Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Br J Clin Pharmacol ; 86(12): 2530-2534, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31426120

RESUMEN

Regulatory science underpins the objective evaluation of medicinal products. It is therefore imperative that regulatory science and expertise remain at the cutting edge so that innovations of ever-increasing complexity are translated safely and swiftly into effective, high-quality therapies. We undertook a comprehensive examination of the evolution of science and technology impacting on medicinal product evaluation over the next 5-10 years and this horizon-scanning activity was complemented by extensive stakeholder interviews, resulting in a number of significant recommendations. Highlighted in particular was the need for expertise and regulatory science research to fill knowledge gaps in both more fundamental, longer-term research, with respect to technological and product-specific challenges. A model is proposed to realise these objectives in Europe, comprising a synergistic relationship between the European Medicines Agency, the European Medicines Regulatory Network and academic research centres to establish a novel regulatory science and innovation platform.


Asunto(s)
Control de Medicamentos y Narcóticos , Conocimiento , Europa (Continente) , Humanos
3.
BMJ Open ; 9(5): e026764, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-31133588

RESUMEN

OBJECTIVES: Society is confronted with the rapid emergence of innovation in science and technology. To manage this, horizon scanning is being adopted globally to identify, assess and prioritise innovations and trends at an early stage of their development. This enables decision-makers to be better informed and to prepare for change. The aim of this paper is to systematically identify and evaluate horizon scanning methodologies employed in the healthcare and biomedical fields. METHODS: A systematic literature review was performed using PubMed and Embase and was supplemented with grey literature searches (2008-2018). The principal methodologies used in horizon scanning were extracted. RESULTS: Approximately 100 articles were summarised in a literature map. The search revealed many examples of horizon scanning across disciplines. Challenges, such as the need to refine prioritisation criteria, manage uncertainty inherent in the findings and improve the dissemination of identified issues, have been highlighted. CONCLUSION: Horizon scanning, when performed appropriately, is a flexible and potentially reliable tool, with a wide variety of methods. Horizon scanning can inform and influence decision-making, through identifying opportunities and challenges, from an organisational to an international level. Further research to identify the most effective methodologies available would add depth to this landscape and enable the evolution of best practice to most efficiently anticipate novel developments and innovations.


Asunto(s)
Atención a la Salud/tendencias , Difusión de Innovaciones , Evaluación de la Tecnología Biomédica/tendencias , Toma de Decisiones , Humanos , Formulación de Políticas
4.
Trials ; 19(1): 642, 2018 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-30454061

RESUMEN

BACKGROUND: In recent years, experience on the application of adaptive designs in confirmatory clinical trials has accumulated. Although planning such trials comes at the cost of additional operational complexity, adaptive designs offer the benefit of flexibility to update trial design and objectives as data accrue. In 2007, the European Medicines Agency (EMA) provided guidance on confirmatory clinical trials with adaptive (or flexible) designs. In order to better understand how adaptive trials are implemented in practice and how they may impact medicine approval within the EMA centralised procedure, we followed on 59 medicines for which an adaptive clinical trial had been submitted to the EMA Scientific Advice (SA) and analysed previously in a dedicated EMA survey of scientific advice letters. We scrutinized in particular the submission of the corresponding medicines for a marketing authorisation application (MAA). We also discuss the current regulatory perspective as regards the implementation of adaptive designs in confirmatory clinical trials. METHODS: Using the internal EMA MAA database, the AdisInsight database and related trial registries, we analysed how many of these 59 trials actually started, the completion status, results, the time to trial start, the adaptive elements finally implemented after SA, their possible influence on the success of the trial and corresponding product approval. RESULTS: Overall 31 trials out of 59 (53%) were retrieved. Thirty of them (97%) have been started and 23 (74%) concluded. Nine of these trials (39% out of 23) demonstrated a significant treatment effect on their primary endpoint and 4 (17% out of 23) supported a marketing authorisation (MA). An additional two trials were stopped using pre-defined criteria for futility, efficiently identifying trials on which further resources should not be spent. Median time to trial start after SA letter was given by EMA was 5 months. In the investigated trial registries, at least 18 trial (58% of 31 retrieved trials) designs were implemented with adaptive elements, which were predominantly dose selection, sample size reassessment (SSR) and stopping for futility (SFF). Among the 11 completed trials including adaptive elements, 6 demonstrated a significant treatment effect on their primary endpoint (55%). CONCLUSIONS: Adaptive designs are now well established in the drug development landscape. If properly pre-planned, adaptations can play a key role in the success of some of these trials, for example to help successfully select the most promising dose regimens for phase II/III trials. Interim analyses can also enable stopping of trials for futility when they do not hold their promises. Type I error rate control, trial integrity and results consistency between the different stages of the analyses are fundamental aspects to be discussed thoroughly. Engaging early dialogue with regulators and implementing the scientific advice received is strongly recommended, since much experience in discussing adaptive designs and assessing their results has been accumulated.


