therapy accompanied by a more rational alimentary routine. Twenty-eight children diagnosed with inoperable NF-1 or PN were recruited at a major pediatric oncological center. Twenty-two patients donated 156 blood samples in steady state for nonparametric population pharmacokinetic modeling. An equation was developed experimentally for estimating model error. Eleven three-compartment models were compared in terms of statistical performance. Monte Carlo simulations were performed to validate a limited external model using six additional patients and to compare the trough-to-peak SLT concentration ratios simulated for various dosing regimens to develop better control over exposure. A pharmacokinetic model that included total body weight as a covariate provided the best fit between predicted and observed concentrations (r = 0.994) and the best performance statistics. In the first Monte Carlo simulation, measured concentrations fell within the 0.01%–95% (median: 19.7%) quantiles of the simulated ranges. The second simulation revealed that 6-h (q6h), 8-h (q8h), and 12-h (q12h) dosing intervals would yield comparable trough-to-peak concentration ratios, with medians of 0.126 (range: 0.001–0.335), 0.104 (0.000–0.306), and 0.065 (0.000–0.279), respectively. The nonparametric population model provides efficient priors for making individual predictions of SLT concentrations. The simulation did not reveal any disadvantages of q6h or q8h dosing.

在禁食状态下每日两次口服selumetinib (SLT)是唯一被批准用于治疗不能手术的I型神经纤维瘤病(NF-1)和丛状神经纤维瘤(PN)的药物选择。在儿童中,暴露于SLT是高度可变的,禁食是一个很大的负担。治疗药物监测和药代动力学模型可以支持个体化治疗,并伴有更合理的饮食常规。在一家主要的儿科肿瘤中心招募了28名被诊断为不能手术的NF-1或PN的儿童。22名患者捐献了156份稳态血液样本,用于非参数群体药代动力学建模。实验建立了模型误差估计方程。在统计性能方面比较了11个三室模型。通过蒙特卡罗模拟验证了另外6名患者的有限外部模型,并比较了不同给药方案模拟的SLT波谷-峰浓度比,以更好地控制暴露。以总体重为协变量的药代动力学模型提供了预测浓度与观察浓度之间的最佳拟合(r = 0.994)和最佳性能统计。在第一次蒙特卡罗模拟中,测量到的浓度落在模拟范围的0.01%-95%(中位数:19.7%)分位数内。第二次模拟显示,6小时(q6h)、8小时(q8h)和12小时(q12h)的给药间隔可以产生相当的波谷-峰浓度比,中位数分别为0.126(范围:0.001-0.335)、0.104(0.000-0.306)和0.065(0.000-0.279)。非参数总体模型为单独预测SLT浓度提供了有效的先验。模拟没有显示q6h或q8h给药的任何缺点。
{"title":"A Nonparametric Population Pharmacokinetic Model of Selumetinib in Pediatric Patients Diagnosed With Neurofibromatosis-I or Plexiform Neurofibromas","authors":"Zoltán Köllő,&nbsp;Janka Kovács,&nbsp;Michael N. Neely,&nbsp;Barna Vásárhelyi,&nbsp;Edit Brückner,&nbsp;Attila J. Szabo,&nbsp;Miklós Garami,&nbsp;Gellért Balázs Karvaly","doi":"10.1002/psp4.70156","DOIUrl":"10.1002/psp4.70156","url":null,"abstract":"<p>A twice-daily administration of oral selumetinib (SLT) in the fasted state is the only approved pharmaceutical option for treating inoperable neurofibromatosis type I (NF-1) and plexiform neurofibromas (PN). In children, exposure to SLT is highly variable, and fasting presents a substantial burden. Therapeutic drug monitoring and pharmacokinetic modeling can support individualized therapy accompanied by a more rational alimentary routine. Twenty-eight children diagnosed with inoperable NF-1 or PN were recruited at a major pediatric oncological center. Twenty-two patients donated 156 blood samples in steady state for nonparametric population pharmacokinetic modeling. An equation was developed experimentally for estimating model error. Eleven three-compartment models were compared in terms of statistical performance. Monte Carlo simulations were performed to validate a limited external model using six additional patients and to compare the trough-to-peak SLT concentration ratios simulated for various dosing regimens to develop better control over exposure. A pharmacokinetic model that included total body weight as a covariate provided the best fit between predicted and observed concentrations (<i>r</i> = 0.994) and the best performance statistics. In the first Monte Carlo simulation, measured concentrations fell within the 0.01%–95% (median: 19.7%) quantiles of the simulated ranges. The second simulation revealed that 6-h (q6h), 8-h (q8h), and 12-h (q12h) dosing intervals would yield comparable trough-to-peak concentration ratios, with medians of 0.126 (range: 0.001–0.335), 0.104 (0.000–0.306), and 0.065 (0.000–0.279), respectively. The nonparametric population model provides efficient priors for making individual predictions of SLT concentrations. The simulation did not reveal any disadvantages of q6h or q8h dosing.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70156","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647595","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 of the PK/PD Changes on MASLD-Related Population—An Example From Simultaneous Acetaminophen Parent-Metabolite PBPK/PD Modeling masld相关人群PK/PD变化的评估——以对乙酰氨基酚亲本代谢物PBPK/PD同步建模为例
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-25 DOI: 10.1002/psp4.70161
Shanshan Zhao, Lan Zhang

Patients with metabolic dysfunction-associated steatotic liver disease (MASLD) may exhibit altered pharmacokinetics (PK) and pharmacodynamics (PD) of drugs compared with healthy populations. However, no physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) model has been specifically developed for MASLD. Acetaminophen (APAP), a widely used analgesic, was selected to develop a PBPK/PD model predicting PK/PD changes of APAP and its metabolites in MASLD-related populations. Based on a comprehensive review of published APAP PK studies and examination of existing PBPK models, a simultaneous parent-metabolite PBPK model for APAP was developed and optimized in healthy people. The model simulated the dynamics of APAP and its five major metabolites: APAP-glucuronide (APAP-glu), APAP-sulfate (APAP-sul), N-acetyl-p-benzoquinone imine (NAPQI), APAP-cysteine (APAP-cys), and APAP-mercapturate (APAP-merc). The validated model was expanded to MASLD-related populations, including overweight, obese, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and cirrhosis with different severities. Finally, a PD model was integrated to correlate APAP's PK with pain relief scores. The PBPK model reproduced published clinical PK data for APAP and its metabolites in healthy and MASLD-related populations. At therapeutic doses, the toxic NAPQI remained at very low levels. APAP's pain relief efficacy was retained, but onset time may change in MASLD-related populations. This PBPK/PD approach provides a strategy for projecting drug exposure in MASLD-related populations, even without specific PK or PD data. It highlights modeling's utility for personalized medicine in MASLD patients and MASLD treatment drug development.

