?
当前位置:澳门黄金赌城 > 小柯机器人 >详情
欧洲药物管理局批准癌症药物的随机对照试验偏倚风险评估
作者:小柯机器人 发布时间:2019/9/23 13:12:36

英国伦敦政治经济学院Huseyin Naci课题组取得一项新突破。他们对2014-2016年欧洲药物管理局(EMA)支持批准癌症药物的随机对照试验的设计特征、偏倚风险和报告进行了一项横断面分析。 该项研究成果发表在2019年9月18日出版的《英国医学期刊》上。

2014-2016年间,EMA在54项关键研究的基础上批准了32种新癌症药物。其中41项(76%)为随机对照试验(39项已发表),13项(24%)为非随机研究或单臂研究。只有10项随机对照试验(26%)将总生存率作为主要终点,其余试验则评估无进展生存期和有效率等替代指标。

总体而言,19项随机对照试验(49%)被判定为其主要结果存在偏倚高风险。缺失结果数据(10项)和缺失结果衡量方法(7项)是导致偏倚高风险的主要因素。将总生存率作为主要终点的随机对照试验偏倚高风险所占比率(2/10,20% )显著低于评估替代疗效终点的试验(16/29,55%)。

若分开考虑监管文件和科学文献中提供的信息,则八项随机对照试验(21%)的偏倚总风险差别较大,这反映了两种信息来源报告的不足之处。在对10种药物进行偏倚风险评估时,监管机构确定了一些研究领域外的额外缺陷,包括临床获益的程度、不恰当的参照对象和非首选的研究终点,科学出版物没有披露这些局限。

综上,2014-2016年间,EMA批准新癌症药物最关键的研究基础是随机对照试验。然而,根据他们的设计、操作或分析,几乎一半的研究被判定为偏倚高风险,由于癌症试验的复杂性,其中一些或许不可避免。监管文件和科学文献的报告中存在漏洞。期刊出版物不承认监管文件中现有证据的主要局限性。

附:英文原文

Title: Design characteristics, risk of bias, and reporting of randomised controlled trials supporting approvals of cancer drugs by European Medicines Agency, 2014-16: cross sectional analysis

Author: Huseyin Naci, Courtney Davis, Jelena Savovi, Julian P T Higgins, Jonathan A C Sterne, Bishal Gyawali, Xochitl Romo-Sandoval, Nicola Handley, Christopher M Booth

Issue&Volume: Volume 366 Issue 8214

Abstract:

Objective To examine the design characteristics, risk of bias, and reporting adequacy of pivotal randomised controlled trials of cancer drugs approved by the European Medicines Agency (EMA).

Design Cross sectional analysis.

Setting European regulatory documents, clinical trial registry records, protocols, journal publications, and supplementary appendices.

Eligibility criteria Pivotal randomised controlled trials of new cancer drugs approved by the EMA between 2014 and 2016.

Main outcome measures Study design characteristics (randomisation, comparators, and endpoints); risk of bias using the revised Cochrane tool (bias arising from the randomisation process, deviations from intended interventions, missing outcome data, measurement of the outcome, and selection of the reported result); and reporting adequacy (completeness and consistency of information in trial protocols, publications, supplementary appendices, clinical trial registry records, and regulatory documents).

Results Between 2014 and 2016, the EMA approved 32 new cancer drugs on the basis of 54 pivotal studies. Of these, 41 (76%) were randomised controlled trials and 13 (24%) were either non-randomised studies or single arm studies. 39/41 randomised controlled trials had available publications and were included in our study. Only 10 randomised controlled trials (26%) measured overall survival as either a primary or coprimary endpoint, with the remaining trials evaluating surrogate measures such as progression free survival and response rates. Overall, 19 randomised controlled trials (49%) were judged to be at high risk of bias for their primary outcome. Concerns about missing outcome data (n=10) and measurement of the outcome (n=7) were the most common domains leading to high risk of bias judgments. Fewer randomised controlled trials that evaluated overall survival as the primary endpoint were at high risk of bias than those that evaluated surrogate efficacy endpoints (2/10 (20%) v 16/29 (55%), respectively). When information available in regulatory documents and the scientific literature was considered separately, overall risk of bias judgments differed for eight randomised controlled trials (21%), which reflects reporting inadequacies in both sources of information. Regulators identified additional deficits beyond the domains captured in risk of bias assessments for 10 drugs (31%). These deficits included magnitude of clinical benefit, inappropriate comparators, and non-preferred study endpoints, which were not disclosed as limitations in scientific publications.

Conclusions Most pivotal studies forming the basis of EMA approval of new cancer drugs between 2014 and 2016 were randomised controlled trials. However, almost half of these were judged to be at high risk of bias based on their design, conduct, or analysis, some of which might be unavoidable because of the complexity of cancer trials. Regulatory documents and the scientific literature had gaps in their reporting. Journal publications did not acknowledge the key limitations of the available evidence identified in regulatory documents.

DOI: 10.1136/bmj.l5221

Source: https://www.bmj.com/content/366/bmj.l5221

期刊信息

BMJ-British Medical Journal:《英国医学杂志》,创刊于1840年。隶属于BMJ出版集团,最新IF:27.604
官方网址:http://www.bmj.com/
投稿链接:https://mc.manuscriptcentral.com/bmj

?