Asunto(s)
Ensayos Clínicos Adaptativos como Asunto/métodos , Aprobación de Drogas , Agencias Gubernamentales , Comercialización de los Servicios de Salud , Proyectos de Investigación , Ensayos Clínicos Adaptativos como Asunto/legislación & jurisprudencia , Aprobación de Drogas/legislación & jurisprudencia , Determinación de Punto Final , Europa (Continente) , Agencias Gubernamentales/legislación & jurisprudencia , Regulación Gubernamental , Humanos , Comercialización de los Servicios de Salud/legislación & jurisprudencia , Proyectos de Investigación/legislación & jurisprudencia , Tamaño de la Muestra , Factores de Tiempo
5.
Trials ; 15: 383, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25278265

RESUMEN

BACKGROUND: Since the first methodological publications on adaptive study design approaches in the 1990s, the application of these approaches in drug development has raised increasing interest among academia, industry and regulators. The European Medicines Agency (EMA) as well as the Food and Drug Administration (FDA) have published guidance documents addressing the potentials and limitations of adaptive designs in the regulatory context. Since there is limited experience in the implementation and interpretation of adaptive clinical trials, early interaction with regulators is recommended. The EMA offers such interactions through scientific advice and protocol assistance procedures. METHODS: We performed a text search of scientific advice letters issued between 1 January 2007 and 8 May 2012 that contained relevant key terms. Letters containing questions related to adaptive clinical trials in phases II or III were selected for further analysis. From the selected letters, important characteristics of the proposed design and its context in the drug development program, as well as the responses of the Committee for Human Medicinal Products (CHMP)/Scientific Advice Working Party (SAWP), were extracted and categorized. For 41 more recent procedures (1 January 2009 to 8 May 2012), additional details of the trial design and the CHMP/SAWP responses were assessed. In addition, case studies are presented as examples. RESULTS: Over a range of 5½ years, 59 scientific advices were identified that address adaptive study designs in phase II and phase III clinical trials. Almost all were proposed as confirmatory phase III or phase II/III studies. The most frequently proposed adaptation was sample size reassessment, followed by dropping of treatment arms and population enrichment. While 12 (20%) of the 59 proposals for an adaptive clinical trial were not accepted, the great majority of proposals were accepted (15, 25%) or conditionally accepted (32, 54%). In the more recent 41 procedures, the most frequent concerns raised by CHMP/SAWP were insufficient justifications of the adaptation strategy, type I error rate control and bias. CONCLUSIONS: For the majority of proposed adaptive clinical trials, an overall positive opinion was given albeit with critical comments. Type I error rate control, bias and the justification of the design are common issues raised by the CHMP/SAWP.


