首页 > 最新文献

Clinical Trials最新文献

英文 中文
Optimizing accrual to a large-scale, clinically integrated randomized trial in anesthesiology: A 2-year analysis of recruitment. 优化麻醉学大规模临床综合随机试验的招募:为期两年的招募分析。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-19 DOI: 10.1177/17407745241255087
Hanae K Tokita, Melissa Assel, Joanna Serafin, Emily Lin, Leslie Sarraf, Geema Masson, Tracy-Ann Moo, Jonas A Nelson, Brett A Simon, Andrew J Vickers

Background: Performing large randomized trials in anesthesiology is often challenging and costly. The clinically integrated randomized trial is characterized by simplified logistics embedded into routine clinical practice, enabling ease and efficiency of recruitment, offering an opportunity for clinicians to conduct large, high-quality randomized trials under low cost. Our aims were to (1) demonstrate the feasibility of the clinically integrated trial design in a high-volume anesthesiology practice and (2) assess whether trial quality improvement interventions led to more balanced accrual among study arms and improved trial compliance over time.

Methods: This is an interim analysis of recruitment to a cluster-randomized trial investigating three nerve block approaches for mastectomy with immediate implant-based reconstruction: paravertebral block (arm 1), paravertebral plus interpectoral plane blocks (arm 2), and serratus anterior plane plus interpectoral plane blocks (arm 3). We monitored accrual and consent rates, clinician compliance with the randomized treatment, and availability of outcome data. Assessment after the initial year of implementation showed a slight imbalance in study arms suggesting areas for improvement in trial compliance. Specific improvement interventions included increasing the frequency of communication with the consenting staff and providing direct feedback to clinician investigators about their individual recruitment patterns. We assessed overall accrual rates and tested for differences in accrual, consent, and compliance rates pre- and post-improvement interventions.

Results: Overall recruitment was extremely high, accruing close to 90% of the eligible population. In the pre-intervention period, there was evidence of bias in the proportion of patients being accrued and receiving the monthly block, with higher rates in arm 3 (90%) compared to arms 1 (81%) and 2 (79%, p = 0.021). In contrast, in the post-intervention period, there was no statistically significant difference between groups (p = 0.8). Eligible for randomization rate increased from 89% in the pre-intervention period to 95% in the post-intervention period (difference 5.7%; 95% confidence interval = 2.2%-9.4%, p = 0.002). Consent rate increased from 95% to 98% (difference of 3.7%; 95% confidence interval = 1.1%-6.3%; p = 0.004). Compliance with the randomized nerve block approach was maintained at close to 100% and availability of primary outcome data was 100%.

Conclusion: The clinically integrated randomized trial design enables rapid trial accrual with a high participant compliance rate in a high-volume anesthesiology practice. Continuous monitoring of accrual, consent, and compliance rates is necessary to maintain and improve trial conduct and reduce potential biases. This trial methodology serves as a template for the implementation of other large, low-cost randomized

背景:在麻醉学领域开展大型随机试验通常具有挑战性且成本高昂。临床综合随机试验的特点是将简化的后勤工作嵌入到常规临床实践中,使招募工作变得简单高效,为临床医生以低成本开展大型、高质量的随机试验提供了机会。我们的目的是:(1) 证明临床综合试验设计在大容量麻醉科实践中的可行性;(2) 评估试验质量改进干预措施是否能使各研究臂之间的应征人数更加均衡,并随着时间的推移改善试验的依从性:这是一项集群随机试验的中期招募分析,该试验研究了乳房切除术与即时植入物重建的三种神经阻滞方法:椎旁阻滞(第 1 组)、椎旁加胸骨间平面阻滞(第 2 组)和锯肌前平面加胸骨间平面阻滞(第 3 组)。我们对应征率和同意率、临床医生对随机治疗的依从性以及结果数据的可用性进行了监测。实施最初一年后的评估显示,研究臂略有失衡,这表明试验依从性有待改善。具体的改进措施包括增加与同意人员沟通的频率,并向临床研究人员提供有关其个人招募模式的直接反馈。我们对总体招募率进行了评估,并检测了改进干预前后招募率、同意率和依从率的差异:结果:总体招募率非常高,接近 90% 的合格人群都参与了招募。在干预前,有证据表明患者的入组比例和每月接受治疗的比例存在偏差,与第一组(81%)和第二组(79%,P = 0.021)相比,第三组的比例更高(90%)。相比之下,在干预后阶段,组间差异无统计学意义(p = 0.8)。符合随机化条件的比例从干预前的 89% 提高到干预后的 95%(差异为 5.7%;95% 置信区间 = 2.2%-9.4%,p = 0.002)。同意率从 95% 提高到 98%(差异为 3.7%;95% 置信区间 = 1.1%-6.3%;P = 0.004)。随机神经阻滞方法的依从性保持在接近 100%,主要结果数据的可用性达到 100%:结论:临床综合随机试验设计能在麻醉科高工作量的实践中实现试验的快速累积和较高的参与者依从率。有必要对试验的参与率、同意率和符合率进行持续监控,以保持和改善试验的进行并减少潜在的偏差。该试验方法可作为麻醉科实施其他大型、低成本随机试验的模板。
{"title":"Optimizing accrual to a large-scale, clinically integrated randomized trial in anesthesiology: A 2-year analysis of recruitment.","authors":"Hanae K Tokita, Melissa Assel, Joanna Serafin, Emily Lin, Leslie Sarraf, Geema Masson, Tracy-Ann Moo, Jonas A Nelson, Brett A Simon, Andrew J Vickers","doi":"10.1177/17407745241255087","DOIUrl":"https://doi.org/10.1177/17407745241255087","url":null,"abstract":"<p><strong>Background: </strong>Performing large randomized trials in anesthesiology is often challenging and costly. The clinically integrated randomized trial is characterized by simplified logistics embedded into routine clinical practice, enabling ease and efficiency of recruitment, offering an opportunity for clinicians to conduct large, high-quality randomized trials under low cost. Our aims were to (1) demonstrate the feasibility of the clinically integrated trial design in a high-volume anesthesiology practice and (2) assess whether trial quality improvement interventions led to more balanced accrual among study arms and improved trial compliance over time.</p><p><strong>Methods: </strong>This is an interim analysis of recruitment to a cluster-randomized trial investigating three nerve block approaches for mastectomy with immediate implant-based reconstruction: paravertebral block (arm 1), paravertebral plus interpectoral plane blocks (arm 2), and serratus anterior plane plus interpectoral plane blocks (arm 3). We monitored accrual and consent rates, clinician compliance with the randomized treatment, and availability of outcome data. Assessment after the initial year of implementation showed a slight imbalance in study arms suggesting areas for improvement in trial compliance. Specific improvement interventions included increasing the frequency of communication with the consenting staff and providing direct feedback to clinician investigators about their individual recruitment patterns. We assessed overall accrual rates and tested for differences in accrual, consent, and compliance rates pre- and post-improvement interventions.</p><p><strong>Results: </strong>Overall recruitment was extremely high, accruing close to 90% of the eligible population. In the pre-intervention period, there was evidence of bias in the proportion of patients being accrued and receiving the monthly block, with higher rates in arm 3 (90%) compared to arms 1 (81%) and 2 (79%, p = 0.021). In contrast, in the post-intervention period, there was no statistically significant difference between groups (p = 0.8). Eligible for randomization rate increased from 89% in the pre-intervention period to 95% in the post-intervention period (difference 5.7%; 95% confidence interval = 2.2%-9.4%, p = 0.002). Consent rate increased from 95% to 98% (difference of 3.7%; 95% confidence interval = 1.1%-6.3%; p = 0.004). Compliance with the randomized nerve block approach was maintained at close to 100% and availability of primary outcome data was 100%.</p><p><strong>Conclusion: </strong>The clinically integrated randomized trial design enables rapid trial accrual with a high participant compliance rate in a high-volume anesthesiology practice. Continuous monitoring of accrual, consent, and compliance rates is necessary to maintain and improve trial conduct and reduce potential biases. This trial methodology serves as a template for the implementation of other large, low-cost randomized","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141418251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Scaling and interpreting treatment effects in clinical trials using restricted mean survival time. 使用受限平均生存时间对临床试验中的治疗效果进行缩放和解释。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-13 DOI: 10.1177/17407745241254995
Theodore Karrison, Chen Hu, James Dignam