与健康人群相比,代谢功能障碍相关的脂肪变性肝病(MASLD)患者可能表现出药物的药代动力学(PK)和药效学(PD)的改变。然而,目前还没有专门针对MASLD的基于生理学的药代动力学/药效学(PBPK/PD)模型。选择广泛使用的镇痛药对乙酰氨基酚(APAP),建立预测masld相关人群中APAP及其代谢物PK/PD变化的PBPK/PD模型。在全面回顾已发表的APAP PK研究和检查现有PBPK模型的基础上,建立并优化了健康人APAP的同时亲本代谢物PBPK模型。该模型模拟了APAP及其五种主要代谢物:APAP-葡萄糖醛酸酯(APAP-glu)、APAP-硫酸酯(APAP-sul)、n -乙酰基-对苯醌亚胺(NAPQI)、APAP-半胱氨酸(APAP-cys)和APAP-巯基(APAP-merc)的动力学。经验证的模型扩展到masld相关人群,包括超重、肥胖、非酒精性脂肪性肝病(NAFLD)、非酒精性脂肪性肝炎(NASH)和不同严重程度的肝硬化。最后,将一个PD模型整合到APAP的PK与疼痛缓解评分之间。PBPK模型重现了健康人群和masld相关人群中APAP及其代谢物的临床PK数据。在治疗剂量下,毒性NAPQI保持在非常低的水平。APAP的镇痛效果保持不变,但在masld相关人群中起效时间可能发生变化。这种PBPK/PD方法提供了一种预测masld相关人群药物暴露的策略,即使没有特定的PK或PD数据。它强调了建模在MASLD患者的个性化医疗和MASLD治疗药物开发中的效用。
{"title":"Evaluation of the PK/PD Changes on MASLD-Related Population—An Example From Simultaneous Acetaminophen Parent-Metabolite PBPK/PD Modeling","authors":"Shanshan Zhao,&nbsp;Lan Zhang","doi":"10.1002/psp4.70161","DOIUrl":"10.1002/psp4.70161","url":null,"abstract":"<p>Patients with metabolic dysfunction-associated steatotic liver disease (MASLD) may exhibit altered pharmacokinetics (PK) and pharmacodynamics (PD) of drugs compared with healthy populations. However, no physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) model has been specifically developed for MASLD. Acetaminophen (APAP), a widely used analgesic, was selected to develop a PBPK/PD model predicting PK/PD changes of APAP and its metabolites in MASLD-related populations. Based on a comprehensive review of published APAP PK studies and examination of existing PBPK models, a simultaneous parent-metabolite PBPK model for APAP was developed and optimized in healthy people. The model simulated the dynamics of APAP and its five major metabolites: APAP-glucuronide (APAP-glu), APAP-sulfate (APAP-sul), N-acetyl-p-benzoquinone imine (NAPQI), APAP-cysteine (APAP-cys), and APAP-mercapturate (APAP-merc). The validated model was expanded to MASLD-related populations, including overweight, obese, nonalcoholic fatty liver disease (NAFLD), nonalcoholic steatohepatitis (NASH), and cirrhosis with different severities. Finally, a PD model was integrated to correlate APAP's PK with pain relief scores. The PBPK model reproduced published clinical PK data for APAP and its metabolites in healthy and MASLD-related populations. At therapeutic doses, the toxic NAPQI remained at very low levels. APAP's pain relief efficacy was retained, but onset time may change in MASLD-related populations. This PBPK/PD approach provides a strategy for projecting drug exposure in MASLD-related populations, even without specific PK or PD data. It highlights modeling's utility for personalized medicine in MASLD patients and MASLD treatment drug development.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70161","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602831","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
B Cell Differentiation Model for Identifying Predictors of Responses to Rituximab-Mediated B Cell Depletion in Rheumatic Diseases B细胞分化模型用于识别风湿疾病对利妥昔单抗介导的B细胞耗竭反应的预测因子。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-25 DOI: 10.1002/psp4.70151
Tomohisa Nakada, Donald E. Mager

Rituximab (RTX), an anti-CD20 monoclonal antibody, has been used to treat autoimmune diseases such as rheumatoid arthritis (RA). However, variability in therapeutic response to RTX remains a challenge. Here, a systems model is developed to mimic B cell differentiation leading to antibody-secreting cells (ASCs), including plasmablasts (PBs) and plasma cells (PCs). The model features the localization of B cell subsets in the bone marrow and secondary lymphoid organs and incorporates the internalization process of the CD20–RTX complex. To reproduce clinical data from patients with RA receiving RTX and glucocorticoids, pharmacokinetic models for the drugs were built and respective pharmacodynamic profiles of CD19+ and CD20+ cells and PBs were well captured by optimizing model parameters, which were estimated with good precision. As ASCs are the primary source of pathogenic autoantibodies in RA, the extent and duration of ASC depletion were hypothesized as drivers of therapeutic response to RTX. Global sensitivity analyses identified the CD20–RTX binding affinity and elimination rate constant (i.e., Fcγ-mediated degradation, internalization) as major determinants of both CD19+ cells and ASCs. The influence of baseline PBs and PCs on ASCs was also suggested, providing potential mechanisms underlying responder and non-responder variability. The model accurately reproduced the temporal changes in CD19+ cells after combination treatment with RTX and glucocorticoids suggesting successful model validation. This study provides a mechanistic framework and insights into key drivers of responses to CD20-depletion treatment using B cell dynamics as an indirect biomarker of clinical endpoints, which might ultimately improve therapeutic outcomes.

