首页 > 最新文献

Journal of Biopharmaceutical Statistics最新文献

英文 中文
The proportional treatment effect: A metric that empowers and connects. 比例治疗效果:一种赋权和联系的指标。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-19 DOI: 10.1080/10543406.2025.2602479
Guoqiao Wang, Yijie Liao, Caiyan Li, Kun Jin, Yan Li, Gary Cutter

Clinical trials with continuous endpoints evaluate efficacy by comparing the difference in mean changes from baseline between groups. However, clinicians often interpret results in terms of a proportional reduction rather than an absolute difference. An alternative approach is to reparametrize this difference as a proportional treatment effect (PTE), calculated by dividing the difference by the placebo mean change. PTE is not a new metric per se, but a specific reparameterization gaining traction in certain clinical contexts. We demonstrate that, in theory, PTE can be more powerful than the simple difference in means while still controlling the type I error rate. This is achieved using the delta method, as implemented in well-established computational tools like the R package 'msm' and the SAS procedure 'NLMIXED'. By analyzing data from phase III trials, we illustrate how a PTE connects treatment outcomes across various endpoints and different presentation formats. The availability of these well-established statistical tools for estimating proportional treatment effects, combined with this theoretical demonstration, suggests an alternative test statistic for clinical trials with continuous endpoints.

具有连续终点的临床试验通过比较两组从基线到平均变化的差异来评估疗效。然而,临床医生通常根据比例减少而不是绝对差异来解释结果。另一种方法是将这种差异作为比例治疗效果(PTE)重新参数化,通过将差异除以安慰剂的平均变化来计算。PTE本身并不是一个新的度量,而是在某些临床环境中获得牵引力的特定重新参数化。我们证明,从理论上讲,PTE可以比简单的均值差更强大,同时仍然控制I型错误率。这是使用delta方法实现的,正如在成熟的计算工具中实现的那样,如R包‘msm’和SAS过程‘NLMIXED’。通过分析III期试验的数据,我们说明了PTE如何将不同终点和不同呈现格式的治疗结果联系起来。这些完善的统计工具可用于估计比例治疗效果,结合这一理论论证,为具有连续终点的临床试验提供了另一种检验统计。
{"title":"The proportional treatment effect: A metric that empowers and connects.","authors":"Guoqiao Wang, Yijie Liao, Caiyan Li, Kun Jin, Yan Li, Gary Cutter","doi":"10.1080/10543406.2025.2602479","DOIUrl":"https://doi.org/10.1080/10543406.2025.2602479","url":null,"abstract":"<p><p>Clinical trials with continuous endpoints evaluate efficacy by comparing the difference in mean changes from baseline between groups. However, clinicians often interpret results in terms of a proportional reduction rather than an absolute difference. An alternative approach is to reparametrize this difference as a proportional treatment effect (PTE), calculated by dividing the difference by the placebo mean change. PTE is not a new metric per se, but a specific reparameterization gaining traction in certain clinical contexts. We demonstrate that, in theory, PTE can be more powerful than the simple difference in means while still controlling the type I error rate. This is achieved using the delta method, as implemented in well-established computational tools like the R package 'msm' and the SAS procedure 'NLMIXED'. By analyzing data from phase III trials, we illustrate how a PTE connects treatment outcomes across various endpoints and different presentation formats. The availability of these well-established statistical tools for estimating proportional treatment effects, combined with this theoretical demonstration, suggests an alternative test statistic for clinical trials with continuous endpoints.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-9"},"PeriodicalIF":1.2,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Saddlepoint p-values for the class of bivariate two-sample tests under generalized randomized block design. 广义随机区组设计下二元双样本检验类的鞍点p值。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-05 DOI: 10.1080/10543406.2025.2592617
Abd El-Raheem M Abd El-Raheem, Ibrahim A A Shanan, Mona Hosny

Bivariate data arise in several research fields, such as clinical trials and reliability studies. In clinical trials, patients are distributed into treatment groups using randomization designs, which prevent selection bias and incidental bias. Generalized randomized block design is among clinical trials' most famous and widely used randomization designs. This article investigates the use of the saddlepoint method to approximate the underlying permutation distributions of bivariate two-sample tests under a generalized randomized block design. Additionally, the saddlepoint method is utilized to approximate the tail probability of these tests. Through comprehensive simulation studies, the accuracy of this approximation method is thoroughly evaluated, revealing a significant improvement in precision compared to the asymptotic normal approximation.

双变量数据出现在一些研究领域,如临床试验和可靠性研究。在临床试验中,使用随机化设计将患者分配到治疗组,以防止选择偏倚和偶然偏倚。广义随机区组设计是临床试验中最著名、应用最广泛的随机化设计之一。本文研究了在广义随机块设计下使用鞍点法近似二元双样本检验的潜在排列分布。此外,利用鞍点法来近似这些试验的尾概率。通过全面的仿真研究,对该近似方法的精度进行了全面评估,与渐近正态近似相比,精度有了显著提高。
{"title":"Saddlepoint p-values for the class of bivariate two-sample tests under generalized randomized block design.","authors":"Abd El-Raheem M Abd El-Raheem, Ibrahim A A Shanan, Mona Hosny","doi":"10.1080/10543406.2025.2592617","DOIUrl":"https://doi.org/10.1080/10543406.2025.2592617","url":null,"abstract":"<p><p>Bivariate data arise in several research fields, such as clinical trials and reliability studies. In clinical trials, patients are distributed into treatment groups using randomization designs, which prevent selection bias and incidental bias. Generalized randomized block design is among clinical trials' most famous and widely used randomization designs. This article investigates the use of the saddlepoint method to approximate the underlying permutation distributions of bivariate two-sample tests under a generalized randomized block design. Additionally, the saddlepoint method is utilized to approximate the tail probability of these tests. Through comprehensive simulation studies, the accuracy of this approximation method is thoroughly evaluated, revealing a significant improvement in precision compared to the asymptotic normal approximation.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-12"},"PeriodicalIF":1.2,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145679535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
TITE-STEIN: Time-to-event simple toxicity and efficacy interval design to accelerate phase I/II trials. TITE-STEIN:时间到事件的简单毒性和疗效间隔设计,加速I/II期试验。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-04 DOI: 10.1080/10543406.2025.2592615
Hao Sun, Jieqi Tu, Revathi Ananthakrishnan, Eunhee Kim

Oncology dose-finding trials are shifting from identifying the maximum-tolerated dose (MTD) to determining the optimal biological dose (OBD), driven by the need for efficient methods that consider both toxicity and efficacy. This is particularly important for novel therapies, such as immunotherapies and molecularly targeted therapies, which often exhibit non-monotonic dose-efficacy curves. The Simple Toxicity and Efficacy Interval (STEIN) design has demonstrated strong performance in accommodating diverse dose-efficacy patterns and incorporating both toxicity and efficacy outcomes to select the OBD. However, the rapid accrual of patients and the often-delayed onset of toxicity and/or efficacy pose challenges to timely adaptive-dose decisions. To address these challenges, we propose TITE-STEIN, a model-assisted design that incorporates time-to-event (TITE) outcomes for toxicity and/or efficacy, by extending STEIN. In this article, we demonstrate that TITE-STEIN significantly shortens the trial duration compared to STEIN. Furthermore, by integrating an OBD verification procedure during OBD selection, TITE-STEIN effectively mitigates the risk of exposing patients to inadmissible doses when the OBD does not exist. Extensive simulations demonstrate that TITE-STEIN outperforms existing TITE designs, including TITE-BONI12, TITE-BOIN-ET, LO-TC, and Joint TITE-CRM, by selecting the OBD more accurately, allocating more patients to it, and improving overdose control.

肿瘤剂量发现试验正在从确定最大耐受剂量(MTD)转向确定最佳生物剂量(OBD),这是由于需要同时考虑毒性和疗效的有效方法。这对于新疗法尤其重要,例如免疫疗法和分子靶向疗法,它们通常表现出非单调的剂量-功效曲线。简单毒性和疗效区间(STEIN)设计在适应不同剂量-疗效模式和结合毒性和疗效结果来选择OBD方面表现出很强的性能。然而,患者的快速累积和毒性和/或疗效的延迟发作对及时的适应性剂量决定提出了挑战。为了应对这些挑战,我们提出了TITE-STEIN,这是一种模型辅助设计,通过扩展STEIN,将毒性和/或疗效的时间-事件(TITE)结果纳入其中。在本文中,我们证明了与STEIN相比,TITE-STEIN显著缩短了试验持续时间。此外,通过在OBD选择过程中整合OBD验证程序,TITE-STEIN有效地降低了在OBD不存在时患者暴露于不可接受剂量的风险。大量的模拟表明,通过更准确地选择OBD,分配更多的患者,并改善过量控制,TITE- stein优于现有的TITE设计,包括TITE- boni12, TITE- boin - et, LO-TC和Joint TITE- crm。
{"title":"TITE-STEIN: Time-to-event simple toxicity and efficacy interval design to accelerate phase I/II trials.","authors":"Hao Sun, Jieqi Tu, Revathi Ananthakrishnan, Eunhee Kim","doi":"10.1080/10543406.2025.2592615","DOIUrl":"https://doi.org/10.1080/10543406.2025.2592615","url":null,"abstract":"<p><p>Oncology dose-finding trials are shifting from identifying the maximum-tolerated dose (MTD) to determining the optimal biological dose (OBD), driven by the need for efficient methods that consider both toxicity and efficacy. This is particularly important for novel therapies, such as immunotherapies and molecularly targeted therapies, which often exhibit non-monotonic dose-efficacy curves. The Simple Toxicity and Efficacy Interval (STEIN) design has demonstrated strong performance in accommodating diverse dose-efficacy patterns and incorporating both toxicity and efficacy outcomes to select the OBD. However, the rapid accrual of patients and the often-delayed onset of toxicity and/or efficacy pose challenges to timely adaptive-dose decisions. To address these challenges, we propose TITE-STEIN, a model-assisted design that incorporates time-to-event (TITE) outcomes for toxicity and/or efficacy, by extending STEIN. In this article, we demonstrate that TITE-STEIN significantly shortens the trial duration compared to STEIN. Furthermore, by integrating an OBD verification procedure during OBD selection, TITE-STEIN effectively mitigates the risk of exposing patients to inadmissible doses when the OBD does not exist. Extensive simulations demonstrate that TITE-STEIN outperforms existing TITE designs, including TITE-BONI12, TITE-BOIN-ET, LO-TC, and Joint TITE-CRM, by selecting the OBD more accurately, allocating more patients to it, and improving overdose control.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-18"},"PeriodicalIF":1.2,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145670769","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Estimands for long-term follow-up trials in gene therapy products. 对基因治疗产品长期随访试验的估计。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-02 DOI: 10.1080/10543406.2025.2592619
Shihua Wen, Patricia Anderson, Ran Duan, Oleksandr Sverdlov, Alan Y Chiang

Gene and genetically modified cell therapies offer incredible potential but may come with certain risks and unknowns of delayed adverse events due to unique characteristics of these products. Major regulatory agencies all require long-term follow-up (LTFU) trials, which can be as long as 15 years, to monitor potential delayed adverse events and the durability of effectiveness. Various innovative approaches have been proposed to reduce the operational burden of gene and genetically modified cell therapy LTFU trials. In this article, the authors aim to apply the ICH E9(R1) estimand framework in the context of such LTFU trials, which can be beneficial during the protocol development stage. A hypothetical LTFU study is used to illustrate the estimand considerations discussed in the current manuscript. Proper estimand specifications for a LTFU study of a gene or genetically modified cell therapy can help add clarity to the study design, data collection, and statistical analysis plan, and help ensure the study is robust, transparent, and capable of addressing the important research questions posed by these advanced therapies.

基因和转基因细胞疗法提供了令人难以置信的潜力,但由于这些产品的独特特性,可能会带来一定的风险和未知的延迟不良事件。主要监管机构都要求进行长达15年的长期随访(LTFU)试验,以监测潜在的延迟不良事件和有效性的持久性。已经提出了各种创新方法来减少基因和转基因细胞治疗LTFU试验的操作负担。在本文中,作者旨在将ICH E9(R1)估计框架应用于此类LTFU试验的背景下,这在协议开发阶段可能是有益的。一个假设的LTFU研究被用来说明当前手稿中讨论的估计和考虑因素。对基因或基因修饰细胞疗法的LTFU研究进行适当的评估和规范可以帮助增加研究设计、数据收集和统计分析计划的清晰度,并有助于确保研究是稳健、透明的,能够解决这些先进疗法提出的重要研究问题。
{"title":"Estimands for long-term follow-up trials in gene therapy products.","authors":"Shihua Wen, Patricia Anderson, Ran Duan, Oleksandr Sverdlov, Alan Y Chiang","doi":"10.1080/10543406.2025.2592619","DOIUrl":"https://doi.org/10.1080/10543406.2025.2592619","url":null,"abstract":"<p><p>Gene and genetically modified cell therapies offer incredible potential but may come with certain risks and unknowns of delayed adverse events due to unique characteristics of these products. Major regulatory agencies all require long-term follow-up (LTFU) trials, which can be as long as 15 years, to monitor potential delayed adverse events and the durability of effectiveness. Various innovative approaches have been proposed to reduce the operational burden of gene and genetically modified cell therapy LTFU trials. In this article, the authors aim to apply the ICH E9(R1) estimand framework in the context of such LTFU trials, which can be beneficial during the protocol development stage. A hypothetical LTFU study is used to illustrate the estimand considerations discussed in the current manuscript. Proper estimand specifications for a LTFU study of a gene or genetically modified cell therapy can help add clarity to the study design, data collection, and statistical analysis plan, and help ensure the study is robust, transparent, and capable of addressing the important research questions posed by these advanced therapies.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-12"},"PeriodicalIF":1.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145656387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
M-DODII: Bayesian dose optimization design for randomized phase II study with multiple indications. M-DODII:多适应症随机II期研究的贝叶斯剂量优化设计
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-27 DOI: 10.1080/10543406.2025.2589731
Sasha Amdur Kravets, Ziji Yu, Rachael Liu, Jianchang Lin

The landscape of oncology drug development is transitioning from traditional cytotoxic chemotherapy drugs to novel agents, such as molecularly targeted therapies (MTA) or immunotherapies. Conventional dose optimization methods based on chemotherapy that assume a monotone dose-response relationship might not be ideal for the development of these novel therapies. Recognizing these limitations, the US FDA has introduced Project Optimus, an initiative aimed to reform the current paradigm of dose optimization. In addition to dose optimization, another critical objective for early phase proof-of-concept clinical trials is indication selection. However, there are limited methodologies that can address dose optimization and indication selection simultaneously. In this paper, we propose a Bayesian Dose Optimization Design for Randomized Phase II trials with Multiple Indications (M-DODII) that integrates Bayesian continuous monitoring and Bayesian pick-the-winner approach, utilizing efficacy and toxicity endpoints to inform dose selection for multiple indications simultaneously. Through simulation studies, we demonstrate that M-DODII has favorable operating characteristics with controlled selection error. Compared to other adaptive designs, M-DODII shows a lower probability of choosing a suboptimal dose, a higher probability of selecting the optimal dose, and reduced total sample size.

肿瘤药物开发的前景正在从传统的细胞毒性化疗药物过渡到新型药物,如分子靶向治疗(MTA)或免疫治疗。传统的基于化疗的剂量优化方法假设剂量-反应关系单调,可能不适合这些新疗法的发展。认识到这些局限性,美国FDA推出了Optimus项目,这是一项旨在改革当前剂量优化范例的倡议。除了剂量优化外,早期概念验证临床试验的另一个关键目标是适应症选择。然而,能够同时解决剂量优化和适应症选择的方法有限。在本文中,我们提出了一种多适应症随机II期试验(M-DODII)的贝叶斯剂量优化设计,该设计集成了贝叶斯连续监测和贝叶斯选择赢家方法,利用疗效和毒性终点同时为多种适应症的剂量选择提供信息。通过仿真研究,我们证明了M-DODII具有良好的操作特性,选择误差可控。与其他自适应设计相比,M-DODII选择次优剂量的概率较低,选择最佳剂量的概率较高,并且总样本量减小。
{"title":"M-DODII: Bayesian dose optimization design for randomized phase II study with multiple indications.","authors":"Sasha Amdur Kravets, Ziji Yu, Rachael Liu, Jianchang Lin","doi":"10.1080/10543406.2025.2589731","DOIUrl":"https://doi.org/10.1080/10543406.2025.2589731","url":null,"abstract":"<p><p>The landscape of oncology drug development is transitioning from traditional cytotoxic chemotherapy drugs to novel agents, such as molecularly targeted therapies (MTA) or immunotherapies. Conventional dose optimization methods based on chemotherapy that assume a monotone dose-response relationship might not be ideal for the development of these novel therapies. Recognizing these limitations, the US FDA has introduced Project Optimus, an initiative aimed to reform the current paradigm of dose optimization. In addition to dose optimization, another critical objective for early phase proof-of-concept clinical trials is indication selection. However, there are limited methodologies that can address dose optimization and indication selection simultaneously. In this paper, we propose a Bayesian Dose Optimization Design for Randomized Phase II trials with Multiple Indications (M-DODII) that integrates Bayesian continuous monitoring and Bayesian pick-the-winner approach, utilizing efficacy and toxicity endpoints to inform dose selection for multiple indications simultaneously. Through simulation studies, we demonstrate that M-DODII has favorable operating characteristics with controlled selection error. Compared to other adaptive designs, M-DODII shows a lower probability of choosing a suboptimal dose, a higher probability of selecting the optimal dose, and reduced total sample size.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-14"},"PeriodicalIF":1.2,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145642993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Enhancing dose selection in phase I cancer trials: Extending the Bayesian Logistic Regression Model with non-DLT adverse events integration. 加强I期癌症试验的剂量选择:扩展非dlt不良事件整合的贝叶斯逻辑回归模型。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-24 DOI: 10.1080/10543406.2025.2589734
Andrea Nizzardo, Luca Genetti, Marco Pergher

This work introduces the Burdened Bayesian Logistic Regression Model (BBLRM), an enhancement of the Bayesian Logistic Regression Model (BLRM) for dose-finding in phase I oncology trials. The BLRM determines the maximum tolerated dose (MTD) based on dose-limiting toxicities (DLTs). However, clinicians often perceive model-based designs like BLRM as complex and less conservative than rule-based designs, such as the widely used 3 + 3 method. To address these concerns, BBLRM incorporates non-DLT adverse events (nDLTAEs), which, although not severe enough to be DLTs, indicate potential toxicity risks at higher doses. BBLRM introduces an additional parameter δ to account for nDLTAEs, adjusting toxicity probability estimates to make dose escalation more conservative while maintaining accurate MTD allocation. This parameter, generated basing on the proportion of patients experiencing nDLTAEs, is tuned to balance conservatism with model performance, reducing the risk of selecting overly toxic doses. Additionally, involving clinicians in identifying nDLTAEs enhances their engagement in the dose-finding process. A simulation study compares BBLRM with two other BLRM methods and a two-stage Continual Reassessment Method (CRM) incorporating nDLTAEs. Results show that BBLRM reduces the proportion of toxic doses selected as MTD without compromising the accuracy in MTD identification. These findings suggest that integrating nDLTAEs can improve the safety and acceptance of model-based designs in phase I oncology trials.

这项工作介绍了负担贝叶斯逻辑回归模型(BBLRM),这是贝叶斯逻辑回归模型(BLRM)的增强,用于I期肿瘤试验的剂量发现。BLRM根据剂量限制毒性(dlt)确定最大耐受剂量(MTD)。然而,临床医生通常认为基于模型的设计(如BLRM)比基于规则的设计(如广泛使用的3 + 3方法)更复杂,更不保守。为了解决这些问题,BBLRM纳入了非dlt不良事件(nDLTAEs),这些不良事件虽然没有严重到足以成为dlt,但表明在高剂量下存在潜在的毒性风险。BBLRM引入了一个额外的参数δ来解释nDLTAEs,调整毒性概率估计,使剂量递增更加保守,同时保持准确的MTD分配。该参数是根据经历nDLTAEs的患者比例生成的,经过调整以平衡保守性和模型性能,降低了选择过度毒性剂量的风险。此外,让临床医生参与确定ndltae可以提高他们在剂量确定过程中的参与度。仿真研究比较了BBLRM与其他两种BLRM方法以及包含nDLTAEs的两阶段持续再评估方法(CRM)。结果表明,在不影响MTD鉴定准确性的前提下,BBLRM降低了MTD毒性剂量的选择比例。这些发现表明,整合nDLTAEs可以提高I期肿瘤试验中基于模型设计的安全性和接受度。
{"title":"Enhancing dose selection in phase I cancer trials: Extending the Bayesian Logistic Regression Model with non-DLT adverse events integration.","authors":"Andrea Nizzardo, Luca Genetti, Marco Pergher","doi":"10.1080/10543406.2025.2589734","DOIUrl":"https://doi.org/10.1080/10543406.2025.2589734","url":null,"abstract":"<p><p>This work introduces the Burdened Bayesian Logistic Regression Model (BBLRM), an enhancement of the Bayesian Logistic Regression Model (BLRM) for dose-finding in phase I oncology trials. The BLRM determines the maximum tolerated dose (MTD) based on dose-limiting toxicities (DLTs). However, clinicians often perceive model-based designs like BLRM as complex and less conservative than rule-based designs, such as the widely used 3 + 3 method. To address these concerns, BBLRM incorporates non-DLT adverse events (nDLTAEs), which, although not severe enough to be DLTs, indicate potential toxicity risks at higher doses. BBLRM introduces an additional parameter δ to account for nDLTAEs, adjusting toxicity probability estimates to make dose escalation more conservative while maintaining accurate MTD allocation. This parameter, generated basing on the proportion of patients experiencing nDLTAEs, is tuned to balance conservatism with model performance, reducing the risk of selecting overly toxic doses. Additionally, involving clinicians in identifying nDLTAEs enhances their engagement in the dose-finding process. A simulation study compares BBLRM with two other BLRM methods and a two-stage Continual Reassessment Method (CRM) incorporating nDLTAEs. Results show that BBLRM reduces the proportion of toxic doses selected as MTD without compromising the accuracy in MTD identification. These findings suggest that integrating nDLTAEs can improve the safety and acceptance of model-based designs in phase I oncology trials.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-21"},"PeriodicalIF":1.2,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145589649","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Recovery of overall survival information from treatment switching in oncology trials using multiple imputation. 在肿瘤试验中使用多重输入从治疗转换中恢复总体生存信息。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1080/10543406.2025.2571224
Jianbo Xu

