IQWiG

IQWiG
  • 文章类型: Journal Article
    背景:对于时间到事件的端点,另外还开发了3种获益评估方法,目的是对新批准的治疗方法的临床获益程度进行无偏见了解.美国临床肿瘤学会(ASCO)使用风险比点估计(HR-PE)定义了连续评分。欧洲医学肿瘤学会(ESMO)和德国医疗保健质量和效率研究所(IQWiG)开发了使用95%HR置信区间(HR-CI)的下限和上限的顺序结果的方法。分别。我们描述了用于额外利益评估的所有三个框架,旨在对不同利益相关者进行公平比较。此外,我们确定哪个ASCO评分与哪个ESMO/IQWiG类别一致.
    方法:在具有不同故障时间分布和处理效果的综合模拟研究中,我们使用Spearman相关性和描述性度量比较了所有方法。为了确定符合ESMO/IQWiG类别的ASCO值,使用了最大化加权科恩的Kappa方法。
    结果:我们的研究描述了ASCO/IQWiG之间的高度正相关和ASCO/ESMO之间的低正相关。小于17、17至20、20至24和大于24的ASCO分数对应于ESMO类别。使用ASCO值21和38作为截止值表示IQWiG类别。
    结论:我们调查了方法的统计学方面,因此实施了所有方法的略微缩减版本。
    结论:IQWiG和ASCO比ESMO更保守,通常授予与真实效果无关的最大类别,并且有可能因各种故障时间分布而过度补偿。ASCO具有与IQWiG相似的特性。延迟治疗效果和动力不足/过度研究在一定程度上影响所有方法。然而,ESMO是最自由的。
    结论:对于额外的收益评估,美国临床肿瘤学会(ASCO)使用风险比点估计值(HR-PE)作为其连续评分.相比之下,欧洲医学肿瘤学会(ESMO)和德国医疗保健质量和效率研究所(IQWiG)使用下限和上限95%的HR置信区间(HR-CI)到特定阈值,分别。ESMO慷慨地分配最大分数,而IQWiG更保守。本研究提供了IQWiG和ASCO之间的首次比较,并描述了所有三个额外收益评估框架,旨在对不同利益相关者进行公平比较。此外,确定符合ESMO和IQWiG类别的ASCO阈值,能够以公平的方式对实践中的方法进行比较。IQWiG和ASCO是比较保守的方法,虽然ESMO授予最高类别的高百分比,特别是在各种故障时间分布的情况下。ASCO具有与IQWiG相似的特性。延迟的治疗效果和/-过度研究影响所有方法。然而,ESMO是最自由的。小于17、17至20、20至24和大于24的ASCO分数对应于ESMO的类别。使用ASCO值21和38作为截止值表示IQWiG的类别。
    BACKGROUND: For time-to-event endpoints, three additional benefit assessment methods have been developed aiming at an unbiased knowledge about the magnitude of clinical benefit of newly approved treatments. The American Society of Clinical Oncology (ASCO) defines a continuous score using the hazard ratio point estimate (HR-PE). The European Society for Medical Oncology (ESMO) and the German Institute for Quality and Efficiency in Health Care (IQWiG) developed methods with an ordinal outcome using lower and upper limits of the 95% HR confidence interval (HR-CI), respectively. We describe all three frameworks for additional benefit assessment aiming at a fair comparison across different stakeholders. Furthermore, we determine which ASCO score is consistent with which ESMO/IQWiG category.
    METHODS: In a comprehensive simulation study with different failure time distributions and treatment effects, we compare all methods using Spearman\'s correlation and descriptive measures. For determination of ASCO values consistent with categories of ESMO/IQWiG, maximizing weighted Cohen\'s Kappa approach was used.
    RESULTS: Our research depicts a high positive relationship between ASCO/IQWiG and a low positive relationship between ASCO/ESMO. An ASCO score smaller than 17, 17 to 20, 20 to 24, and greater than 24 corresponds to ESMO categories. Using ASCO values of 21 and 38 as cutoffs represents IQWiG categories.
    CONCLUSIONS: We investigated the statistical aspects of the methods and hence implemented slightly reduced versions of all methods.