Background: Restricted mean survival time is the expected duration of survival up to a chosen time of restriction τ. For comparison studies, the difference in restricted mean survival times between two groups provides a summary measure of the treatment effect that is free of assumptions regarding the relative shape of the two survival curves, such as proportional hazards. However, it can be difficult to judge the magnitude of the effect from a comparison of restricted means due to the truncation of observation at time τ.

Methods: In this article, we describe additional ways of expressing the treatment effect based on restricted means that can be helpful in this regard. These include the ratio of restricted means, the ratio of life-years (or time) lost, and the average integrated difference between the survival curves, equal to the difference in restricted means divided by τ. These alternative metrics are straightforward to calculate and provide a means for scaling the effect size as an aid to interpretation. Examples from two randomized, multicenter clinical trials in prostate cancer, NRG/RTOG 0521 and NRG/RTOG 0534, with primary endpoints of overall survival and biochemical/radiological progression-free survival, respectively, are presented to illustrate the ideas.

Results: The four effect measures (restricted mean survival time difference, restricted mean survival time ratio, time lost ratio, and average survival rate difference) were 0.45 years, 1.05, 0.81, and 0.038 for RTOG 0521 and 1.36 years, 1.17, 0.56, and 0.12 for RTOG 0534 with τ = 12 and 11 years, respectively. Thus, for example, the 0.45-year difference in the first trial translates into a 19% reduction in time lost and a 3.8% average absolute difference between the survival curves over the 12-year horizon, a modest effect size, whereas the 1.36-year difference in the second trial corresponds to a 44% reduction in time lost and a 12% absolute survival difference, a rather large effect.

Conclusions: In addition to the difference in restricted mean survival times, these alternative measures can be helpful in determining whether the magnitude of the treatment effect is clinically meaningful.

背景:对于比较研究而言,两组间受限平均生存时间的差异提供了一个治疗效果的概括衡量指标,它不需要对两组生存曲线的相对形状(如比例危险)进行假设。然而,由于在时间τ处的观察被截断,因此很难从受限平均值的比较中判断效果的大小:在本文中,我们介绍了基于受限均值来表示治疗效果的其他方法,这些方法在这方面可能会有所帮助。这些方法包括受限均值之比、损失的生命年(或时间)之比以及生存曲线之间的平均综合差异(等于受限均值之差除以τ)。本文以两项前列腺癌多中心随机临床试验(NRG/RTOG 0521 和 NRG/RTOG 0534)为例,分别以总生存期和无生化/放射进展生存期为主要终点,来说明上述观点:RTOG 0521 的四个效应指标(受限平均生存时间差、受限平均生存时间比、时间损失比和平均生存率差)分别为 0.45 年、1.05 年、0.81 年和 0.038 年,RTOG 0534 的四个效应指标(τ = 12 年和 11 年)分别为 1.36 年、1.17 年、0.56 年和 0.12 年。因此,举例来说,第一项试验中0.45年的差异相当于减少了19%的时间损失,12年生存曲线之间的平均绝对差异为3.8%,效应大小适中;而第二项试验中1.36年的差异相当于减少了44%的时间损失,12%的绝对生存差异,效应相当大:除了限制性平均生存时间的差异外,这些替代指标还有助于确定治疗效果的大小是否具有临床意义。
{"title":"Scaling and interpreting treatment effects in clinical trials using restricted mean survival time.","authors":"Theodore Karrison, Chen Hu, James Dignam","doi":"10.1177/17407745241254995","DOIUrl":"10.1177/17407745241254995","url":null,"abstract":"<p><strong>Background: </strong>Restricted mean survival time is the expected duration of survival up to a chosen time of restriction <math><mrow><mi>τ</mi></mrow></math>. For comparison studies, the difference in restricted mean survival times between two groups provides a summary measure of the treatment effect that is free of assumptions regarding the relative shape of the two survival curves, such as proportional hazards. However, it can be difficult to judge the magnitude of the effect from a comparison of restricted means due to the truncation of observation at time <math><mrow><mi>τ</mi></mrow></math>.</p><p><strong>Methods: </strong>In this article, we describe additional ways of expressing the treatment effect based on restricted means that can be helpful in this regard. These include the ratio of restricted means, the ratio of life-years (or time) lost, and the average integrated difference between the survival curves, equal to the difference in restricted means divided by <math><mrow><mi>τ</mi><mo>.</mo></mrow></math> These alternative metrics are straightforward to calculate and provide a means for scaling the effect size as an aid to interpretation. Examples from two randomized, multicenter clinical trials in prostate cancer, NRG/RTOG 0521 and NRG/RTOG 0534, with primary endpoints of overall survival and biochemical/radiological progression-free survival, respectively, are presented to illustrate the ideas.</p><p><strong>Results: </strong>The four effect measures (restricted mean survival time difference, restricted mean survival time ratio, time lost ratio, and average survival rate difference) were 0.45 years, 1.05, 0.81, and 0.038 for RTOG 0521 and 1.36 years, 1.17, 0.56, and 0.12 for RTOG 0534 with <math><mrow><mi>τ</mi></mrow></math> = 12 and 11 years, respectively. Thus, for example, the 0.45-year difference in the first trial translates into a 19% reduction in time lost and a 3.8% average absolute difference between the survival curves over the 12-year horizon, a modest effect size, whereas the 1.36-year difference in the second trial corresponds to a 44% reduction in time lost and a 12% absolute survival difference, a rather large effect.</p><p><strong>Conclusions: </strong>In addition to the difference in restricted mean survival times, these alternative measures can be helpful in determining whether the magnitude of the treatment effect is clinically meaningful.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Adaptive Bayesian information borrowing methods for finding and optimizing subgroup-specific doses. 自适应贝叶斯信息借用法,用于寻找和优化亚组特异性剂量。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2024-01-19 DOI: 10.1177/17407745231212193
Jingyi Zhang, Ruitao Lin, Xin Chen, Fangrong Yan