In oncology trials, patients in both the control and experimental arms can receive different subsequent anti-cancer therapies (SATs) after discontinuing their randomized study drugs, a phenomenon commonly referred to as treatment switching. SATs may have the potential to extend overall survival (OS) in patients treated with the control and experimental drugs. Without recovering the information from the SATs, the statistical power of the clinical trials could be drastically reduced, thus making it difficult or impossible to meet the efficacy objective. This article presents a novel statistical method for imputing the post-switching survival time multiple times to derive the point estimate of the true hazard ratio (HR) of OS between the experimental and control drugs and the associated 95% confidence interval (CI). The proposed method provides an effective solution for recovering lost information in the OS caused by SATs. It also offers an efficient way to evaluate the true causal treatment effect, potentially increasing the statistical power. Additionally, this method can be used for patients with a crossover from a placebo to an experimental treatment in placebo-controlled trials. Simulation studies demonstrated that the proposed method performed well and reliably, and applications to oncology trials using the simulated data are provided.

在肿瘤学试验中,对照组和实验组的患者在停止随机研究药物后都可以接受不同的后续抗癌治疗(SATs),这种现象通常被称为治疗转换。SATs有可能延长接受对照和实验药物治疗的患者的总生存期(OS)。如果不能从sat中恢复信息,临床试验的统计效力可能会大大降低,从而难以或不可能达到疗效目标。本文提出了一种新的统计方法,将切换后生存时间多次输入,以导出实验药物与对照药物之间OS的真实风险比(HR)和相关的95%置信区间(CI)的点估计。本文提出的方法为恢复由sat引起的操作系统中丢失的信息提供了有效的解决方案。它还提供了一种有效的方法来评估真正的因果处理效果,潜在地提高了统计能力。此外,该方法可用于安慰剂对照试验中从安慰剂到实验性治疗交叉的患者。仿真研究表明,该方法性能良好、可靠,并将模拟数据应用于肿瘤临床试验。
{"title":"Recovery of overall survival information from treatment switching in oncology trials using multiple imputation.","authors":"Jianbo Xu","doi":"10.1080/10543406.2025.2571224","DOIUrl":"https://doi.org/10.1080/10543406.2025.2571224","url":null,"abstract":"<p><p>In oncology trials, patients in both the control and experimental arms can receive different subsequent anti-cancer therapies (SATs) after discontinuing their randomized study drugs, a phenomenon commonly referred to as treatment switching. SATs may have the potential to extend overall survival (OS) in patients treated with the control and experimental drugs. Without recovering the information from the SATs, the statistical power of the clinical trials could be drastically reduced, thus making it difficult or impossible to meet the efficacy objective. This article presents a novel statistical method for imputing the post-switching survival time multiple times to derive the point estimate of the true hazard ratio (HR) of OS between the experimental and control drugs and the associated 95% confidence interval (CI). The proposed method provides an effective solution for recovering lost information in the OS caused by SATs. It also offers an efficient way to evaluate the true causal treatment effect, potentially increasing the statistical power. Additionally, this method can be used for patients with a crossover from a placebo to an experimental treatment in placebo-controlled trials. Simulation studies demonstrated that the proposed method performed well and reliably, and applications to oncology trials using the simulated data are provided.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-26"},"PeriodicalIF":1.2,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566435","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Statistical approaches to evaluate the positive control drug using the hERG assay. 使用hERG测定法评价阳性对照药物的统计方法。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-19 DOI: 10.1080/10543406.2025.2575939
Yu-Ting Weng, Dalong Huang

The assessment of human ether-a-go-go-related gene (hERG) safety assay is essential for estimating the risk that a drug will cause delayed repolarization and QT interval prolongation prior to human administration. Quantitative assessment of hERG safety assay similarity presents significant challenges due to the absence of consensus methodology and substantial inter-laboratory variability in hERG assay performance. We developed a statistical framework to conduct quantitative assessment of hERG safety assay similarity for drug products between sponsor's laboratories and laboratories that follow the ICH E14 S7b Q&A Best Practice recommended protocol. Our approach employs fixed margin equivalence testing methodology. Using real-world and/or simulated data, we demonstrate that the proposed equivalence testing methods successfully identify similar hERG assays between laboratories for 28 Comprehensive In Vitro Proarrhythmia Assay (CiPA) drugs. The testing results align with the domain experts' assessments, validating the framework's utility for regulatory decision-making.