    CONCLUSIONS: IQWiG and ASCO are more conservative than ESMO, which often awards the maximal category independent of the true effect and is at risk of overcompensating with various failure time distributions. ASCO has similar characteristics as IQWiG. Delayed treatment effects and underpowered/overpowered studies influence all methods in some degree. Nevertheless, ESMO is the most liberal one.
    CONCLUSIONS: For the additional benefit assessment, the American Society of Clinical Oncology (ASCO) uses the hazard ratio point estimate (HR-PE) for their continuous score. In contrast, the European Society for Medical Oncology (ESMO) and the German Institute for Quality and Efficiency in Health Care (IQWiG) use the lower and upper 95% HR confidence interval (HR-CI) to specific thresholds, respectively. ESMO generously assigns maximal scores, while IQWiG is more conservative.This research provides the first comparison between IQWiG and ASCO and describes all three frameworks for additional benefit assessment aiming for a fair comparison across different stakeholders. Furthermore, thresholds for ASCO consistent with ESMO and IQWiG categories are determined, enabling a comparison of the methods in practice in a fair manner.IQWiG and ASCO are the more conservative methods, while ESMO awards high percentages of maximal categories, especially with various failure time distributions. ASCO has similar characteristics as IQWiG. Delayed treatment effects and under/-overpowered studies influence all methods. Nevertheless, ESMO is the most liberal one. An ASCO score smaller than 17, 17 to 20, 20 to 24, and greater than 24 correspond to the categories of ESMO. Using ASCO values of 21 and 38 as cutoffs represents categories of IQWiG.
    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
    从2025年开始,健康技术开发人员(HTD)必须提交欧盟HTA档案。联合临床评估(JCA)旨在简化整个欧洲的HTA流程和药品的获取。目前,德国HTA机构IQWiG和G-BA积极塑造JCA方法。在这里,我们检查德国HTA档案要求是否适合JCA。我们将德国档案中安全性终点和亚组分析的数量与IQWIG获益评估中考虑的分析进行比较,并评估G-BA是否考虑了这些分析。我们进一步调查了德国档案模板的最新变化对分析数量的影响。使用当前模板,与以前的模板相比,HTDs报告的不良事件(AE)分析中位数为2.6倍,亚组类别为1.1倍。IQWiG不考虑33%的AE分析和73%的HTD在当前模板下呈现的亚组类别。在76%的病例中,G-BA认为与IQWiG相同的不良事件。在大多数情况下,亚组未被G-BA评论,独立于模板(以前:93%,目前的85%),在关于额外福利的结论中没有考虑(以前:77%,当前69%)。因此,档案模板的变化大大增加了HTD的工作量,但是HTA机构似乎没有考虑其他分析。随着JCA范围的扩大,这种影响可能会被放大。为了减轻重复的努力,并确保按照HTAR的设想迅速获得医药产品,我们建议精心挑选和精确的档案要求,早期磋商,和早期的HTD参与。
    From 2025, Health Technology Developers (HTDs) have to submit EU HTA dossiers. The joint clinical assessment (JCA) aims to streamline HTA processes and access to medicinal products across Europe. Currently, German HTA bodies IQWiG and G-BA actively shape the JCA methodology. Here we examine if German HTA dossier requirements are suitable for the JCA. We compare the number of safety endpoint and subgroup analyses in German dossiers with analyses considered in IQWIG\'s benefit assessment and evaluate if these analyses were considered by the G-BA. We further investigated how the number of analyses was affected by the latest change in the German dossier template. With the current template, HTDs report in median 2.6 times more analyses on adverse events (AE) and 1.1 times more subgroup categories than in the previous template. IQWiG does not consider 33% of AE analyses and 73% of the subgroup categories presented by the HTD under the current template. G-BA considered the same AE as IQWiG in 76% of cases. Subgroups were uncommented by G-BA in most cases, independent of the template (previous: 93%, current 85%) and unconsidered in the conclusion on additional benefit (previous: 77%, current 69%). Thus, changes in the dossier template drastically increased HTD workload, but additional analyses seem unconsidered by the HTA bodies. With a broader scope in JCA, this effect could be amplified. To mitigate duplicative efforts and ensure prompt availability of medicinal products as envisioned by the HTAR, we suggest well-chosen and precise dossier requirements, early consultations, and early HTD engagement.