Rituximab (RTX)是一种抗cd20单克隆抗体,已被用于治疗自身免疫性疾病,如类风湿关节炎(RA)。然而,RTX治疗反应的可变性仍然是一个挑战。本文建立了一个系统模型来模拟导致抗体分泌细胞(ASCs)的B细胞分化,包括浆母细胞(PBs)和浆细胞(PCs)。该模型的特点是B细胞亚群定位于骨髓和次级淋巴器官,并纳入了CD20-RTX复合物的内化过程。为了重现接受RTX和糖皮质激素治疗的RA患者的临床数据,我们建立了药物的药代动力学模型,并通过优化模型参数,很好地捕获了CD19+、CD20+细胞和PBs各自的药效学特征,估计精度较高。由于ASC是RA中致病性自身抗体的主要来源,ASC消耗的程度和持续时间被假设为RTX治疗反应的驱动因素。全局敏感性分析发现CD20-RTX结合亲和力和消除速率常数(即fc γ介导的降解、内化)是CD19+细胞和ASCs的主要决定因素。还提出了基线PBs和PCs对ASCs的影响,提供了应答者和非应答者变异的潜在机制。该模型准确再现了RTX和糖皮质激素联合治疗后CD19+细胞的时间变化,表明模型验证成功。这项研究提供了一个机制框架和洞察cd20耗竭治疗反应的关键驱动因素,使用B细胞动力学作为临床终点的间接生物标志物,这可能最终改善治疗结果。
{"title":"B Cell Differentiation Model for Identifying Predictors of Responses to Rituximab-Mediated B Cell Depletion in Rheumatic Diseases","authors":"Tomohisa Nakada,&nbsp;Donald E. Mager","doi":"10.1002/psp4.70151","DOIUrl":"10.1002/psp4.70151","url":null,"abstract":"<p>Rituximab (RTX), an anti-CD20 monoclonal antibody, has been used to treat autoimmune diseases such as rheumatoid arthritis (RA). However, variability in therapeutic response to RTX remains a challenge. Here, a systems model is developed to mimic B cell differentiation leading to antibody-secreting cells (ASCs), including plasmablasts (PBs) and plasma cells (PCs). The model features the localization of B cell subsets in the bone marrow and secondary lymphoid organs and incorporates the internalization process of the CD20–RTX complex. To reproduce clinical data from patients with RA receiving RTX and glucocorticoids, pharmacokinetic models for the drugs were built and respective pharmacodynamic profiles of CD19<sup>+</sup> and CD20<sup>+</sup> cells and PBs were well captured by optimizing model parameters, which were estimated with good precision. As ASCs are the primary source of pathogenic autoantibodies in RA, the extent and duration of ASC depletion were hypothesized as drivers of therapeutic response to RTX. Global sensitivity analyses identified the CD20–RTX binding affinity and elimination rate constant (i.e., Fcγ-mediated degradation, internalization) as major determinants of both CD19<sup>+</sup> cells and ASCs. The influence of baseline PBs and PCs on ASCs was also suggested, providing potential mechanisms underlying responder and non-responder variability. The model accurately reproduced the temporal changes in CD19<sup>+</sup> cells after combination treatment with RTX and glucocorticoids suggesting successful model validation. This study provides a mechanistic framework and insights into key drivers of responses to CD20-depletion treatment using B cell dynamics as an indirect biomarker of clinical endpoints, which might ultimately improve therapeutic outcomes.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70151","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602798","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
Exposure-Efficacy Meta-Model of Nintedanib in Adult Patients With Chronic Fibrosing Interstitial Lung Diseases 尼达尼布治疗成人慢性纤维化间质性肺疾病的暴露-疗效meta模型
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1002/psp4.70132
Sonja Hartmann, Julie Janssen, Jakob Ribbing, Susanne Stowasser, Julia Korell

The tyrosine kinase inhibitor, nintedanib, reduces the rate of decline in forced vital capacity (FVC) in a comparable manner in patients with idiopathic pulmonary fibrosis (IPF), other forms of progressive pulmonary fibrosis (PPF), and systemic sclerosis-associated ILD (SSc-ILD). The recommended dose of nintedanib in all indications is 150 mg twice daily (BID). Data from Phase II and III trials in IPF, PPF, and SSc-ILD were incorporated into a meta-model to holistically investigate the relationship between nintedanib exposure and efficacy. Using data from 2642 patients with IPF, PPF, or SSc-ILD treated with nintedanib doses ranging from 50 to 150 mg BID, disease progression models with a maximum drug effect on the annual rate of change in absolute FVC (i.e., mL), FVC %predicted, and FVC Z-score were developed. The estimated plasma concentration producing 50% of the maximum drug effect (EC50) ranged from 6.21 to 10.4 nM (with respect to nintedanib trough concentration) across the explored FVC-based endpoints. While the disease progression for absolute FVC (mL), FVC %predicted, and FVC Z-score was different between IPF and PPF patients compared to SSc-ILD patients, the relative treatment effect of nintedanib, described by a disease-modifying Emax effect, was comparable across indications. The majority of patients achieve exposure levels at or exceeding the EC50 with the approved starting dose of 150 mg BID.

酪氨酸激酶抑制剂尼达尼布(nintedanib)在特发性肺纤维化(IPF)、其他形式的进行性肺纤维化(PPF)和系统性硬化症相关的ILD (SSc-ILD)患者中,以类似的方式降低了强迫肺活量(FVC)的下降速度。尼达尼布在所有适应症中的推荐剂量为150mg,每日两次(BID)。IPF、PPF和SSc-ILD的II期和III期试验数据被纳入一个元模型,以全面调查尼达尼布暴露与疗效之间的关系。使用来自2642例IPF、PPF或SSc-ILD患者的数据,尼达尼布的剂量范围为50 - 150mg BID,开发了对绝对FVC(即mL)、预测FVC %和FVC z评分的年变化率具有最大药物效应的疾病进展模型。在研究的基于fvc的终点上,产生最大药物效应50%的估计血浆浓度(EC50)范围为6.21至10.4 nM(相对于尼达尼布谷浓度)。虽然与SSc-ILD患者相比,IPF和PPF患者的绝对FVC (mL)、FVC %预测和FVC z -评分的疾病进展不同,但尼达尼布的相对治疗效果(由疾病改善Emax效应描述)在不同适应症之间具有可比性。大多数患者在批准的起始剂量为150mg BID时达到或超过EC50的暴露水平。
{"title":"Exposure-Efficacy Meta-Model of Nintedanib in Adult Patients With Chronic Fibrosing Interstitial Lung Diseases","authors":"Sonja Hartmann,&nbsp;Julie Janssen,&nbsp;Jakob Ribbing,&nbsp;Susanne Stowasser,&nbsp;Julia Korell","doi":"10.1002/psp4.70132","DOIUrl":"10.1002/psp4.70132","url":null,"abstract":"<p>The tyrosine kinase inhibitor, nintedanib, reduces the rate of decline in forced vital capacity (FVC) in a comparable manner in patients with idiopathic pulmonary fibrosis (IPF), other forms of progressive pulmonary fibrosis (PPF), and systemic sclerosis-associated ILD (SSc-ILD). The recommended dose of nintedanib in all indications is 150 mg twice daily (BID). Data from Phase II and III trials in IPF, PPF, and SSc-ILD were incorporated into a meta-model to holistically investigate the relationship between nintedanib exposure and efficacy. Using data from 2642 patients with IPF, PPF, or SSc-ILD treated with nintedanib doses ranging from 50 to 150 mg BID, disease progression models with a maximum drug effect on the annual rate of change in absolute FVC (i.e., mL), FVC %predicted, and FVC Z-score were developed. The estimated plasma concentration producing 50% of the maximum drug effect (EC<sub>50</sub>) ranged from 6.21 to 10.4 nM (with respect to nintedanib trough concentration) across the explored FVC-based endpoints. While the disease progression for absolute FVC (mL), FVC %predicted, and FVC Z-score was different between IPF and PPF patients compared to SSc-ILD patients, the relative treatment effect of nintedanib, described by a disease-modifying E<sub>max</sub> effect, was comparable across indications. The majority of patients achieve exposure levels at or exceeding the EC<sub>50</sub> with the approved starting dose of 150 mg BID.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70132","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145563184","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
Exposure–Response Modeling of Monthly Migraine Days for Efficacy of Atogepant in Patients With Episodic or Chronic Migraine 对发作性或慢性偏头痛患者的暴露-反应模型:每月偏头痛天数。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1002/psp4.70154
Louisa Schlachter, Denise Beck, Ramesh R. Boinpally, Sven Stodtmann

This work aimed to develop an appropriate model to evaluate the exposure–response relationship (ERR) for monthly migraine days (MMD) in atogepant's key migraine prevention clinical trials to support dose selection. The ERR between atogepant concentration and MMD over time was analyzed utilizing data from one phase 2b/3 and three phase 3 studies in patients with episodic or chronic migraine (EM/CM). Several distribution models were evaluated for placebo data, whereas two modified normal distributions were introduced enabling bounded MMD modeling. Exposure metrics and shapes for ERR were tested for the most suitable distribution. Stepwise covariate search, visual predictive checks, and plots of model-predicted MMD over the range of exposure metrics were utilized in model development, evaluation, and selection. The final MMD exposure–response model was able to model patients with EM/CM simultaneously and was based on a modified normal distribution with Emax ERR on Cmin. The model adequately described the observed data over time. Due to the Emax relationship, MMD at Week 9–12 plateaued around their model-based atogepant Cmin-EC90 of 3.71 nM, which is similar to most Cmin exposures seen at the 10 mg once-daily regimen. All approved atogepant dosages for EM/CM achieve effective concentrations to inhibit the calcitonin gene-peptide receptor by 90%. Patients who have been failed by conventional oral migraine preventive treatments or patients with a higher baseline MMD may require a longer treatment period to reach atogepant's maximal effect. No significant difference in efficacy was evident in patients exposed to prior oral migraine preventives compared to treatment-naïve patients.

本研究旨在建立一个合适的模型来评估atgegent关键偏头痛预防临床试验中每月偏头痛天数(MMD)的暴露-反应关系(ERR),以支持剂量选择。利用在发作性或慢性偏头痛(EM/CM)患者中进行的一项2b/3期和三项3期研究的数据,分析了伴随剂浓度与烟雾病之间随时间的ERR。对安慰剂数据的几种分布模型进行了评估,而引入了两种修正的正态分布,从而实现了有界烟雾模型。测试ERR的暴露度量和形状以确定最合适的分布。逐步协变量搜索、视觉预测检查和暴露度量范围内模型预测的烟雾度图被用于模型开发、评估和选择。最终的烟雾暴露-反应模型能够同时对EM/CM患者进行建模,并基于Emax ERR对Cmin的修正正态分布。该模型充分描述了随时间变化的观测数据。由于Emax关系,在第9-12周时,MMD在基于模型的联合剂Cmin- ec90为3.71 nM附近趋于稳定,这与大多数Cmin暴露在10 mg每日一次的方案中相似。所有批准的EM/CM联合剂剂量均达到抑制降钙素基因肽受体90%的有效浓度。常规口服偏头痛预防治疗失败的患者或基线烟雾度较高的患者可能需要更长的治疗期才能达到联合剂的最大效果。与treatment-naïve患者相比,先前暴露于口服偏头痛预防药物的患者在疗效上没有明显差异。
{"title":"Exposure–Response Modeling of Monthly Migraine Days for Efficacy of Atogepant in Patients With Episodic or Chronic Migraine","authors":"Louisa Schlachter,&nbsp;Denise Beck,&nbsp;Ramesh R. Boinpally,&nbsp;Sven Stodtmann","doi":"10.1002/psp4.70154","DOIUrl":"10.1002/psp4.70154","url":null,"abstract":"<p>This work aimed to develop an appropriate model to evaluate the exposure–response relationship (ERR) for monthly migraine days (MMD) in atogepant's key migraine prevention clinical trials to support dose selection. The ERR between atogepant concentration and MMD over time was analyzed utilizing data from one phase 2b/3 and three phase 3 studies in patients with episodic or chronic migraine (EM/CM). Several distribution models were evaluated for placebo data, whereas two modified normal distributions were introduced enabling bounded MMD modeling. Exposure metrics and shapes for ERR were tested for the most suitable distribution. Stepwise covariate search, visual predictive checks, and plots of model-predicted MMD over the range of exposure metrics were utilized in model development, evaluation, and selection. The final MMD exposure–response model was able to model patients with EM/CM simultaneously and was based on a modified normal distribution with <i>E</i><sub>max</sub> ERR on <i>C</i><sub>min</sub>. The model adequately described the observed data over time. Due to the <i>E</i><sub>max</sub> relationship, MMD at Week 9–12 plateaued around their model-based atogepant <i>C</i><sub>min</sub>-EC<sub>90</sub> of 3.71 nM, which is similar to most <i>C</i><sub>min</sub> exposures seen at the 10 mg once-daily regimen. All approved atogepant dosages for EM/CM achieve effective concentrations to inhibit the calcitonin gene-peptide receptor by 90%. Patients who have been failed by conventional oral migraine preventive treatments or patients with a higher baseline MMD may require a longer treatment period to reach atogepant's maximal effect. No significant difference in efficacy was evident in patients exposed to prior oral migraine preventives compared to treatment-naïve patients.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70154","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573380","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
Pharmacokinetic Model Selection for Personalized Infliximab Dosing in IBD IBD患者英夫利昔单抗个体化用药的药代动力学模型选择。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1002/psp4.70152
Sahira Chaiben, Peggy Gandia, Thibaut Jamme, Nicolas Congy, Didier Concordet

Infliximab, a monoclonal antibody used for immune-mediated diseases, shows high interpatient pharmacokinetic variability. Prolonged exposure increases the risk of adverse effects and costs, making dose personalization essential to balance safety, efficacy, and cost-effectiveness. Population pharmacokinetic models support individualized dosing, but different models may predict varying drug exposure for the same patient. This study aims to identify compatible models for each patient and assess the impact of model selection on dosing. This retrospective study included adult Crohn's disease patients receiving infliximab. Published pharmacokinetic models were screened. Model-patient compatibility was evaluated using Multivariate Exact Discrepancy through 100,000 Monte Carlo simulations. The Metropolis-Hastings algorithm generated individual parameter distributions. For each model-patient pair, the median and 90% confidence interval of the dose required to achieve a target exposure of 2079 mg*day/L were computed. Sixteen models were tested. No model was compatible with all patients. Dosing was calculated only for compatible pairs. The average median dose was 9.25 mg/kg, with an average imprecision of 6.63 mg/kg. The highest median dose reached 23.21 mg/kg, reflecting inter-model differences, while the greatest imprecision (25.69 mg/kg) stemmed from patient variability. This concentration-based method personalizes dosing via pharmacokinetic profiling. Patients can be classified into three groups: (1) those for whom all models provide similar recommendations, indicating high reliability across models; (2) those incompatible with all models, for whom the posology recommended by the manufacturer should be prioritized; and (3) those for whom some models are compatible but intensified therapeutic drug monitoring is required.