Asunto(s)
Comités Consultivos , Ensayos Clínicos Fase II como Asunto/métodos , Ensayos Clínicos Fase III como Asunto/métodos , Correspondencia como Asunto , Comercialización de los Servicios de Salud , Proyectos de Investigación , Comités Consultivos/normas , Ensayos Clínicos Fase II como Asunto/normas , Ensayos Clínicos Fase III como Asunto/normas , Europa (Continente) , Humanos , Guías de Práctica Clínica como Asunto , Proyectos de Investigación/normas , Tamaño de la Muestra
6.
Clin Cancer Res ; 20(6): 1458-68, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634469

RESUMEN

The European Union (EU) legal framework for medical device regulation is currently under revision. The European Commission has proposed a new framework to ensure that medical devices serve the needs and ensure the safety of European citizens, aiming for a framework that is fit for purpose, more transparent, and better adapted to scientific and technological progress. The proposed new framework is described as an evolution of the current regime keeping the same legal approach. An important proposed change is that companion diagnostics will no longer be considered as low risk and subject to self-certification by the manufacturer. According to the new proposal, companion diagnostics will be classified as high individual risk or moderate public health risk (category C) and require conformity assessment by a notified body. It has also been proposed that evidence of the clinical utility of the device for the intended purpose should be required for companion diagnostics. In this article, we review the EU legal framework relevant for companion diagnostics, describe the proposed changes, and summarize the available scientific guidance from the European Medicines Agency and its regulatory experience with cancer drug development including companion diagnostics. See all articles in this CCR Focus section, "The Precision Medicine Conundrum: Approaches to Companion Diagnostic Co-development."


Asunto(s)
Antineoplásicos/normas , Técnicas de Diagnóstico Molecular/normas , Terapia Molecular Dirigida/normas , Unión Europea , Humanos , Neoplasias/tratamiento farmacológico , Estados Unidos , United States Food and Drug Administration
7.
Regul Toxicol Pharmacol ; 68(3): 312-6, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24447908

RESUMEN

In this article we analyse the Environmental Risk Assessment (ERA) of 59 medicinal products for human use authorised in the EU through the centralised procedure between 2011 and 2012, to establish whether company submissions are compliant with the European Medicines Agency (EMA) guideline and complete in terms of data and study reports provided. The most frequent questions raised by EU regulatory authorities are described, together with an evaluation of the presence and quality of ERA-related information in published regulatory assessment documents. The results of this review show recent improvement in ERA-related data presented in regulatory assessment documents available to the public while also highlighting a need to develop further guidance on environmental issues in order to assist applicants improve their ERA dossiers and overcome current shortcomings.


Asunto(s)
Ambiente , Preparaciones Farmacéuticas , Aprobación de Drogas , Unión Europea , Humanos , Medición de Riesgo
8.
Pharmacogenomics ; 11(12): 1637-47, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21142906

RESUMEN

The 2010 US FDA-Drug Industry Association (DIA) Pharmacogenomics (PGx) Workshop follows a series that began in 2002 bringing together multidisciplinary experts spanning regulatory authorities, medical research, healthcare and industry. This report summarizes the 'Building PGx into Labels' sessions from the workshop, which discussed the critical elements in developing PGx outcomes leading to product labels that inform efficacy and/or safety. Examples were drawn from US prescribing information, which integrated PGx knowledge into medical decisions (e.g., panitumumab, warfarin and clopidogrel). Attendees indicated the need for broader dialog and for guidelines on evidentiary considerations for PGx to be included into product labels. Also discussed was the understanding of appropriate PGx placement on labels; how to encourage adoption by medical communities of label recommendations on PGx tests; and, given the global nature of drug development, worldwide considerations including European Summary of Product Characteristics.


Asunto(s)
Biomarcadores Farmacológicos/análisis , Diseño de Fármacos , Industria Farmacéutica , Etiquetado de Medicamentos/métodos , Programas de Gobierno , Farmacogenética/normas , Industria Farmacéutica/normas , Etiquetado de Medicamentos/legislación & jurisprudencia , Etiquetado de Medicamentos/tendencias , Farmacogenética/tendencias , Estados Unidos , United States Food and Drug Administration
9.
Nat Rev Drug Discov ; 9(6): 435-45, 2010 06.
Artículo en Inglés | MEDLINE | ID: mdl-20514070

RESUMEN

Heterogeneity in the underlying mechanisms of disease processes and inter-patient variability in drug responses are major challenges in drug development. To address these challenges, biomarker strategies based on a range of platforms, such as microarray gene-expression technologies, are increasingly being applied to elucidate these sources of variability and thereby potentially increase drug development success rates. With the aim of enhancing understanding of the regulatory significance of such biomarker data by regulators and sponsors, the US Food and Drug Administration initiated a programme in 2004 to allow sponsors to submit exploratory genomic data voluntarily, without immediate regulatory impact. In this article, a selection of case studies from the first 5 years of this programme - which is now known as the voluntary exploratory data submission programme, and also involves collaboration with the European Medicines Agency - are discussed, and general lessons are highlighted.