评估人乙醚-a-go-go相关基因(hERG)安全性测定对于估计药物在给药前引起复极延迟和QT间期延长的风险至关重要。由于缺乏一致的方法和hERG分析性能的大量实验室间差异,hERG安全分析相似性的定量评估提出了重大挑战。我们开发了一个统计框架,用于在申办者实验室和遵循ICH E14 S7b问答最佳实践推荐方案的实验室之间对药品hERG安全性测定相似性进行定量评估。我们的方法采用固定边际等效检验方法。使用真实世界和/或模拟数据,我们证明了所提出的等效性测试方法成功地识别了28种综合体外心律失常原测定(CiPA)药物在实验室之间的相似hERG测定。测试结果与领域专家的评估一致,验证了框架对监管决策的效用。
{"title":"Statistical approaches to evaluate the positive control drug using the hERG assay.","authors":"Yu-Ting Weng, Dalong Huang","doi":"10.1080/10543406.2025.2575939","DOIUrl":"https://doi.org/10.1080/10543406.2025.2575939","url":null,"abstract":"<p><p>The assessment of human ether-a-go-go-related gene (hERG) safety assay is essential for estimating the risk that a drug will cause delayed repolarization and QT interval prolongation prior to human administration. Quantitative assessment of hERG safety assay similarity presents significant challenges due to the absence of consensus methodology and substantial inter-laboratory variability in hERG assay performance. We developed a statistical framework to conduct quantitative assessment of hERG safety assay similarity for drug products between sponsor's laboratories and laboratories that follow the ICH E14 S7b Q&A Best Practice recommended protocol. Our approach employs fixed margin equivalence testing methodology. Using real-world and/or simulated data, we demonstrate that the proposed equivalence testing methods successfully identify similar hERG assays between laboratories for 28 Comprehensive <i>In Vitro</i> Proarrhythmia Assay (CiPA) drugs. The testing results align with the domain experts' assessments, validating the framework's utility for regulatory decision-making.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-17"},"PeriodicalIF":1.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145551786","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Blinded sample size re-estimation in a crossover study. 交叉研究中的盲法样本量重新估计。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-18 DOI: 10.1080/10543406.2025.2575947
Shaofei Zhao, Balakrishna Hosmane, Chen Chen, Yi-Lin Chiu

Bioequivalence studies play a pivotal role in drug development by establishing the clinical equivalence of two drug formulations. These studies often utilize crossover designs to facilitate within-subject treatment comparisons, optimizing statistical power with fewer subjects. However, uncertainty regarding the variance of a new drug or formulation during planning presents a challenge for sample size determination. While adaptive designs offer a potential solution, their application in crossover studies is less explored compared to group sequential designs, and many existing adaptive methods require data unblinding during the trial. Only two blinded sample size re-estimation approaches have been developed in crossover settings to date. In this paper, we propose a novel method for blinded within-subject variance estimation at interim analysis and re-estimate the sample size to achieve the desired power. We thoroughly investigate its analytical properties and introduce a refined, unbiased estimator. Through extensive simulation studies, our method shows comparable performance to existing blinded approaches and offers a distinct advantage in scenarios with small treatment differences and large subject variances.

生物等效性研究通过确定两种药物制剂的临床等效性,在药物开发中起着关键作用。这些研究通常采用交叉设计来促进受试者内的治疗比较,在较少受试者的情况下优化统计能力。然而,在计划过程中,关于新药或配方差异的不确定性对样本量的确定提出了挑战。虽然自适应设计提供了一种潜在的解决方案,但与组序贯设计相比,其在交叉研究中的应用探索较少,而且许多现有的自适应方法需要在试验期间对数据进行解盲。迄今为止,在交叉设置中只有两种盲法样本量重新估计方法被开发出来。在本文中,我们提出了一种在中期分析中盲法估计受试者内方差的新方法,并重新估计样本量以达到期望的功率。我们深入研究了它的解析性质,并引入了一个改进的无偏估计量。通过广泛的模拟研究,我们的方法显示出与现有盲法相当的性能,并在治疗差异小、受试者差异大的情况下提供了明显的优势。
{"title":"Blinded sample size re-estimation in a crossover study.","authors":"Shaofei Zhao, Balakrishna Hosmane, Chen Chen, Yi-Lin Chiu","doi":"10.1080/10543406.2025.2575947","DOIUrl":"https://doi.org/10.1080/10543406.2025.2575947","url":null,"abstract":"<p><p>Bioequivalence studies play a pivotal role in drug development by establishing the clinical equivalence of two drug formulations. These studies often utilize crossover designs to facilitate within-subject treatment comparisons, optimizing statistical power with fewer subjects. However, uncertainty regarding the variance of a new drug or formulation during planning presents a challenge for sample size determination. While adaptive designs offer a potential solution, their application in crossover studies is less explored compared to group sequential designs, and many existing adaptive methods require data unblinding during the trial. Only two blinded sample size re-estimation approaches have been developed in crossover settings to date. In this paper, we propose a novel method for blinded within-subject variance estimation at interim analysis and re-estimate the sample size to achieve the desired power. We thoroughly investigate its analytical properties and introduce a refined, unbiased estimator. Through extensive simulation studies, our method shows comparable performance to existing blinded approaches and offers a distinct advantage in scenarios with small treatment differences and large subject variances.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-19"},"PeriodicalIF":1.2,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145551806","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
A Bayesian design for dual-agent dose optimization with targeted therapies. 靶向治疗双药剂量优化的贝叶斯设计。
IF 1.2 4区 医学 Q4 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-16 DOI: 10.1080/10543406.2025.2557533
José L Jiménez, Mourad Tighiouart

In this article, we propose a phase I-II design in two stages for the combination of molecularly targeted therapies. The design is motivated by a published case study that combines MEK and PIK3CA inhibitors; a setting in which higher dose levels do not necessarily translate into higher efficacy responses. The goal is therefore to identify dose combination(s) with a prespecified desirable risk-benefit trade-off. We propose a flexible cubic spline to model the marginal distribution of the efficacy response. In stage I, patients are allocated following the escalation with overdose control (EWOC) principle whereas, in stage II, we adaptively randomize patients to the available experimental dose combinations based on the continuously updated model parameters. A simulation study is presented to assess the design's performance under different scenarios, as well as to evaluate its sensitivity to the sample size and to model misspecification. Compared to a recently published dose finding algorithm for biologic drugs, our design is safer and more efficient at identifying optimal dose combinations.

在本文中,我们提出了一个分两个阶段的I-II期设计,用于结合分子靶向治疗。该设计的动机来自于一项已发表的案例研究,该研究结合了MEK和PIK3CA抑制剂;较高剂量水平不一定转化为较高疗效反应的环境。因此,目标是确定具有预先规定的理想风险-效益权衡的剂量组合。我们提出了一个灵活的三次样条来模拟功效响应的边际分布。在第一阶段,患者按照递增与过量控制(EWOC)原则进行分配,而在第二阶段,我们根据不断更新的模型参数自适应地将患者随机分配到可用的实验剂量组合中。通过仿真研究来评估该设计在不同情况下的性能,以及评估其对样本量和模型错配的敏感性。与最近发表的生物药物剂量查找算法相比,我们的设计在确定最佳剂量组合方面更安全,更有效。
{"title":"A Bayesian design for dual-agent dose optimization with targeted therapies.","authors":"José L Jiménez, Mourad Tighiouart","doi":"10.1080/10543406.2025.2557533","DOIUrl":"https://doi.org/10.1080/10543406.2025.2557533","url":null,"abstract":"<p><p>In this article, we propose a phase I-II design in two stages for the combination of molecularly targeted therapies. The design is motivated by a published case study that combines MEK and PIK3CA inhibitors; a setting in which higher dose levels do not necessarily translate into higher efficacy responses. The goal is therefore to identify dose combination(s) with a prespecified desirable risk-benefit trade-off. We propose a flexible cubic spline to model the marginal distribution of the efficacy response. In stage I, patients are allocated following the escalation with overdose control (EWOC) principle whereas, in stage II, we adaptively randomize patients to the available experimental dose combinations based on the continuously updated model parameters. A simulation study is presented to assess the design's performance under different scenarios, as well as to evaluate its sensitivity to the sample size and to model misspecification. Compared to a recently published dose finding algorithm for biologic drugs, our design is safer and more efficient at identifying optimal dose combinations.</p>","PeriodicalId":54870,"journal":{"name":"Journal of Biopharmaceutical Statistics","volume":" ","pages":"1-16"},"PeriodicalIF":1.2,"publicationDate":"2025-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145535204","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
期刊
Journal of Biopharmaceutical Statistics
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