In precision oncology, integrating multiple cancer patient subgroups into a single master protocol allows for the simultaneous assessment of treatment effects in these subgroups and promotes the sharing of information between them, ultimately reducing sample sizes and costs and enhancing scientific validity. However, the safety and efficacy of these therapies may vary across different subgroups, resulting in heterogeneous outcomes. Therefore, identifying subgroup-specific optimal doses in early-phase clinical trials is crucial for the development of future trials. In this article, we review various innovative Bayesian information-borrowing strategies that aim to determine and optimize subgroup-specific doses. Specifically, we discuss Bayesian hierarchical modeling, Bayesian clustering, Bayesian model averaging or selection, pairwise borrowing, and other relevant approaches. By employing these Bayesian information-borrowing methods, investigators can gain a better understanding of the intricate relationships between dose, toxicity, and efficacy in each subgroup. This increased understanding significantly improves the chances of identifying an optimal dose tailored to each specific subgroup. Furthermore, we present several practical recommendations to guide the design of future early-phase oncology trials involving multiple subgroups when using the Bayesian information-borrowing methods.

在精准肿瘤学中,将多个癌症患者亚组整合到一个主方案中,可以同时评估这些亚组的治疗效果,并促进它们之间的信息共享,最终减少样本量和成本,提高科学有效性。然而,这些疗法在不同亚组中的安全性和疗效可能会有所不同,从而导致不同的结果。因此,在早期临床试验中确定针对亚组的最佳剂量对未来试验的发展至关重要。在本文中,我们回顾了旨在确定和优化亚组特异性剂量的各种创新贝叶斯信息借用策略。具体而言,我们讨论了贝叶斯分层建模、贝叶斯聚类、贝叶斯模型平均或选择、配对借用以及其他相关方法。通过采用这些贝叶斯信息借用方法,研究人员可以更好地了解各亚组中剂量、毒性和疗效之间错综复杂的关系。这种理解的加深大大提高了为每个特定亚组确定最佳剂量的机会。此外,我们还提出了几项实用建议,以指导未来使用贝叶斯信息借用方法设计涉及多个亚组的早期肿瘤学试验。
{"title":"Adaptive Bayesian information borrowing methods for finding and optimizing subgroup-specific doses.","authors":"Jingyi Zhang, Ruitao Lin, Xin Chen, Fangrong Yan","doi":"10.1177/17407745231212193","DOIUrl":"10.1177/17407745231212193","url":null,"abstract":"<p><p>In precision oncology, integrating multiple cancer patient subgroups into a single master protocol allows for the simultaneous assessment of treatment effects in these subgroups and promotes the sharing of information between them, ultimately reducing sample sizes and costs and enhancing scientific validity. However, the safety and efficacy of these therapies may vary across different subgroups, resulting in heterogeneous outcomes. Therefore, identifying subgroup-specific optimal doses in early-phase clinical trials is crucial for the development of future trials. In this article, we review various innovative Bayesian information-borrowing strategies that aim to determine and optimize subgroup-specific doses. Specifically, we discuss Bayesian hierarchical modeling, Bayesian clustering, Bayesian model averaging or selection, pairwise borrowing, and other relevant approaches. By employing these Bayesian information-borrowing methods, investigators can gain a better understanding of the intricate relationships between dose, toxicity, and efficacy in each subgroup. This increased understanding significantly improves the chances of identifying an optimal dose tailored to each specific subgroup. Furthermore, we present several practical recommendations to guide the design of future early-phase oncology trials involving multiple subgroups when using the Bayesian information-borrowing methods.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11132956/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139502337","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 pilot recruitment strategy to enhance ethical and equitable access to Covid-19 pediatric vaccine trials. 一项试点招募战略,旨在提高 Covid-19 儿科疫苗试验的道德性和公平性。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2023-12-23 DOI: 10.1177/17407745231217299
William J Muller, Ravi Jhaveri, Taylor Heald-Sargent, Michelle L Macy, Nia Heard-Garris, Seema Shah, Erin Paquette

Background/aims: The SARS-CoV-2 pandemic disproportionately impacted communities with lower access to health care in the United States, particularly before vaccines were widely available. These same communities are often underrepresented in clinical trials. Efforts to ensure equitable enrollment of participants in trials related to treatment and prevention of Covid-19 can raise concerns about exploitation if communities with lower access to health care are targeted for recruitment.

Methods: To enhance equity while avoiding exploitation, our site developed and implemented a three-part recruitment strategy for pediatric Covid-19 vaccine studies. First, we publicized a registry for potentially interested participants. Next, we applied public health community and social vulnerability indices to categorize the residence of families who had signed up for the registry into three levels to reflect the relative impact of the pandemic on their community: high, medium, and low. Finally, we preferentially offered study participation to interested families living in areas categorized by these indices as having high impact of the Covid-19 pandemic on their community.

Results: This approach allowed us to meet goals for study recruitment based on public health metrics related to disease burden, which contributed to a racially diverse study population that mirrored the surrounding community demographics. While this three-part recruitment strategy improved representation of minoritized groups from areas heavily impacted by the Covid-19 pandemic, important limitations were identified that would benefit from further study.

Conclusion: Future use of this approach to enhance equitable access to research while avoiding exploitation should test different methods to build trust and communicate with underserved communities more effectively.

背景/目的:在美国,SARS-CoV-2 大流行对获得医疗保健机会较少的社区造成了极大的影响,尤其是在疫苗普及之前。这些社区在临床试验中的代表性往往不足。如果招募的对象是医疗条件较差的社区,那么为确保与治疗和预防 Covid-19 相关的试验参与者的公平招募所做的努力可能会引发对剥削的担忧:为了在避免剥削的同时提高公平性,我们的研究机构为儿科 Covid-19 疫苗研究制定并实施了由三部分组成的招募策略。首先,我们对可能感兴趣的参与者进行登记。接下来,我们运用公共卫生社区和社会脆弱性指数将报名参加登记的家庭的居住地分为三个等级,以反映大流行病对其社区的相对影响:高、中、低。最后,我们优先让居住在被这些指数归类为 Covid-19 大流行对其社区影响较大的地区的感兴趣的家庭参与研究:结果:这种方法使我们达到了根据与疾病负担相关的公共卫生指标进行研究招募的目标,从而使研究人群具有种族多样性,与周围社区的人口构成相一致。虽然这种由三部分组成的招募策略提高了受 Covid-19 大流行影响严重地区的少数民族群体的代表性,但也发现了一些重要的局限性,这些局限性将受益于进一步的研究:结论:未来使用这种方法来提高公平参与研究的机会,同时避免剥削,应该测试不同的方法来建立信任,并与服务不足的社区进行更有效的沟通。
{"title":"A pilot recruitment strategy to enhance ethical and equitable access to Covid-19 pediatric vaccine trials.","authors":"William J Muller, Ravi Jhaveri, Taylor Heald-Sargent, Michelle L Macy, Nia Heard-Garris, Seema Shah, Erin Paquette","doi":"10.1177/17407745231217299","DOIUrl":"10.1177/17407745231217299","url":null,"abstract":"<p><strong>Background/aims: </strong>The SARS-CoV-2 pandemic disproportionately impacted communities with lower access to health care in the United States, particularly before vaccines were widely available. These same communities are often underrepresented in clinical trials. Efforts to ensure equitable enrollment of participants in trials related to treatment and prevention of Covid-19 can raise concerns about exploitation if communities with lower access to health care are targeted for recruitment.</p><p><strong>Methods: </strong>To enhance equity while avoiding exploitation, our site developed and implemented a three-part recruitment strategy for pediatric Covid-19 vaccine studies. First, we publicized a registry for potentially interested participants. Next, we applied public health community and social vulnerability indices to categorize the residence of families who had signed up for the registry into three levels to reflect the relative impact of the pandemic on their community: high, medium, and low. Finally, we preferentially offered study participation to interested families living in areas categorized by these indices as having high impact of the Covid-19 pandemic on their community.</p><p><strong>Results: </strong>This approach allowed us to meet goals for study recruitment based on public health metrics related to disease burden, which contributed to a racially diverse study population that mirrored the surrounding community demographics. While this three-part recruitment strategy improved representation of minoritized groups from areas heavily impacted by the Covid-19 pandemic, important limitations were identified that would benefit from further study.</p><p><strong>Conclusion: </strong>Future use of this approach to enhance equitable access to research while avoiding exploitation should test different methods to build trust and communicate with underserved communities more effectively.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138884665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Optimizing the doses of cancer drugs after usual dose finding. 在找到常规剂量后优化抗癌药物的剂量。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2023-12-27 DOI: 10.1177/17407745231213882
Garth W Strohbehn, Walter M Stadler, Philip S Boonstra, Mark J Ratain

Since the middle of the 20th century, oncology's dose-finding paradigm has been oriented toward identifying a drug's maximum tolerated dose, which is then carried forward into phase 2 and 3 trials and clinical practice. For most modern precision medicines, however, maximum tolerated dose is far greater than the minimum dose needed to achieve maximal benefit, leading to unnecessary side effects. Regulatory change may decrease maximum tolerated dose's predominance by enforcing dose optimization of new drugs. Dozens of already approved cancer drugs require re-evaluation, however, introducing a new methodologic and ethical challenge in cancer clinical trials. In this article, we assess the history and current landscape of cancer drug dose finding. We provide a set of strategic priorities for postapproval dose optimization trials of the future. We discuss ethical considerations for postapproval dose optimization trial design and review three major design strategies for these unique trials that would both adhere to ethical standards and benefit patients and funders. We close with a discussion of financial and reporting considerations in the realm of dose optimization. Taken together, we provide a comprehensive, bird's eye view of the postapproval dose optimization trial landscape and offer our thoughts on the next steps required of methodologies and regulatory and funding regimes.

自 20 世纪中叶以来,肿瘤学的剂量研究范式一直以确定药物的最大耐受剂量为导向,然后将其应用于 2、3 期试验和临床实践。然而,对于大多数现代精准药物来说,最大耐受剂量远大于实现最大疗效所需的最小剂量,从而导致不必要的副作用。监管变革可通过强制新药剂量优化来降低最大耐受剂量的主导地位。然而,数十种已获批准的抗癌药物需要重新评估,这给癌症临床试验带来了新的方法学和伦理挑战。在本文中,我们将评估抗癌药物剂量发现的历史和现状。我们为未来的批准后剂量优化试验提出了一系列战略重点。我们讨论了批准后剂量优化试验设计的伦理考虑因素,并回顾了这些独特试验的三大设计策略,它们既符合伦理标准,又有利于患者和资助者。最后,我们讨论了剂量优化领域的财务和报告注意事项。总之,我们对批准后的剂量优化试验进行了全面的鸟瞰,并就方法学、监管和资助制度所需的下一步措施提出了自己的想法。
{"title":"Optimizing the doses of cancer drugs after usual dose finding.","authors":"Garth W Strohbehn, Walter M Stadler, Philip S Boonstra, Mark J Ratain","doi":"10.1177/17407745231213882","DOIUrl":"10.1177/17407745231213882","url":null,"abstract":"<p><p>Since the middle of the 20th century, oncology's dose-finding paradigm has been oriented toward identifying a drug's maximum tolerated dose, which is then carried forward into phase 2 and 3 trials and clinical practice. For most modern precision medicines, however, maximum tolerated dose is far greater than the minimum dose needed to achieve maximal benefit, leading to unnecessary side effects. Regulatory change may decrease maximum tolerated dose's predominance by enforcing dose optimization of <i>new</i> drugs. Dozens of already approved cancer drugs require re-evaluation, however, introducing a new methodologic and ethical challenge in cancer clinical trials. In this article, we assess the history and current landscape of cancer drug dose finding. We provide a set of strategic priorities for postapproval dose optimization trials of the future. We discuss ethical considerations for postapproval dose optimization trial design and review three major design strategies for these unique trials that would both adhere to ethical standards and benefit patients and funders. We close with a discussion of financial and reporting considerations in the realm of dose optimization. Taken together, we provide a comprehensive, bird's eye view of the postapproval dose optimization trial landscape and offer our thoughts on the next steps required of methodologies and regulatory and funding regimes.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139039631","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evaluating whether the proportional odds models to analyse ordinal outcomes in COVID-19 clinical trials is providing clinically interpretable treatment effects: A systematic review. 评估用于分析COVID-19临床试验顺序结果的比例优势模型是否提供临床可解释的治疗效果:一项系统综述。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2023-11-20 DOI: 10.1177/17407745231211272
Masuma Uddin, Nasir Z Bashir, Brennan C Kahan

Background: After an initial recommendation from the World Health Organisation, trials of patients hospitalised with COVID-19 often include an ordinal clinical status outcome, which comprises a series of ordered categorical variables, typically ranging from 'Alive and discharged from hospital' to 'Dead'. These ordinal outcomes are often analysed using a proportional odds model, which provides a common odds ratio as an overall measure of effect, which is generally interpreted as the odds ratio for being in a higher category. The common odds ratio relies on the assumption of proportional odds, which implies an identical odds ratio across all ordinal categories; however, there is generally no statistical or biological basis for which this assumption should hold; and when violated, the common odds ratio may be a biased representation of the odds ratios for particular categories within the ordinal outcome. In this study, we aimed to evaluate to what extent the common odds ratio in published COVID-19 trials differed to simple binary odds ratios for clinically important outcomes.

Methods: We conducted a systematic review of randomised trials evaluating interventions for patients hospitalised with COVID-19, which used a proportional odds model to analyse an ordinal clinical status outcome, published between January 2020 and May 2021. We assessed agreement between the common odds ratio and the odds ratio from a standard logistic regression model for three clinically important binary outcomes: 'Alive', 'Alive without mechanical ventilation', and 'Alive and discharged from hospital'.

Results: Sixteen randomised clinical trials, comprising 38 individual comparisons, were included in this study; of these, only 6 trials (38%) formally assessed the proportional odds assumption. The common odds ratio differed by more than 25% compared to the binary odds ratios in 55% of comparisons for the outcome 'Alive', 37% for 'Alive without mechanical ventilation', and 24% for 'Alive and discharged from hospital'. In addition, the common odds ratio systematically underestimated the odds ratio for the outcome 'Alive' by -16.8% (95% confidence interval: -28.7% to -2.9%, p = 0.02), though differences for the other outcomes were smaller and not statistically significant (-8.4% for 'Alive without mechanical ventilation' and 3.6% for 'Alive and discharged from hospital'). The common odds ratio was statistically significant for 18% of comparisons, while the binary odds ratio was significant in 5%, 16%, and 3% of comparisons for the outcomes 'Alive', 'Alive without mechanical ventilation', and 'Alive and discharged from hospital', respectively.

Conclusion: The common odds ratio from proportional odds models often differs substantially to odds ratios from clinically important binary outcomes, and similar to composite outcomes, a beneficial common OR from a proportional odds model does not

背景:根据世界卫生组织的初步建议,对COVID-19住院患者的试验通常包括顺序临床状态结果,该结果由一系列有序的分类变量组成,通常从“活着并出院”到“死亡”。这些顺序结果通常使用比例赔率模型进行分析,该模型提供了一个共同的赔率比作为效果的总体衡量标准,通常将其解释为处于较高类别的赔率比。共同的优势比依赖于比例优势的假设,这意味着在所有有序类别中具有相同的优势比;然而,这种假设通常没有统计学或生物学依据;当违反时,共同比值比可能是顺序结果中特定类别的比值比的有偏表示。在本研究中,我们旨在评估已发表的COVID-19试验中的常见优势比与临床重要结果的简单二元优势比的差异程度。方法:我们对评估COVID-19住院患者干预措施的随机试验进行了系统回顾,使用比例优势模型分析了2020年1月至2021年5月期间发表的顺序临床状态结果。我们通过标准逻辑回归模型评估了常见优势比和优势比之间的一致性,这些优势比来自三个重要的临床二元结局:“活着”、“没有机械通气的活着”和“活着并出院”。结果:本研究纳入16项随机临床试验,包括38个个体比较;其中,只有6项试验(38%)正式评估了比例赔率假设。与55%的“存活”结果、37%的“无机械通气存活”结果和24%的“存活并出院”结果相比,普通优势比相差超过25%。此外,常见优势比系统地低估了“存活”结果的优势比-16.8%(95%置信区间:-28.7%至-2.9%,p = 0.02),尽管其他结果的差异较小且无统计学意义(“无机械通气存活”为-8.4%,“存活并出院”为3.6%)。在18%的比较中,共同优势比具有统计学意义,而在“存活”、“无机械通气存活”和“存活并出院”结果的比较中,二元优势比分别在5%、16%和3%的比较中具有统计学意义。结论:比例优势模型得出的共同优势比通常与临床重要的二元结果的优势比存在很大差异,与复合结果相似,比例优势模型得出的有益的共同优势比并不一定表明在有序结果中对最重要的类别有有益的影响。
{"title":"Evaluating whether the proportional odds models to analyse ordinal outcomes in COVID-19 clinical trials is providing clinically interpretable treatment effects: A systematic review.","authors":"Masuma Uddin, Nasir Z Bashir, Brennan C Kahan","doi":"10.1177/17407745231211272","DOIUrl":"10.1177/17407745231211272","url":null,"abstract":"<p><strong>Background: </strong>After an initial recommendation from the World Health Organisation, trials of patients hospitalised with COVID-19 often include an ordinal clinical status outcome, which comprises a series of ordered categorical variables, typically ranging from 'Alive and discharged from hospital' to 'Dead'. These ordinal outcomes are often analysed using a proportional odds model, which provides a common odds ratio as an overall measure of effect, which is generally interpreted as the odds ratio for being in a higher category. The common odds ratio relies on the assumption of proportional odds, which implies an identical odds ratio across all ordinal categories; however, there is generally no statistical or biological basis for which this assumption should hold; and when violated, the common odds ratio may be a biased representation of the odds ratios for particular categories within the ordinal outcome. In this study, we aimed to evaluate to what extent the common odds ratio in published COVID-19 trials differed to simple binary odds ratios for clinically important outcomes.</p><p><strong>Methods: </strong>We conducted a systematic review of randomised trials evaluating interventions for patients hospitalised with COVID-19, which used a proportional odds model to analyse an ordinal clinical status outcome, published between January 2020 and May 2021. We assessed agreement between the common odds ratio and the odds ratio from a standard logistic regression model for three clinically important binary outcomes: 'Alive', 'Alive without mechanical ventilation', and 'Alive and discharged from hospital'.</p><p><strong>Results: </strong>Sixteen randomised clinical trials, comprising 38 individual comparisons, were included in this study; of these, only 6 trials (38%) formally assessed the proportional odds assumption. The common odds ratio differed by more than 25% compared to the binary odds ratios in 55% of comparisons for the outcome 'Alive', 37% for 'Alive without mechanical ventilation', and 24% for 'Alive and discharged from hospital'. In addition, the common odds ratio systematically underestimated the odds ratio for the outcome 'Alive' by -16.8% (95% confidence interval: -28.7% to -2.9%, <i>p</i> = 0.02), though differences for the other outcomes were smaller and not statistically significant (-8.4% for 'Alive without mechanical ventilation' and 3.6% for 'Alive and discharged from hospital'). The common odds ratio was statistically significant for 18% of comparisons, while the binary odds ratio was significant in 5%, 16%, and 3% of comparisons for the outcomes 'Alive', 'Alive without mechanical ventilation', and 'Alive and discharged from hospital', respectively.</p><p><strong>Conclusion: </strong>The common odds ratio from proportional odds models often differs substantially to odds ratios from clinically important binary outcomes, and similar to composite outcomes, a beneficial common OR from a proportional odds model does not","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11134983/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138046292","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 patient perspective on dose optimization for anticancer treatments: A new era of cancer drug dosing-Challenging the "more is better" dogma. 从患者角度看抗癌治疗的剂量优化:抗癌药物剂量的新时代--挑战 "越多越好 "的教条。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2024-02-22 DOI: 10.1177/17407745241232428
Julia Maués, Anne Loeser, Janice Cowden, Sheila Johnson, Martha Carlson, Shing Lee