Asunto(s)
Aprobación de Drogas , Perfilación de la Expresión Génica , United States Food and Drug Administration , Alanina Transaminasa/sangre , Azetidinas/efectos adversos , Azetidinas/uso terapéutico , Bencilaminas/efectos adversos , Bencilaminas/uso terapéutico , Carcinoma de Células Renales/diagnóstico , Europa (Continente) , Fluorouracilo/efectos adversos , Marcadores Genéticos , Humanos , Cooperación Internacional , Neoplasias Renales/diagnóstico , Trasplante de Riñón , Farmacogenética , Piperazinas/farmacocinética , Piperazinas/uso terapéutico , Clorhidrato de Prasugrel , Medicina de Precisión , Tiofenos/farmacocinética , Tiofenos/uso terapéutico , Estados Unidos
10.
Nat Biotechnol ; 28(5): 446-54, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20458314

RESUMEN

Application of any new biomarker to support safety-related decisions during regulated phases of drug development requires provision of a substantial data set that critically assesses analytical and biological performance of that biomarker. Such an approach enables stakeholders from industry and regulatory bodies to objectively evaluate whether superior standards of performance have been met and whether specific claims of fit-for-purpose use are supported. It is therefore important during the biomarker evaluation process that stakeholders seek agreement on which critical experiments are needed to test that a biomarker meets specific performance claims, how new biomarker and traditional comparators will be measured and how the resulting data will be merged, analyzed and interpreted.


Asunto(s)
Biomarcadores , Descubrimiento de Drogas , Preparaciones Farmacéuticas , Animales , Descubrimiento de Drogas/legislación & jurisprudencia , Descubrimiento de Drogas/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Preparaciones Farmacéuticas/normas
11.
Clin Cases Miner Bone Metab ; 5(2): 149-54, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-22460999

RESUMEN

The identification of new risk factors for specific diseases is an enduring theme in medical research. Advances in molecular biology, genetics, and computational biology are accelerating the pace of this work. The research seeks to increase our understanding of the causes of diseases, but there is also hope that the recognition of new risk factors will lead to improved methods for identifying persons who are in the early stages of, or at high risk for, the diseases of concern. Research has shown, however, that a genomic biomarker must have a much stronger association with the disease outcome than we ordinarily see in etiologic research if it is to provide a basis for early diagnosis or prediction in individual patients. However, even if the literature contains ~150,000 reports of disease-associated molecular markers, there are still very few validated biomarkers of proven and robust clinical utility. At present there is no established, standardized means for validating the association between a marker (or set of markers) and clinical outcomes. The Regulatory Authorities have undertaken a number of initiatives in order to enhance the use of biomarkers in drug development, to promote a more informed drug development and maximise the benefit of innovative medicines to the patients.

12.
J Biomed Biotechnol ; 2003(1): 3-8, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12686717

RESUMEN

The evaluation of quality, safety, and efficacy of medicinal products by the European Medicines Evaluation Agency (EMEA) via the centralized procedure is the only available regulatory procedure for obtaining marketing authorization for gene therapy (GT) medicinal products in the European Union. The responsibility for the authorization of clinical trials remains with the national competent authorities (NCA) acting in a harmonized framework from the scientific viewpoint. With the entry into force of a new directive on good clinical practice implementation in clinical trials as of 1 May 2004, procedural aspects will also be harmonized at EU level. Scientifically sound development of medicinal products is the key for the successful registration of dossiers and for contributing to the promotion and protection of public health. The objective of this paper is to introduce the EMEA regulatory processes and scientific activities relevant to GT medicinal products.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA