dose expansion

  • 文章类型: Journal Article
    背景:蛋白质精氨酸甲基转移酶5(PRMT5)甲基化多种底物在癌症中失调,包括剪接体机械部件。PF-06939999是一种选择性小分子PRMT5抑制剂。
    方法:该I期剂量递增和扩展试验(NCT03854227)纳入了患有选定实体瘤的患者。PF-06939999在28天周期中每天口服一次或两次(q.d./b.i.d.)。目的是评估PF-06939999的安全性和耐受性,以确定最大耐受剂量(MTD)和推荐的第2部分剂量(RP2D)。并评估药代动力学(PK),药效学[血浆对称二甲基精氨酸(SDMA)水平的变化],和抗肿瘤活性。
    结果:在第1部分剂量递增中,28例患者接受PF-06939999(0.5mgq.d.至6mgb.i.d.)。24例患者中有4例(17%)报告了剂量限制性毒性:血小板减少症(n=2,6mgb.i.d.),贫血(n=1,8mgq.d.),和中性粒细胞减少症(n=1,6mgq.d.)。PF-06939999暴露量随剂量增加而增加。到第15天达到稳态PK。血浆SDMA在稳态下降低(58%-88%)。血浆SDMA的调节是剂量依赖性的。没有确定MTD。在第二部分剂量扩张中,26例患者接受PF-069399996mgq.d.(RP2D)。总体(第1部分+第2部分),最常见的≥3级治疗相关不良事件包括贫血(28%),血小板减少症/血小板计数减少(22%),疲劳(6%),和中性粒细胞减少症(4%)。3例患者(6.8%)确认部分缓解(头颈部鳞状细胞癌,n=1;非小细胞肺癌,n=2),19例(43.2%)病情稳定。没有发现预测性生物标志物。
    结论:PF-06939999在一部分患者中表现出可耐受的安全性和客观的临床反应,表明PRMT5是一个有趣的癌症靶标,具有临床验证。然而,未发现预测性生物标志物.PRMT5在癌症生物学中的作用是复杂的,需要进一步的临床前,机械调查,以确定用于患者选择的预测性生物标志物。
    BACKGROUND: Protein arginine methyltransferase 5 (PRMT5) methylates multiple substrates dysregulated in cancer, including spliceosome machinery components. PF-06939999 is a selective small-molecule PRMT5 inhibitor.
    METHODS: This phase I dose-escalation and -expansion trial (NCT03854227) enrolled patients with selected solid tumors. PF-06939999 was administered orally once or twice a day (q.d./b.i.d.) in 28-day cycles. The objectives were to evaluate PF-06939999 safety and tolerability to identify maximum tolerated dose (MTD) and recommended part 2 dose (RP2D), and assess pharmacokinetics (PK), pharmacodynamics [changes in plasma symmetric dimethylarginine (SDMA) levels], and antitumor activities.
    RESULTS: In part 1 dose escalation, 28 patients received PF-06939999 (0.5 mg q.d. to 6 mg b.i.d.). Four of 24 (17%) patients reported dose-limiting toxicities: thrombocytopenia (n = 2, 6 mg b.i.d.), anemia (n = 1, 8 mg q.d.), and neutropenia (n = 1, 6 mg q.d.). PF-06939999 exposure increased with dose. Steady-state PK was achieved by day 15. Plasma SDMA was reduced at steady state (58%-88%). Modulation of plasma SDMA was dose dependent. No MTD was determined. In part 2 dose expansion, 26 patients received PF-06939999 6 mg q.d. (RP2D). Overall (part 1 + part 2), the most common grade ≥3 treatment-related adverse events included anemia (28%), thrombocytopenia/platelet count decreased (22%), fatigue (6%), and neutropenia (4%). Three patients (6.8%) had confirmed partial response (head and neck squamous cell carcinoma, n = 1; non-small-cell lung cancer, n = 2), and 19 (43.2%) had stable disease. No predictive biomarkers were identified.
    CONCLUSIONS: PF-06939999 demonstrated a tolerable safety profile and objective clinical responses in a subset of patients, suggesting that PRMT5 is an interesting cancer target with clinical validation. However, no predictive biomarker was identified. The role of PRMT5 in cancer biology is complex and requires further preclinical, mechanistic investigation to identify predictive biomarkers for patient selection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    I期癌症试验越来越多地纳入剂量扩展队列(DEC),反映了对获取更多研究药物信息的需求日益增长。协议通常无法为DEC提供样本大小的理由或分析计划。在这项研究中,我们为DECs的设计开发了一个统计框架。
    我们假设研究药物的最大耐受剂量(MTD)已在试验的剂量递增阶段确定。我们使用80%的置信下限和90%的置信上限作为反应率和毒性率,分别,作为剂量扩展阶段的决策阈值。我们参考预定的最小有效响应率和最大安全DLT率计算操作特性。
    我们应用我们的框架来指定DEC计划的系统。该设计包括三个部分:1)在MTD登记的受试者数量,2)指示临时药物功效所需的最少回应次数,和3)允许指示药物安全性的剂量限制性毒性(DLT)的最大数目。我们在癌症免疫治疗试验的应用中展示了我们的方法。
    我们简单实用的工具可以创建DEC设计,以适当地解决试验的安全性和有效性目标。
    Phase I cancer trials increasingly incorporate dose-expansion cohorts (DECs), reflecting a growing demand to acquire more information about investigational drugs. Protocols commonly fail to provide a sample-size justification or analysis plan for the DEC. In this study, we develop a statistical framework for the design of DECs.
    We assume the maximum tolerated dose (MTD) for the investigational drug has been identified in the dose-escalation stage of the trial. We use the 80% lower confidence bound and the 90% upper confidence bound for the response and toxicity rates, respectively, as decision thresholds for the dose-expansion stage. We calculate the operating characteristics with reference to prespecified minimum effective response rates and maximum safe DLT rates.
    We apply our framework to specify a system of DEC plans. The design comprises three components: 1) the number of subjects enrolled at the MTD, 2) the minimum number of responses necessary to indicate provisional drug efficacy, and 3) the maximum number of dose-limiting toxicities (DLTs) permitted to indicate drug safety. We demonstrate our method in an application to a cancer immunotherapy trial.
    Our simple and practical tool enables creation of DEC designs that appropriately address the safety and efficacy objectives of the trial.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    迅速增加的I期剂量发现研究,特别是那些基于标准3+3设计的,正在延长纳入剂量扩展队列(DEC)的时间,以便更好地表征实验药物的毒性特征并研究疾病特异性队列。这些试验包括两个阶段:通常的剂量递增阶段,旨在建立最大耐受剂量(MTD),以及增加额外患者的剂量扩张阶段,通常具有不同的资格标准,以及收集其他信息的地方。当前的协议并不总是指定是否以及如何根据从DEC累积的新数据来更新MTD。在本文中,我们提出了通过根据其他信息重新评估MTD来监测DEC安全性的方法.我们的工作假设是,无论用于剂量递增的设计如何,在DEC期间,我们正在目标剂量附近进行实验,毒性率可接受。我们通过在MTD的初始估计的附近继续实验来完善我们对MTD的初始估计。在这种评估中提供的辅助信息可以包括毒性,药代动力学,功效或其他终点。我们考虑专门针对DEC的目标的方法,该方法可以单独或同时检查有效性和安全性,并通过模拟比较建议的测试。
    A rapidly increasing number of Phase I dose-finding studies, and in particular those based on the standard 3+3 design, are being prolonged with the inclusion of dose expansion cohorts (DEC) in order to better characterize the toxicity profiles of experimental agents and to study disease-specific cohorts. These trials consist of two phases: the usual dose escalation phase that aims to establish the maximum tolerated dose (MTD), and the dose expansion phase that accrues additional patients, often with different eligibility criteria, and where additional information is collected. Current protocols do not always specify whether and how the MTD will be updated in light of the new data accumulated from the DEC. In this paper, we propose methods that allow monitoring of safety in the DEC by re-evaluating the MTD in light of additional information. Our working assumption is that, regardless of the design being used for dose escalation, during the DEC we are experimenting in the neighborhood of a target dose with an acceptable rate of toxicity. We refine our initial estimate of the MTD by continuing experimentation in the immediate vicinity of the initial estimate of the MTD. The auxiliary information provided in such an evaluation can include toxicity, pharmacokinetic, efficacy or other endpoints. We consider approaches specifically focused on the aims of DEC that examine efficacy alone or simultaneously with safety and compare the proposed tests via simulations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    We focus on Phase I dose finding studies as they are currently undertaken. The design and analysis of these trials have changed over the last years and, in particular, it is now rare for a Phase I study to not include one or more dose-expansion cohorts (DEC). It is common to see DEC involving several hundred patients, building on an initial dose escalation study that may have no >20 to 30 patients. There has been recent focus by researchers on the design of DEC and the analysis of DEC data. It is reasonable to explicitly account for the uncertainty in the estimation of the MTD, the dose upon which the whole of the DEC is currently based. In this paper, we focus on the dose escalation phase prior to the DEC, with the purpose of adapting it to the needs of DEC. Specifically, before beginning the DEC phase, we need to identify those dose levels that will be taken into the DEC. We define a useful concept for this purpose, the co-MTD, and the results support that the estimated MTD and co-MTD contain the true MTD with high probability. We also provide stopping rules for when the data support that the dose escalation can end and the dose expansion can begin. Simulated trials support the use of the proposed approach and provide additional information on how this approach compares with current practice.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    A relatively recent development in the design of Phase I dose-finding studies is the inclusion of expansion cohort(s), that is, the inclusion of several more patients at a level considered to be the maximum tolerated dose established at the conclusion of the \'pure\' Phase I part. Little attention has been given to the additional statistical analysis, including design considerations, that we might wish to consider for this more involved design. For instance, how can we best make use of new information that may confirm or may tend to contradict the estimate of the maximum tolerated dose based on the dose escalation phase. Those patients included during the dose expansion phase may possess different eligibility criteria. During the expansion phase, we will also wish to have an eye on any evidence of efficacy, an aspect that clearly distinguishes such studies from the classical Phase I study. Here, we present a methodology that enables us to continue the monitoring of safety in the dose expansion cohort while simultaneously trying to assess efficacy and, in particular, which disease types may be the most promising to take forward for further study. The most elementary problem is where we only wish to take account of further toxicity information obtained during the dose expansion cohort, and where the initial design was model based or the standard 3+3. More complex set-ups also involve efficacy and the presence of subgroups. Copyright © 2016 John Wiley & Sons, Ltd.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号