The Patient-Centered Dosing Initiative, a patient-led effort advocating for a paradigm shift in determining cancer drug dosing strategies, pioneers a departure from traditional oncology drug dosing practices. Historically, oncology drug dosing relies on identifying the maximum tolerated dose through phase 1 dose escalation methodology, favoring higher dosing for greater efficacy, often leading to higher toxicity. However, this approach is not universally applicable, especially for newer treatments like targeted therapies and immunotherapies. Patient-Centered Dosing Initiative challenges this "more is better" ethos, particularly as metastatic breast cancer patients themselves, as they not only seek longevity but also a high quality of life since most metastatic breast cancer patients stay on treatment for the rest of their lives. Surveying 1221 metastatic breast cancer patients and 119 oncologists revealed an evident need for flexible dosing strategies, advocating personalized care discussions based on patient attributes. The survey results also demonstrated an openness toward flexible dosing and a willingness from both patients and clinicians to discuss dosing as part of their care. Patient-centered dosing emphasizes dialogue between clinicians and patients, delving into treatment efficacy-toxicity trade-offs. Similarly, clinical trial advocacy for multiple dosing regimens encourages adaptive strategies, moving away from strict adherence to maximum tolerated dose, supported by recent research in optimizing drug dosages. Recognizing the efficacy-effectiveness gap between clinical trials and real-world practice, Patient-Centered Dosing Initiative underscores the necessity for patient-centered dosing strategies. A focus on individual patient attributes aligns with initiatives like Project Optimus and Project Renewal, aiming to optimize drug dosages for improved treatment outcomes at both the pre- and post-approval phases. Patient-Centered Dosing Initiative's efforts extend to patient education, providing tools to initiate dosage-related conversations with physicians. In addition, it emphasizes physician-patient dialogues and post-marketing studies as essential in determining optimal dosing and refining drug regimens. A dose-finding paradigm prioritizing drug safety, tolerability, and efficacy benefits all stakeholders, reducing emergency care needs and missed treatments for patients, aligning with oncologists' and patients' shared goals. Importantly, it represents a win-win scenario across healthcare sectors. In summary, the Patient-Centered Dosing Initiative drives transformative changes in cancer drug dosing, emphasizing patient well-being and personalized care, aiming to enhance treatment outcomes and optimize oncology drug delivery.

以患者为中心的用药倡议 "是一项由患者主导的工作,倡导转变癌症药物用药策略的模式,率先打破了传统的肿瘤药物用药惯例。从历史上看,肿瘤药物剂量依赖于通过第一阶段剂量升级方法确定最大耐受剂量,倾向于加大剂量以提高疗效,但往往会导致毒性增加。然而,这种方法并不普遍适用,尤其是对于靶向疗法和免疫疗法等较新的治疗方法。以患者为中心的用药倡议挑战了这种 "越多越好 "的理念,尤其是转移性乳腺癌患者本身,因为他们不仅追求长寿,还追求高质量的生活,因为大多数转移性乳腺癌患者终生都在接受治疗。对 1221 名转移性乳腺癌患者和 119 名肿瘤学家进行的调查显示,他们明显需要灵活的用药策略,提倡根据患者的特质进行个性化护理讨论。调查结果还表明,患者和临床医生对灵活用药持开放态度,并愿意将用药讨论作为治疗的一部分。以患者为中心的用药方式强调临床医生与患者之间的对话,深入探讨治疗效果与毒性之间的权衡。同样,临床试验提倡多种给药方案,鼓励采取适应性策略,不再严格遵守最大耐受剂量,这也得到了近期优化药物剂量研究的支持。认识到临床试验与实际应用之间的疗效差距,以患者为中心的用药倡议强调了以患者为中心的用药策略的必要性。对患者个体属性的关注与 "优化项目 "和 "更新项目 "等计划相一致,旨在优化药物剂量,以改善批准前和批准后阶段的治疗效果。以患者为中心的剂量倡议 "将工作延伸至患者教育,提供与医生展开剂量相关对话的工具。此外,它还强调医患对话和上市后研究对于确定最佳剂量和完善药物治疗方案至关重要。优先考虑药物安全性、耐受性和疗效的剂量确定范式有利于所有利益相关者,可减少患者的紧急护理需求和错过的治疗,符合肿瘤学家和患者的共同目标。重要的是,它代表了一种跨医疗保健领域的双赢方案。总之,"以患者为中心的剂量倡议 "推动了抗癌药物剂量的变革,强调了患者福祉和个性化护理,旨在提高治疗效果并优化肿瘤药物的交付。
{"title":"The patient perspective on dose optimization for anticancer treatments: A new era of cancer drug dosing-Challenging the \"more is better\" dogma.","authors":"Julia Maués, Anne Loeser, Janice Cowden, Sheila Johnson, Martha Carlson, Shing Lee","doi":"10.1177/17407745241232428","DOIUrl":"10.1177/17407745241232428","url":null,"abstract":"<p><p>The Patient-Centered Dosing Initiative, a patient-led effort advocating for a paradigm shift in determining cancer drug dosing strategies, pioneers a departure from traditional oncology drug dosing practices. Historically, oncology drug dosing relies on identifying the maximum tolerated dose through phase 1 dose escalation methodology, favoring higher dosing for greater efficacy, often leading to higher toxicity. However, this approach is not universally applicable, especially for newer treatments like targeted therapies and immunotherapies. Patient-Centered Dosing Initiative challenges this \"more is better\" ethos, particularly as metastatic breast cancer patients themselves, as they not only seek longevity but also a high quality of life since most metastatic breast cancer patients stay on treatment for the rest of their lives. Surveying 1221 metastatic breast cancer patients and 119 oncologists revealed an evident need for flexible dosing strategies, advocating personalized care discussions based on patient attributes. The survey results also demonstrated an openness toward flexible dosing and a willingness from both patients and clinicians to discuss dosing as part of their care. Patient-centered dosing emphasizes dialogue between clinicians and patients, delving into treatment efficacy-toxicity trade-offs. Similarly, clinical trial advocacy for multiple dosing regimens encourages adaptive strategies, moving away from strict adherence to maximum tolerated dose, supported by recent research in optimizing drug dosages. Recognizing the efficacy-effectiveness gap between clinical trials and real-world practice, Patient-Centered Dosing Initiative underscores the necessity for patient-centered dosing strategies. A focus on individual patient attributes aligns with initiatives like Project Optimus and Project Renewal, aiming to optimize drug dosages for improved treatment outcomes at both the pre- and post-approval phases. Patient-Centered Dosing Initiative's efforts extend to patient education, providing tools to initiate dosage-related conversations with physicians. In addition, it emphasizes physician-patient dialogues and post-marketing studies as essential in determining optimal dosing and refining drug regimens. A dose-finding paradigm prioritizing drug safety, tolerability, and efficacy benefits all stakeholders, reducing emergency care needs and missed treatments for patients, aligning with oncologists' and patients' shared goals. Importantly, it represents a win-win scenario across healthcare sectors. In summary, the Patient-Centered Dosing Initiative drives transformative changes in cancer drug dosing, emphasizing patient well-being and personalized care, aiming to enhance treatment outcomes and optimize oncology drug delivery.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139930421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Adaptive phase I-II clinical trial designs identifying optimal biological doses for targeted agents and immunotherapies. 适应性 I-II 期临床试验设计,确定靶向药物和免疫疗法的最佳生物剂量。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2024-01-11 DOI: 10.1177/17407745231220661
Yong Zang, Beibei Guo, Yingjie Qiu, Hao Liu, Mateusz Opyrchal, Xiongbin Lu