    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
    患者报告的结果(PRO)数据越来越多地包含在肿瘤药物的健康技术评估(HTA)提交中。这项研究旨在提供关键HTA机构在肿瘤学中PRO证据要求的差异,并呼吁其协调。德国HTA机构提供的方法指导,法国,和英国,本综述对20项肿瘤学病例研究的HTA报告进行了分析。HTA机构在如何收集PRO数据方面存在差异,报告和分析,以及如何在肿瘤学HTA中审查和考虑数据。HTA机构可以在与监管机构和赞助商合作协调PRO方法指导方面发挥关键作用。
    患者报告的结果(PRO)是由正在经历疾病或正在接受治疗的人直接提供的信息。无需临床医生或护理人员的额外解释。与其他结果指标一样,PRO可以包括在卫生技术评估机构在其审查中使用的证据主体中。在这篇文章中,作者总结了主要卫生技术评估机构发布的指导文件,并回顾了过去的20项癌症药物案例研究,以了解不同机构在决定新的癌症治疗建议时如何使用PRO.
    Patient-reported outcome (PRO) data are increasingly being included in Health Technology Assessment (HTA) submissions for oncology drugs. This study aims to provide differences in PRO evidence requirements in oncology across key HTA bodies and calls for its harmonization. Method guidance provided by HTA bodies in Germany, France, and the UK, and analysis of HTA reports of 20 oncology case studies were evaluated in this review. Differences exist between HTA bodies regarding guidance on how PRO data should be collected, reported and analyzed as well as how the data are reviewed and considered in oncology HTAs. HTA bodies can play a key role to harmonize PRO method guidance in collaboration with regulators and sponsors.
    Patient-reported outcomes (PRO) are information provided directly by the person who is experiencing a disease or undergoing a treatment, without additional interpretation by a clinician or caregiver. Along with other outcome measures, PROs may be included in the body of evidence used by health technology assessment bodies in their review. In this article, the authors summarize the guidance documents published by key health technology assessment agencies and reviewed 20 past cancer drug case studies to understand how different agencies use PROs when deciding on recommendations for new cancer treatments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    UNASSIGNED: To analyze how ophthalmic drugs fared in the early benefit assessment (EBA) after its introduction in Germany up to 2020 and to quantify its impact on their negotiated prices.
    UNASSIGNED: Relevant documents were screened and essential content on added benefit outcomes and the underlying evidence was extracted next to pricing information. In addition to descriptive statistics, cross-stakeholder analyses and agreement statistics were implemented.
    UNASSIGNED: Thirteen completed EBA were identified involving eight drugs. Only four drugs (30.8%) received an added benefit. The OR for no added benefit of ophthalmic drugs versus all other drugs was 2.971 (0.902-9.781). The agreement between manufacturers\' claims and decision-maker appraisals is fair (kappa 0.435). In all cases, evidence was derived for RCTs, but for different reasons, not all of them allowed direct comparisons with the comparator as defined by the decision-maker. The negotiated rebates on manufacturer\'s selling prices varied from 6.8% up to 47.4%. Nevertheless, the rebates for ophthalmic drugs (median 14.5%) were lower than those for all negotiated drugs (median 24%).
    UNASSIGNED: Over the past decade, the EBA of ophthalmic drugs was not necessarily a success story, but in most of the cases, the drugs were successful in the market.
    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: Letter
    BACKGROUND: The question of how to set the cost-effectiveness threshold for new, innovative medicines is a matter of ongoing controversy. One prominent proposal suggests that the cost-effectiveness threshold adopted by the U.K. National Institute for Health and Care Excellence (NICE) should represent the opportunity cost for the U.K. National Health Service resulting from the adoption of new medicines. The purpose of this article is to compare this proposal for the U.K. with the approach chosen by the Institute for Quality and Efficiency in Health Care (IQWiG) in Germany, which relies on indication-specific cost-effectiveness thresholds.