英夫利昔单抗是一种用于免疫介导疾病的单克隆抗体,在患者间表现出很高的药代动力学变异性。长时间接触增加了不良反应的风险和成本,使剂量个性化对于平衡安全性、有效性和成本效益至关重要。人群药代动力学模型支持个体化给药,但不同的模型可能预测同一患者不同的药物暴露。本研究旨在为每位患者确定兼容的模型,并评估模型选择对给药的影响。这项回顾性研究包括接受英夫利昔单抗治疗的成年克罗恩病患者。筛选已发表的药代动力学模型。通过100,000次蒙特卡罗模拟,使用多元精确差异评估模型患者相容性。Metropolis-Hastings算法生成单个参数分布。对于每对模型患者,计算达到2079 mg*day/L目标暴露所需剂量的中位数和90%置信区间。共测试了16个模型。没有一种模型与所有患者兼容。只计算相容对的剂量。平均中位剂量为9.25 mg/kg,平均不精确度为6.63 mg/kg。最高中位剂量达到23.21 mg/kg,反映了模型间的差异,而最大的不精确性(25.69 mg/kg)源于患者的差异。这种基于浓度的方法通过药代动力学分析个性化给药。患者可分为三组:(1)所有模型提供相似建议的患者,表明模型之间的可靠性较高;(2)与所有型号不兼容的,应优先考虑厂家推荐的型号;(3)某些模式兼容但需要加强治疗药物监测的。
{"title":"Pharmacokinetic Model Selection for Personalized Infliximab Dosing in IBD","authors":"Sahira Chaiben,&nbsp;Peggy Gandia,&nbsp;Thibaut Jamme,&nbsp;Nicolas Congy,&nbsp;Didier Concordet","doi":"10.1002/psp4.70152","DOIUrl":"10.1002/psp4.70152","url":null,"abstract":"<p>Infliximab, a monoclonal antibody used for immune-mediated diseases, shows high interpatient pharmacokinetic variability. Prolonged exposure increases the risk of adverse effects and costs, making dose personalization essential to balance safety, efficacy, and cost-effectiveness. Population pharmacokinetic models support individualized dosing, but different models may predict varying drug exposure for the same patient. This study aims to identify compatible models for each patient and assess the impact of model selection on dosing. This retrospective study included adult Crohn's disease patients receiving infliximab. Published pharmacokinetic models were screened. Model-patient compatibility was evaluated using Multivariate Exact Discrepancy through 100,000 Monte Carlo simulations. The Metropolis-Hastings algorithm generated individual parameter distributions. For each model-patient pair, the median and 90% confidence interval of the dose required to achieve a target exposure of 2079 mg*day/L were computed. Sixteen models were tested. No model was compatible with all patients. Dosing was calculated only for compatible pairs. The average median dose was 9.25 mg/kg, with an average imprecision of 6.63 mg/kg. The highest median dose reached 23.21 mg/kg, reflecting inter-model differences, while the greatest imprecision (25.69 mg/kg) stemmed from patient variability. This concentration-based method personalizes dosing via pharmacokinetic profiling. Patients can be classified into three groups: (1) those for whom all models provide similar recommendations, indicating high reliability across models; (2) those incompatible with all models, for whom the posology recommended by the manufacturer should be prioritized; and (3) those for whom some models are compatible but intensified therapeutic drug monitoring is required.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70152","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145563208","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
Exposure-Response Analysis for Time-to-Event Data in the Presence of Adaptive Dosing: Efficient Approaches and Pitfalls 自适应剂量下时间-事件数据的暴露-响应分析:有效方法和缺陷。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-20 DOI: 10.1002/psp4.70149
Alexandra Lavalley-Morelle, Félicien Le Louedec, Richard Anziano, France Mentré, Martin Bergstrand

Analyzing exposure-response (E-R) relationships for time-to-event (TTE) endpoints presents challenges due to the inherent time-dependent nature of the data. Some authors address these difficulties by using a fixed timepoint approach, where exposure is assessed at a predetermined time rather than dynamically over time. (e.g., initial exposure or last exposure). The aim of the current work is to compare the use of time-static and time-varying metrics to assess the E-R relationship through simulations. PK exposures were simulated from a one-compartment model and TTE data from a parametric proportional hazard model, involving the weekly average PK concentration as a time-varying covariate. Several scenarios were considered to handle the type of dosing (fixed or adaptive), the accumulation of the drug (low or strong), the type of event (efficacy, safety or independent), and the timing of the event onset (early or late). Wald tests on the exposure effect parameter were performed to assess the significance of the E-R relationship. For each simulation scenario, the type-I error and the power of the Wald tests were reported, revealing that no time-static metric consistently produced reliable results across all conditions. In order to ensure adequate statistical properties, we recommend using time-varying exposure, which shows good performance across all scenarios.

由于数据固有的时间依赖性,分析时间到事件(TTE)端点的暴露-响应(E-R)关系带来了挑战。一些作者通过使用固定时间点方法来解决这些困难,在这种方法中,暴露是在预定的时间而不是随时间动态评估的。(例如,初次暴露或最后暴露)。当前工作的目的是通过模拟比较使用时间静态和时变指标来评估E-R关系。采用单室模型模拟PK暴露,采用参数比例风险模型模拟TTE数据,将周平均PK浓度作为时变协变量。考虑了几种情况来处理剂量类型(固定或适应性),药物积累(低或强),事件类型(有效性,安全性或独立性)以及事件发生的时间(早或晚)。对暴露效应参数进行Wald检验,以评估E-R关系的显著性。对于每个模拟场景,报告了i型误差和Wald测试的功率,表明没有时间静态度量在所有条件下始终产生可靠的结果。为了确保充分的统计特性,我们建议使用时变曝光,它在所有场景中都显示出良好的性能。
{"title":"Exposure-Response Analysis for Time-to-Event Data in the Presence of Adaptive Dosing: Efficient Approaches and Pitfalls","authors":"Alexandra Lavalley-Morelle,&nbsp;Félicien Le Louedec,&nbsp;Richard Anziano,&nbsp;France Mentré,&nbsp;Martin Bergstrand","doi":"10.1002/psp4.70149","DOIUrl":"10.1002/psp4.70149","url":null,"abstract":"<p>Analyzing exposure-response (E-R) relationships for time-to-event (TTE) endpoints presents challenges due to the inherent time-dependent nature of the data. Some authors address these difficulties by using a fixed timepoint approach, where exposure is assessed at a predetermined time rather than dynamically over time. (e.g., initial exposure or last exposure). The aim of the current work is to compare the use of time-static and time-varying metrics to assess the E-R relationship through simulations. PK exposures were simulated from a one-compartment model and TTE data from a parametric proportional hazard model, involving the weekly average PK concentration as a time-varying covariate. Several scenarios were considered to handle the type of dosing (fixed or adaptive), the accumulation of the drug (low or strong), the type of event (efficacy, safety or independent), and the timing of the event onset (early or late). Wald tests on the exposure effect parameter were performed to assess the significance of the E-R relationship. For each simulation scenario, the type-I error and the power of the Wald tests were reported, revealing that no time-static metric consistently produced reliable results across all conditions. In order to ensure adequate statistical properties, we recommend using time-varying exposure, which shows good performance across all scenarios.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70149","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145563131","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 Advance of In Silico Evidence to Transform Pediatric Drug Development for Rare Diseases 计算机证据的进展将改变罕见病儿科药物的开发
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-18 DOI: 10.1002/psp4.70139
Jane Knöchel, Ping Zhao, Rajat Desikan, Jiawei Zhou, João A. Abrantes, Lutz Harnisch
<p>Rare diseases (RDs)—defined in the U.S. as those affecting fewer than 200,000 people and in the EU as fewer than 1 in 2000—represent a persistent unmet need. These differing definitions contribute to variation in reported numbers: the U.S. recognized over 7000 rare diseases impacting 25–30 million people (https://www.fda.gov/patients/rare-diseases-fda) while the EU estimates around 36 million affected individuals (https://www.ema.europa.eu/en/human-regulatory-overview/orphan-designation-overview). Most manifest early in life and progress relentlessly (around 70% [https://www.rarediseasesinternational.org/living-with-a-rare-disease/]), yet fewer than 5% currently have approved therapy [<span>1</span>]. Pediatric rare diseases amplify every obstacle of drug development: small and heterogeneous populations, ethical constraints and limited usefulness of conventional clinical trials.</p><p>Recognizing this urgency, initiatives such as the FDA's Rare Disease Innovation Hub and the LEADER 3D Program (https://www.fda.gov/about-fda/accelerating-rare-disease-cures-arc-program/learning-and-education-advance-and-empower-rare-disease-drug-developers-leader-3d) aim to accelerate the development of medicines. Still, as highlighted in Michelle Werner's ASCPT 2025 State-of-the-Art Lecture (https://ascpt2025.eventscribe.net/agenda.asp?BCFO=&pfp=BrowsebyDay&fa=&fb=&fc=&fd=&all=1), attention alone is not enough—innovation requires translation into action. Today, the growing availability of large-scale biological datasets and advanced modeling offers that opportunity. Pharmacometrics and systems pharmacology can transform sparse data into quantitative insights, enabling virtual exploration of therapies and supporting confident decision-making even in the absence of large trials.</p><p>A recent review by Chen et al. outlines the distinct challenges of pediatric RDs [<span>2</span>]—slow disease progression, limited natural-history data, genetic and phenotypic heterogeneity, and uncertain surrogate endpoints. These challenges call for a change in the mindset of conventional drug development, which is based on evidence generation through an extensive clinical program including multiple clinical trials.</p><p>Designing clinical trials for RDs, particularly those with genetic origins, presents unique challenges due to the difficulty in demonstrating immediate clinical improvement. Since resolving the root cause is often unattainable, the primary goal of most current RD treatment is typically to prevent disease progression rather than to elicit a rapid clinical response. This necessitates a deep understanding of the disease's progression timeline and the ability to model outcome metrics over time. Proof-of-concept (PoC) trials for RD often focus on detecting any treatment response—typically a binary outcome—using high-dose strategies to maximize the chance of observing an effect. However, predicting responses across a range of doses requires intr