Targeted agents and immunotherapies have revolutionized cancer treatment, offering promising options for various cancer types. Unlike traditional therapies the principle of "more is better" is not always applicable to these new therapies due to their unique biomedical mechanisms. As a result, various phase I-II clinical trial designs have been proposed to identify the optimal biological dose that maximizes the therapeutic effect of targeted therapies and immunotherapies by jointly monitoring both efficacy and toxicity outcomes. This review article examines several innovative phase I-II clinical trial designs that utilize accumulated efficacy and toxicity outcomes to adaptively determine doses for subsequent patients and identify the optimal biological dose, maximizing the overall therapeutic effect. Specifically, we highlight three categories of phase I-II designs: efficacy-driven, utility-based, and designs incorporating multiple efficacy endpoints. For each design, we review the dose-outcome model, the definition of the optimal biological dose, the dose-finding algorithm, and the software for trial implementation. To illustrate the concepts, we also present two real phase I-II trial examples utilizing the EffTox and ISO designs. Finally, we provide a classification tree to summarize the designs discussed in this article.

靶向药物和免疫疗法为癌症治疗带来了革命性的变化,为各种癌症类型提供了前景广阔的治疗方案。与传统疗法不同,由于其独特的生物医学机制,"多多益善 "的原则并不总是适用于这些新疗法。因此,人们提出了各种 I-II 期临床试验设计,通过联合监测疗效和毒性结果,确定最佳生物剂量,最大限度地发挥靶向疗法和免疫疗法的治疗效果。本综述文章探讨了几种创新的 I-II 期临床试验设计,这些设计利用累积的疗效和毒性结果来适应性地确定后续患者的剂量,并确定最佳生物剂量,从而最大限度地提高整体治疗效果。具体来说,我们重点介绍了三类 I-II 期设计:疗效驱动型设计、基于效用的设计以及包含多个疗效终点的设计。对于每种设计,我们都会回顾剂量-结果模型、最佳生物剂量的定义、剂量寻找算法以及试验实施软件。为了说明这些概念,我们还介绍了两个利用 EffTox 和 ISO 设计的 I-II 期试验实例。最后,我们提供了一个分类树来总结本文所讨论的设计。
{"title":"Adaptive phase I-II clinical trial designs identifying optimal biological doses for targeted agents and immunotherapies.","authors":"Yong Zang, Beibei Guo, Yingjie Qiu, Hao Liu, Mateusz Opyrchal, Xiongbin Lu","doi":"10.1177/17407745231220661","DOIUrl":"10.1177/17407745231220661","url":null,"abstract":"<p><p>Targeted agents and immunotherapies have revolutionized cancer treatment, offering promising options for various cancer types. Unlike traditional therapies the principle of \"more is better\" is not always applicable to these new therapies due to their unique biomedical mechanisms. As a result, various phase I-II clinical trial designs have been proposed to identify the optimal biological dose that maximizes the therapeutic effect of targeted therapies and immunotherapies by jointly monitoring both efficacy and toxicity outcomes. This review article examines several innovative phase I-II clinical trial designs that utilize accumulated efficacy and toxicity outcomes to adaptively determine doses for subsequent patients and identify the optimal biological dose, maximizing the overall therapeutic effect. Specifically, we highlight three categories of phase I-II designs: efficacy-driven, utility-based, and designs incorporating multiple efficacy endpoints. For each design, we review the dose-outcome model, the definition of the optimal biological dose, the dose-finding algorithm, and the software for trial implementation. To illustrate the concepts, we also present two real phase I-II trial examples utilizing the EffTox and ISO designs. Finally, we provide a classification tree to summarize the designs discussed in this article.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11132954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139416580","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
Risk-benefit trade-offs and precision utilities in phase I-II clinical trials. I-II 期临床试验中的风险效益权衡和精确效用。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2023-12-18 DOI: 10.1177/17407745231214750
Pavlos Msaouel, Juhee Lee, Peter F Thall

Background: Identifying optimal doses in early-phase clinical trials is critically important. Therapies administered at doses that are either unsafe or biologically ineffective are unlikely to be successful in subsequent clinical trials or to obtain regulatory approval. Identifying appropriate doses for new agents is a complex process that involves balancing the risks and benefits of outcomes such as biological efficacy, toxicity, and patient quality of life.

Purpose: While conventional phase I trials rely solely on toxicity to determine doses, phase I-II trials explicitly account for both efficacy and toxicity, which enables them to identify doses that provide the most favorable risk-benefit trade-offs. It is also important to account for patient covariates, since one-size-fits-all treatment decisions are likely to be suboptimal within subgroups determined by prognostic variables or biomarkers. Notably, the selection of estimands can influence our conclusions based on the prognostic subgroup studied. For example, assuming monotonicity of the probability of response, higher treatment doses may yield more pronounced efficacy in favorable prognosis compared to poor prognosis subgroups when the estimand is mean or median survival. Conversely, when the estimand is the 3-month survival probability, higher treatment doses produce more pronounced efficacy in poor prognosis compared to favorable prognosis subgroups.

Methods and conclusions: Herein, we first describe why it is essential to consider clinical practice when designing a clinical trial and outline a stepwise process for doing this. We then review a precision phase I-II design based on utilities tailored to prognostic subgroups that characterize efficacy-toxicity risk-benefit trade-offs. The design chooses each patient's dose to optimize their expected utility and allows patients in different prognostic subgroups to have different optimal doses. We illustrate the design with a dose-finding trial of a new therapeutic agent for metastatic clear cell renal cell carcinoma.

背景:在早期临床试验中确定最佳剂量至关重要。用不安全或生物无效的剂量进行治疗不太可能在随后的临床试验中取得成功,也不太可能获得监管部门的批准。为新药确定合适的剂量是一个复杂的过程,涉及平衡生物疗效、毒性和患者生活质量等结果的风险和收益。目的:传统的 I 期试验仅依靠毒性来确定剂量,而 I-II 期试验则明确考虑疗效和毒性,这使它们能够确定风险-收益权衡最有利的剂量。考虑患者的协变量也很重要,因为在由预后变量或生物标志物决定的亚组中,"一刀切 "的治疗决策很可能不是最佳的。值得注意的是,根据所研究的预后亚组,估计因子的选择会影响我们的结论。例如,假设反应概率为单调性,当估计指标为平均生存期或中位生存期时,与预后不良亚组相比,较高的治疗剂量可能对预后良好的亚组产生更明显的疗效。相反,当估计指标为 3 个月生存概率时,与预后良好的亚组相比,较高的治疗剂量对预后不良的亚组产生更明显的疗效:在本文中,我们首先阐述了为什么在设计临床试验时必须考虑临床实践,并概述了设计临床试验的步骤。然后,我们回顾了一种基于效用的 I-II 期精准设计,这种效用是针对预后亚组量身定制的,能体现疗效-毒性风险-效益权衡的特点。该设计选择每位患者的剂量,以优化其预期效用,并允许不同预后亚组的患者使用不同的最佳剂量。我们以一种治疗转移性透明细胞肾细胞癌的新疗法的剂量探索试验来说明这种设计。
{"title":"Risk-benefit trade-offs and precision utilities in phase I-II clinical trials.","authors":"Pavlos Msaouel, Juhee Lee, Peter F Thall","doi":"10.1177/17407745231214750","DOIUrl":"10.1177/17407745231214750","url":null,"abstract":"<p><strong>Background: </strong>Identifying optimal doses in early-phase clinical trials is critically important. Therapies administered at doses that are either unsafe or biologically ineffective are unlikely to be successful in subsequent clinical trials or to obtain regulatory approval. Identifying appropriate doses for new agents is a complex process that involves balancing the risks and benefits of outcomes such as biological efficacy, toxicity, and patient quality of life.</p><p><strong>Purpose: </strong>While conventional phase I trials rely solely on toxicity to determine doses, phase I-II trials explicitly account for both efficacy and toxicity, which enables them to identify doses that provide the most favorable risk-benefit trade-offs. It is also important to account for patient covariates, since one-size-fits-all treatment decisions are likely to be suboptimal within subgroups determined by prognostic variables or biomarkers. Notably, the selection of estimands can influence our conclusions based on the prognostic subgroup studied. For example, assuming monotonicity of the probability of response, higher treatment doses may yield more pronounced efficacy in favorable prognosis compared to poor prognosis subgroups when the estimand is mean or median survival. Conversely, when the estimand is the 3-month survival probability, higher treatment doses produce more pronounced efficacy in poor prognosis compared to favorable prognosis subgroups.</p><p><strong>Methods and conclusions: </strong>Herein, we first describe why it is essential to consider clinical practice when designing a clinical trial and outline a stepwise process for doing this. We then review a precision phase I-II design based on utilities tailored to prognostic subgroups that characterize efficacy-toxicity risk-benefit trade-offs. The design chooses each patient's dose to optimize their expected utility and allows patients in different prognostic subgroups to have different optimal doses. We illustrate the design with a dose-finding trial of a new therapeutic agent for metastatic clear cell renal cell carcinoma.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11132955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138799238","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
Statistical and practical considerations in planning and conduct of dose-optimization trials. 规划和进行剂量优化试验的统计和实际考虑因素。
IF 2.7 3区 医学 Q3 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-06-01 Epub Date: 2024-01-19 DOI: 10.1177/17407745231207085
Ying Yuan, Heng Zhou, Suyu Liu

The U.S. Food and Drug Administration launched Project Optimus with the aim of shifting the paradigm of dose-finding and selection toward identifying the optimal biological dose that offers the best balance between benefit and risk, rather than the maximum tolerated dose. However, achieving dose optimization is a challenging task that involves a variety of factors and is considerably more complicated than identifying the maximum tolerated dose, both in terms of design and implementation. This article provides a comprehensive review of various design strategies for dose-optimization trials, including phase 1/2 and 2/3 designs, and highlights their respective advantages and disadvantages. In addition, practical considerations for selecting an appropriate design and planning and executing the trial are discussed. The article also presents freely available software tools that can be utilized for designing and implementing dose-optimization trials. The approaches and their implementation are illustrated through real-world examples.

美国食品和药物管理局启动 "Optimus 项目 "的目的是将剂量寻找和选择的范式从最大耐受剂量转变为能在效益和风险之间取得最佳平衡的最佳生物剂量。然而,实现剂量优化是一项具有挑战性的任务,涉及多种因素,在设计和实施方面都比确定最大耐受剂量复杂得多。本文全面回顾了剂量优化试验的各种设计策略,包括1/2期和2/3期设计,并强调了它们各自的优缺点。此外,文章还讨论了选择适当设计、规划和执行试验的实际注意事项。文章还介绍了可用于设计和实施剂量优化试验的免费软件工具。文章通过实际案例对这些方法及其实施进行了说明。
{"title":"Statistical and practical considerations in planning and conduct of dose-optimization trials.","authors":"Ying Yuan, Heng Zhou, Suyu Liu","doi":"10.1177/17407745231207085","DOIUrl":"10.1177/17407745231207085","url":null,"abstract":"<p><p>The U.S. Food and Drug Administration launched Project Optimus with the aim of shifting the paradigm of dose-finding and selection toward identifying the optimal biological dose that offers the best balance between benefit and risk, rather than the maximum tolerated dose. However, achieving dose optimization is a challenging task that involves a variety of factors and is considerably more complicated than identifying the maximum tolerated dose, both in terms of design and implementation. This article provides a comprehensive review of various design strategies for dose-optimization trials, including phase 1/2 and 2/3 designs, and highlights their respective advantages and disadvantages. In addition, practical considerations for selecting an appropriate design and planning and executing the trial are discussed. The article also presents freely available software tools that can be utilized for designing and implementing dose-optimization trials. The approaches and their implementation are illustrated through real-world examples.</p>","PeriodicalId":10685,"journal":{"name":"Clinical Trials","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11134987/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139502342","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
Clinical Trials
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:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1