    UNASSIGNED: The \'ideal\' NICE and IQWiG surprisingly share the fundamental principle of inferring the willingness to pay from existing care. For this and other reasons, indication-specific thresholds based on IQWiG\'s methodology do not lead to more inefficiency at the health system\'s level than a generic threshold based on the \'ideal\' NICE (keeping other conditions the same). Also, applying either decision rule to one country will yield a similar long-term growth in population spending. Assuming that everything else is equal, both decision rules are predicted to decrease long-term expenditure growth. Convergence of the two decision rules would require, among others, ruling out waste in the \'ideal\' NICE\'s approach and, for IQWiG\'s approach, using the same relative weights for life expectancy and health-related quality of life as the quality-adjusted-life-year model.
    CONCLUSIONS: This article shows that both decision rules have notable commonalities in terms of inferring the willingness to pay from existing care and the projected impact on long-term growth in population spending.
    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
    BACKGROUND: The purpose of this study was to analyse the impact of commissioned addenda by the Federal Joint Committee (FJC) to the HTA body (IQWiG) and their agreement with FJC decisions and to identify potential additional decisive factors of FJC.
    METHODS: All available relevant documents up to end of 2017 were screened and essential content extracted. Next to descriptive statistics, differences between IQWiG and FJC were tested and explored by agreement statistics (Cohen\'s kappa and Fleiss\' kappa) and ordinal logistic regression.
    RESULTS: Most of the 90 addenda concerned oncological products. In all contingent comparisons, positive changes in added benefit or evidence level on a subpopulation basis (n = 124) prevailed negative ones. Fleiss\' ordinal kappa for agreement of assessments, addenda, and appraisals reached a moderate strength for added benefit (0.474, 95%-CI, 0.408-0.540). Overall agreement between addenda and appraisals on a binary nominal basis is poor for added benefit (Cohen\'s kappa 0.183; 95%-CI: 0.010-0.357) ranging from \"less than by chance\" (respiratory diseases) to \"perfect\" (neurological diseases). The OR of the selected regression model showed that i) mortality, ii) unmet need, the positions of iii) the physicians\' drug commission and iv) medical societies, and v) the annual therapeutic costs of the appropriate comparative therapy had a high influence on FJC\'s appraisals deviating from IQWiG\'s addenda recommendation.
    CONCLUSIONS: IQWiG\'s addenda have a high impact on decision-maker\'s appraisals offering additional analyses of supplementary evidence submitted by the manufacturers. Nevertheless, the agreement between addenda and appraisals varies, highlighting different decisive factors between IQWiG and FJC.
    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
    背景:自2011年1月以来,联邦联合委员会(FJC)对新批准的药物与适当的标准疗法进行了早期益处评估(EBA)。FJC委托医疗保健质量和效率研究所(IQEH)准备初步报告。我们的目标是评估程度,影响,以及对两家机构的额外利益有不同判断的原因。
    方法:我们在FJC网站上搜索了EBA数据,并包括2011年至2017年完成的程序。我们对FJC和IQEH在分歧判断上的差异进行了定量分析,定量分析EBA对市场退出的影响,和定性分析,找出导致判断分歧的潜在因素。
    结果:FJC在30%(457个中的139个)和22%(457个中的101个)匹配的研究问题(P=0.004)中对IQEH进行了评价。在EBA之后,28种药物从德国市场撤出。我们确定了三个可能导致不同判断的潜在因素。IQEH使用了一个独特的阈值概念来定义评级,FJC进行了额外的公开听证会,FJC在坚持严格标准和解释结果方面表现出更大的灵活性。
    结论:FJC和IQEH在早期获益评估方面存在显著差异。为了回应负面的EBA决定,制药公司从德国市场撤出了相当数量的药品。目前的工作揭示了收益评估固有的主观性和可能的差异,因为这两个机构遵守相同的议事规则。
    BACKGROUND: Since January 2011, the Federal Joint Committee (FJC) conducts early benefit assessments (EBA) of newly approved pharmaceutical drugs compared to appropriate standard therapies. The FJC commissions the Institute for Quality and Efficiency in Healthcare (IQEH) to prepare preliminary reports. We aimed to evaluate the extent, impact, and reason for different judgments on added benefit of both institutions.
    METHODS: We searched EBA data on the FJC website and included completed procedures from 2011 to 2017. We conducted a quantitative analysis of the difference between FJC and IQEH on divergent judgments, a quantitative analysis of the impact of EBA on market withdrawal, and a qualitative analysis to identify potential factors contributing to divergent judgments.