罕见病(RDs)——在美国定义为患病人数少于20万人,在欧盟定义为2000年患病人数少于1人——是一个长期未得到满足的需求。这些不同的定义导致了报告数字的差异:美国确认了超过7000种罕见疾病,影响了2500万至3000万人(https://www.fda.gov/patients/rare-diseases-fda),而欧盟估计约有3600万人受影响(https://www.ema.europa.eu/en/human-regulatory-overview/orphan-designation-overview)。大多数表现在生命早期,并且持续发展(约70% [https://www.rarediseasesinternational.org/living-with-a-rare-disease/]]),但目前批准治疗的不到5%。儿科罕见病扩大了药物开发的每一个障碍:小而异质的人群,伦理约束和传统临床试验的有限效用。认识到这一紧迫性,FDA的罕见病创新中心和LEADER 3D计划(https://www.fda.gov/about-fda/accelerating-rare-disease-cures-arc-program/learning-and-education-advance-and-empower-rare-disease-drug-developers-leader-3d)等举措旨在加速药物的开发。然而,正如米歇尔·维尔纳在《2025年ASCPT最新技术讲座》(https://ascpt2025.eventscribe.net/agenda.asp?BCFO=&pfp=BrowsebyDay&fa=&fb=&fc=&fd=&all=1)中所强调的那样,仅仅关注是不够的——创新需要转化为行动。如今,大规模生物数据集和高级建模的日益普及提供了这样的机会。药物计量学和系统药理学可以将稀疏的数据转化为定量的见解,即使在没有大型试验的情况下,也可以对疗法进行虚拟探索,并支持自信的决策。Chen等人最近的一篇综述概述了儿科rd的独特挑战——疾病进展缓慢、自然病史数据有限、遗传和表型异质性以及替代终点不确定。这些挑战要求改变传统药物开发的思维方式,传统药物开发是基于通过包括多个临床试验在内的广泛临床项目来产生证据的。为rd设计临床试验,特别是那些具有遗传来源的rd设计临床试验,由于难以证明立即的临床改善,提出了独特的挑战。由于解决根本原因往往无法实现,目前大多数RD治疗的主要目标通常是防止疾病进展,而不是引起快速的临床反应。这就需要深入了解疾病的进展时间表,并能够随着时间的推移模拟结果指标。RD的概念验证(PoC)试验通常侧重于检测任何治疗反应-通常是二元结果-使用高剂量策略来最大化观察效果的机会。然而,预测不同剂量的反应需要复杂的潜在生物学机制知识,包括特定途径如何受到影响以及这些变化如何转化为可测量的临床结果。基于模型的药物开发(MIDD)方法——跨越药物计量学、定量系统药理学(QSP)和机器学习(ML)——为解决儿科RD的内在挑战提供了一种变革性的手段。这些计算工具通常用于现代药物开发,以增强我们对疾病和药物药理学的理解,并在整个药物开发生命周期的连续体中支持决策。包括儿科rd药物的开发和批准。《药物计量学和系统药理学》的这一主题问题提供了MIDD方法的作用和影响的观点,这些方法正在推进儿科罕见病患者的创新治疗。计算工具现在是现代研发研究的核心。它们允许试验的虚拟设计,治疗的机械探索,以及碎片化知识的自动合成,压缩了开发时间表,提高了决策质量。Duchenne Muscular Dystrophy (DMD)是一种主要影响男孩b[4]的进行性x连锁神经肌肉疾病,它体现了儿童RD药物开发中患者群体有限、表型异质性和伦理约束的挑战。两个互补的计算工具利用疾病进展模型在执行前模拟试验场景。第一个工具是试验模拟器,它可以帮助研究人员优化试验设计——样本量、持续时间和纳入标准——跨越五个常见的DMD功能终点[4]。使用这些工具的案例研究表明,在不影响统计能力的情况下,提高了试验效率,这在患者招募挑战中至关重要。另一个模拟界面结合了机器学习生成的虚拟种群和多变量模型,将功能结果与成像生物标志物[5]联系起来。 对最近试验数据的验证证实了其预测的准确性。直观的图形界面使临床领导和MIDD专家能够协作探索与各自临床药物开发相关的场景范围。QSP框架也在迅速发展。Meno-Tetang等人强调了QSP建模如何增强对生物动力学的理解,告知表达动力学和持久性,并指导剂量优化,同时减轻脱靶效应[6]。在RNA疗法、疫苗、基因和酶替代疗法中的应用表明,QSP模型现在支持设计、翻译和生命周期管理。在此基础上,Saini等人介绍了一种人工智能增强的QSP-Copilot,应用于凝血系统和戈谢病[7](一种隐性遗传溶酶体储存疾病,由葡萄糖脑苷酶缺乏引起,导致葡萄糖神经酰胺[8]积聚)。该工具实现了高精度的自动数据提取(99.1%和100%),同时最小的机构损失。这标志着向可扩展的、可用的、影响更大的QSP工具的关键转变,特别是在儿科rd中,深入的生物学见解是必不可少的。总之,这些进步说明了计算方法是如何在曾经被认为过于罕见或复杂而无法进行严格研究的情况下改变试验计划和转化决策的。随着治疗创新在儿科rd领域的扩展,基于模型的策略在克服小群体、异质人群和零散临床数据的局限性方面被证明是不可或缺的。从庞贝病的酶替代到失调性毛细血管扩张的类固醇给药,再到DMD、HoFH、CAH和ret驱动型癌症的暴露匹配剂量,药物计量学和系统建模实现了量身定制的方案、虚拟队列桥接和机制洞察。这些例子反映了儿科药物开发向精确和高效的更广泛转变——建模不仅仅是一种辅助工具,而是临床决策的核心驱动力。庞贝病(PD)就是一个显著的例子。PD是一种罕见的退行性多系统代谢紊乱,其α-葡萄糖苷酶缺陷导致糖原积聚。根据发病年龄的不同,晚期疾病(LOPD)会导致肌肉无力和呼吸功能不全,而早期疾病(IOPD)和更罕见的疾病会在出生后的第一年就导致心肌病的结果。Rachedi等人开发了一种机制QSP模型,将生物标志物与功能终点联系起来,并在晚发和早发PD患者之间架起虚拟队列,以优化avalglucosidase α -[9]的剂量。该方法为IOPD患儿确定了合适的治疗方案,而无需进行新的、更大规模的比较试验。在患有共济失调毛细血管扩张症(一种由共济失调毛细血管扩张突变基因[10]的双等位致病变异引起的神经退行性疾病)的患者中,一个例子说明了群体PK模型如何表征创新的给药系统。Ozdin等人整合了稀疏的儿童和健康成人数据,通过EryDex系统建立了持续地塞米松释放的模型,预测了每月输注支持长期类固醇治疗的安全、持续暴露,提高了依从性,降低了毒性[10]。罕见病试验,尤其是针对儿童的试验,往往样本量小,因此敏感的终点对于评估药物疗效至关重要。为了解决这个问题,Hamdan等人引入了一个项目反应理论框架,该框架联合模拟了退行性失调性疾病的临床报告(SARA)和数字运动结果,减少了不确定性,提高了统计能力,并有效地
{"title":"The Advance of In Silico Evidence to Transform Pediatric Drug Development for Rare Diseases","authors":"Jane Knöchel,&nbsp;Ping Zhao,&nbsp;Rajat Desikan,&nbsp;Jiawei Zhou,&nbsp;João A. Abrantes,&nbsp;Lutz Harnisch","doi":"10.1002/psp4.70139","DOIUrl":"https://doi.org/10.1002/psp4.70139","url":null,"abstract":"&lt;p&gt;Rare diseases (RDs)—defined in the U.S. as those affecting fewer than 200,000 people and in the EU as fewer than 1 in 2000—represent a persistent unmet need. These differing definitions contribute to variation in reported numbers: the U.S. recognized over 7000 rare diseases impacting 25–30 million people (https://www.fda.gov/patients/rare-diseases-fda) while the EU estimates around 36 million affected individuals (https://www.ema.europa.eu/en/human-regulatory-overview/orphan-designation-overview). Most manifest early in life and progress relentlessly (around 70% [https://www.rarediseasesinternational.org/living-with-a-rare-disease/]), yet fewer than 5% currently have approved therapy [&lt;span&gt;1&lt;/span&gt;]. Pediatric rare diseases amplify every obstacle of drug development: small and heterogeneous populations, ethical constraints and limited usefulness of conventional clinical trials.&lt;/p&gt;&lt;p&gt;Recognizing this urgency, initiatives such as the FDA's Rare Disease Innovation Hub and the LEADER 3D Program (https://www.fda.gov/about-fda/accelerating-rare-disease-cures-arc-program/learning-and-education-advance-and-empower-rare-disease-drug-developers-leader-3d) aim to accelerate the development of medicines. Still, as highlighted in Michelle Werner's ASCPT 2025 State-of-the-Art Lecture (https://ascpt2025.eventscribe.net/agenda.asp?BCFO=&amp;pfp=BrowsebyDay&amp;fa=&amp;fb=&amp;fc=&amp;fd=&amp;all=1), attention alone is not enough—innovation requires translation into action. Today, the growing availability of large-scale biological datasets and advanced modeling offers that opportunity. Pharmacometrics and systems pharmacology can transform sparse data into quantitative insights, enabling virtual exploration of therapies and supporting confident decision-making even in the absence of large trials.&lt;/p&gt;&lt;p&gt;A recent review by Chen et al. outlines the distinct challenges of pediatric RDs [&lt;span&gt;2&lt;/span&gt;]—slow disease progression, limited natural-history data, genetic and phenotypic heterogeneity, and uncertain surrogate endpoints. These challenges call for a change in the mindset of conventional drug development, which is based on evidence generation through an extensive clinical program including multiple clinical trials.&lt;/p&gt;&lt;p&gt;Designing clinical trials for RDs, particularly those with genetic origins, presents unique challenges due to the difficulty in demonstrating immediate clinical improvement. Since resolving the root cause is often unattainable, the primary goal of most current RD treatment is typically to prevent disease progression rather than to elicit a rapid clinical response. This necessitates a deep understanding of the disease's progression timeline and the ability to model outcome metrics over time. Proof-of-concept (PoC) trials for RD often focus on detecting any treatment response—typically a binary outcome—using high-dose strategies to maximize the chance of observing an effect. However, predicting responses across a range of doses requires intr","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"14 11","pages":"1731-1734"},"PeriodicalIF":3.0,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70139","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538066","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
Physiologically-Based Pharmacokinetic Modeling to Support Pediatric Clinical Development: An IQ Working Group Perspective on the Current Status and Challenges 基于生理的药代动力学建模以支持儿科临床发展:IQ工作组对现状和挑战的看法。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-18 DOI: 10.1002/psp4.70141
James W. T. Yates, Michael Zientek, Kunal S. Taskar, Wen Lin, Tycho Heimbach, Stefan Willmann, Jessica Rehmel, Neil Parrott, Michael Hanley, Justine Badee, Yuan Chen, Susan Cole, Loeckie De Zwart, Sebastian Haertter, Rongrong Jiang, Masakatsu Kotsuma, Guiqing Liang, Yu-Wei Lin, Jing Liu, Ying Ou, Juliane Rascher, Naveed A. Shaik, Jan Wahlstrom, Xiaofeng Wang, Guangqing Xiao, Ka Lai Yee, S. Y. Amy Cheung

Pediatric extrapolation strategies issued by health authorities have streamlined pediatric drug development and reduced the unnecessary burden of conducting pediatric clinical studies. In line with these strategies, physiologically based pharmacokinetic (PBPK) models have been utilized extensively for initial dosing regimen and sampling timepoint selection for pediatric studies, as well as dose validation throughout pediatric drug development. Here, the status and challenges of PBPK modeling in pediatric drug development have been summarized by the IQ Pediatric PBPK Working Group. Our work reviews current practices for pediatric PBPK modeling across various therapeutic areas. To enable best practice, we propose an optimized workflow for pediatric PBPK modeling recommendations. Two selected key pediatric PBPK case examples are also described, where modeling impacted the drug label extension to pediatric patients. Moreover, we analyze the current gaps and challenges in our understanding of drug absorption, distribution, metabolism, and elimination in pediatric PBPK model development. Since neonates are the least studied and the most medically fragile, the depth of our understanding of their rapidly evolving physiological processes is limited and so there exist significant modeling gaps which we summarize here. Finally, we provide recommendations, including building a public data repository, leveraging real-world data, and implementing microdose studies for addressing pediatric PBPK modeling challenges.

卫生当局发布的儿科外推策略简化了儿科药物开发,减少了进行儿科临床研究的不必要负担。与这些策略一致,基于生理的药代动力学(PBPK)模型已广泛用于儿科研究的初始给药方案和采样时间点选择,以及整个儿科药物开发过程中的剂量验证。在此,IQ儿科PBPK工作组总结了PBPK建模在儿科药物开发中的现状和挑战。我们的工作回顾了目前在不同治疗领域的儿科PBPK建模实践。为了实现最佳实践,我们提出了儿科PBPK建模建议的优化工作流程。还描述了两个选定的关键儿科PBPK案例示例,其中建模影响了药物标签扩展到儿科患者。此外,我们分析了目前在儿童PBPK模型开发中对药物吸收、分布、代谢和消除的理解方面的差距和挑战。由于新生儿是研究最少,医学上最脆弱的,我们对其快速进化的生理过程的理解深度有限,因此存在显着的建模差距,我们在这里总结。最后,我们提出建议,包括建立公共数据存储库,利用真实世界的数据,并实施微剂量研究,以解决儿科PBPK建模的挑战。
{"title":"Physiologically-Based Pharmacokinetic Modeling to Support Pediatric Clinical Development: An IQ Working Group Perspective on the Current Status and Challenges","authors":"James W. T. Yates,&nbsp;Michael Zientek,&nbsp;Kunal S. Taskar,&nbsp;Wen Lin,&nbsp;Tycho Heimbach,&nbsp;Stefan Willmann,&nbsp;Jessica Rehmel,&nbsp;Neil Parrott,&nbsp;Michael Hanley,&nbsp;Justine Badee,&nbsp;Yuan Chen,&nbsp;Susan Cole,&nbsp;Loeckie De Zwart,&nbsp;Sebastian Haertter,&nbsp;Rongrong Jiang,&nbsp;Masakatsu Kotsuma,&nbsp;Guiqing Liang,&nbsp;Yu-Wei Lin,&nbsp;Jing Liu,&nbsp;Ying Ou,&nbsp;Juliane Rascher,&nbsp;Naveed A. Shaik,&nbsp;Jan Wahlstrom,&nbsp;Xiaofeng Wang,&nbsp;Guangqing Xiao,&nbsp;Ka Lai Yee,&nbsp;S. Y. Amy Cheung","doi":"10.1002/psp4.70141","DOIUrl":"10.1002/psp4.70141","url":null,"abstract":"<p>Pediatric extrapolation strategies issued by health authorities have streamlined pediatric drug development and reduced the unnecessary burden of conducting pediatric clinical studies. In line with these strategies, physiologically based pharmacokinetic (PBPK) models have been utilized extensively for initial dosing regimen and sampling timepoint selection for pediatric studies, as well as dose validation throughout pediatric drug development. Here, the status and challenges of PBPK modeling in pediatric drug development have been summarized by the IQ Pediatric PBPK Working Group. Our work reviews current practices for pediatric PBPK modeling across various therapeutic areas. To enable best practice, we propose an optimized workflow for pediatric PBPK modeling recommendations. Two selected key pediatric PBPK case examples are also described, where modeling impacted the drug label extension to pediatric patients. Moreover, we analyze the current gaps and challenges in our understanding of drug absorption, distribution, metabolism, and elimination in pediatric PBPK model development. Since neonates are the least studied and the most medically fragile, the depth of our understanding of their rapidly evolving physiological processes is limited and so there exist significant modeling gaps which we summarize here. Finally, we provide recommendations, including building a public data repository, leveraging real-world data, and implementing microdose studies for addressing pediatric PBPK modeling challenges.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"15 1","pages":""},"PeriodicalIF":3.0,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.70141","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145548729","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