    RESULTS: FJC rated an added benefit in 30% (139 of 457) and IQEH in 22% (101 of 457) matching research questions (P = 0.004). In the aftermath of EBA, 28 pharmaceutical drugs were withdrawn from the German market. We identified three potential factors that might have contributed to the divergent judgments. IQEH used a unique threshold concept to define the rating, FJC conducted additional public hearings, and FJC showed more flexibility with adherence to stringent criteria and interpretation of results.
    CONCLUSIONS: FJC and IQEH differed significantly in their early benefit assessment. In response to negative EBA decisions, pharmaceutical companies withdrew a considerable number of medicines from the German market. The present work uncovers the subjectivity and possible variance inherent in benefit assessment, as the two institutions observe the same rules of procedure.
    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
    背景:医疗保健质量和效率研究所(IQWiG)用于量化德国九个领域的新药的早期效益评估中的附加效益的决策矩阵非常复杂,可以简化。此外,IQWiG使用的方法受到了多方面的批评:(1)它在其有利于总体生存的评估中对端点的加权不同,并且,因此,致命疾病的药物干预,(2)假设在评估药品制造商提交的档案时,有两个关键试验可用,导致对额外利益的量化产生深远的影响,and,(3)评估主要基于二分端点,因此导致可用证据的信息丢失。
    目的:调查批评是否合理,并提出方法学调整。
    方法:使用统计检验,多项逻辑回归和模拟对截至2016年底的可用档案进行分析。
    结果:结果表明,由于功率损耗,该方法不能确保结果在统计上是有效的,早期效益评估的结果可能会受到损害,尽管支持总体生存率的证据尚不清楚。修改,然而,IQWiG方法有可能解决已识别的问题。
    结论:通过与批准当局对确认性终点的方法趋同,可以简化决策矩阵,改进分析方法,将结果放在更有效的统计基础上。
    BACKGROUND: The decision matrix applied by the Institute for Quality and Efficiency in Health Care (IQWiG) for the quantification of added benefit within the early benefit assessment of new pharmaceuticals in Germany with its nine fields is quite complex and could be simplified. Furthermore, the method used by IQWiG is subject to manifold criticism: (1) it is implicitly weighting endpoints differently in its assessments favoring overall survival and, thereby, drug interventions in fatal diseases, (2) it is assuming that two pivotal trials are available when assessing the dossiers submitted by the pharmaceutical manufacturers, leading to far-reaching implications with respect to the quantification of added benefit, and, (3) it is basing the evaluation primarily on dichotomous endpoints and consequently leading to an information loss of usable evidence.
    OBJECTIVE: To investigate if criticism is justified and to propose methodological adaptations.
    METHODS: Analysis of the available dossiers up to the end of 2016 using statistical tests and multinomial logistic regression and simulations.
    RESULTS: It was shown that due to power losses, the method does not ensure that results are statistically valid and outcomes of the early benefit assessment may be compromised, though evidence on favoring overall survival remains unclear. Modifications, however, of the IQWiG method are possible to address the identified problems.
    CONCLUSIONS: By converging with the approach of approval authorities for confirmatory endpoints, the decision matrix could be simplified and the analysis method could be improved, to put the results on a more valid statistical basis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Medico-economic evaluations estimate, for a given health technology, the added cost and the clinical benefit compared to a reference strategy. The objective here is to analyze the criteria used to measure clinical benefit as the basis for market access and reimbursement decisions for drugs in oncology both in France and in Europe. Prolonged overall survival is the criterion of choice to demonstrate the benefit of an anticancer drug; a survival gain of 2 to 3 months or more would be considered as relevant for a new product versus the comparator. In the absence of survival benefit or mature data on survival, progression-free survival or symptom-free survival and the availability of alternative curative treatments, decrease in drug toxicity and quality of life improvement may be considered. Differences in clinical benefit assessment between regulatory agencies and payers are not specific to France. Case studies show that it is difficult to find a consistency in reimbursement and pricing decisions and to identify factors that may fully explain reimbursement decisions when survival benefit is not demonstrated.
    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
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号