Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Neuromodulation ; 27(3): 447-454, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37306642

RESUMO

BACKGROUND: Implantable neurostimulation devices must be authorized before they are placed on the market. For this purpose, requirements, and processes for assessing their fulfillment, have been defined in different jurisdictions. OBJECTIVE: In this study, we aimed to address differences between the US and European Union (EU) regulatory systems and their relationship to innovation. MATERIALS AND METHODS: A literature review and analysis were conducted using legal texts and guidance documents. RESULTS: The US system has one central body, the Food and Drug Administration, whereas the EU system has several bodies with different responsibilities. The devices themselves are divided into risk classes, which are based on the vulnerability of the human body. This risk class determines the intensity of the review by the market authorization body. In addition to the requirements for development, manufacture, and distribution, the device itself must meet technical and clinical requirements. Compliance with technical requirements is indicated by nonclinical laboratory studies. Proof of efficacy is provided by means of clinical investigations. Procedures are defined for reviewing these elements. Once the market authorization process has been completed, the devices can be placed on the market. In the postmarketing phase, the devices must continue to be monitored, and measures must be initiated, if necessary. CONCLUSIONS: Both US and EU systems are intended to ensure that only safe and effective devices find their way to and remain on the market. The basic approaches of the two systems are comparable. In detail, however, there are differences in ways these goals are achieved.


Assuntos
Próteses e Implantes , Estados Unidos , Humanos , União Europeia , United States Food and Drug Administration
2.
Br J Clin Pharmacol ; 85(8): 1829-1840, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31077427

RESUMO

AIMS: To provide insights into the clinical development pathway for fixed-dose combinations (FDCs), to consider strategies, and to elucidate the path to approval by assessing the body of evidence, as summarized in the European Public Assessment Reports. METHODS: The main resource was the European Public Assessment Reports for 36 FDCs, which included 239 clinical trials with 157 514 patients. The analyses focused on how prior knowledge of the active substances or combination, use of pharmacokinetic-pharmacodynamic modelling, and clinical trial design choice impact the size and strategy of the clinical development programme. RESULTS: FDC products primarily comprised 2 previously approved components (21/36, 71%) and had only 1 approved combination (21/36, 71%). Utilizing previously approved active substances resulted in fewer clinical trials, arms and patients, but FDC doses studied in the clinical development programme. Furthermore, dose-finding trials were performed for less than half of FDCs consisting of 2 previously approved active substances. The standard approach to demonstrate contribution of active substances was through a factorial or single combination study. Finally, the use of pharmacokinetic modelling showed a significant decrease in the number of FDC doses studied. CONCLUSIONS: The field of FDCs seems to be on the rise, utilizing new molecular entities, prior knowledge and re-profiling drugs. However, a way to move FDC development forward might be through new regulatory and scientific paradigms, in which it is encouraged to utilize model-based approaches to develop FDCs with multiple dose levels and dose ratios for exposure-based treatment that will enable personalization.


Assuntos
Combinação de Medicamentos , Desenvolvimento de Medicamentos/métodos , União Europeia , Ensaios Clínicos como Assunto/normas , Relação Dose-Resposta a Droga , Aprovação de Drogas , Desenvolvimento de Medicamentos/normas , Modelos Biológicos , Projetos de Pesquisa/normas
3.
Milbank Q ; 95(2): 261-290, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28589600

RESUMO

Policy Points: Randomized trials-the gold standard of evaluating effectiveness-constitute a small minority of existing evidence on agents given accelerated approval. One-third of randomized trials are in therapeutic areas outside of FDA approval and less than half evaluate the therapeutic benefits of these agents but use them instead as common backbone treatments. Agents receiving accelerated approval are often tested concurrently in several therapeutic areas. For most agents, no substantial time lag is apparent between the average start dates of randomized trials evaluating their effectiveness and those using them as part of background therapies. There appears to be a tendency for therapeutic agents receiving accelerated approval to quickly become an integral component of standard treatment, despite potential shortcomings in their evidence base. CONTEXT: Therapeutic agents treating serious conditions are eligible for Food and Drug Administration (FDA) accelerated approval. The clinical evidence accrued on agents receiving accelerated approval has not been systematically evaluated. Our objective was to assess the timing and characteristics of available studies. METHODS: We first identified clinical studies of novel therapeutic agents receiving accelerated approval. We then (1) categorized those studies as randomized or nonrandomized, (2) explored whether they evaluated the FDA-approved indications, and (3) documented the available treatment comparisons. We also meta-analyzed the difference in start times between randomized studies that (1) did or did not evaluate approved indications and (2) were or were not designed to evaluate the agent's effectiveness. FINDINGS: In total, 37 novel therapeutic agents received accelerated approval between 2000 and 2013. Our search of ClinicalTrials.gov identified 7,757 studies, which included 1,258,315 participants. Only one-third of identified studies were randomized controlled trials. Of 1,631 randomized trials with advanced recruitment status, 906 were conducted in therapeutic areas for which agents received initial accelerated approval, 202 were in supplemental indications, and 523 were outside approved indications. Only 411 out of 906 (45.4%) trials were designed to test the effectiveness of agents that received accelerated approval ("evaluation" trials); others used these agents as common background treatment in both arms ("background" trials). There was no detectable lag between average start times of trials conducted within and outside initially approved indications. Evaluation trials started on average 1.52 years (95% CI: 0.87 to 2.17) earlier than background trials. CONCLUSIONS: Cumulative evidence on agents with accelerated approvals has major limitations. Most clinical studies including these agents are small and nonrandomized, and about a third are conducted in unapproved areas, typically concurrently with those conducted in approved areas. Most randomized trials including these therapeutic agents are not designed to directly evaluate their clinical benefits but to incorporate them as standard treatment.


Assuntos
Aprovação de Drogas/organização & administração , Medicina Baseada em Evidências , United States Food and Drug Administration , Controle de Medicamentos e Entorpecentes , Metanálise como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Tempo , Estados Unidos
4.
Br J Clin Pharmacol ; 83(12): 2615-2625, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28779556

RESUMO

The terms 'licensed', 'unlicensed', and 'off-label', often used in relation to marketing and prescribing medicinal products, may confuse UK prescribers. To market a medicinal product in the UK requires a Marketing Authorization ('product licence') for specified indications under specified conditions, regulated by the Medicines and Healthcare products Regulatory Agency (MHRA). The Marketing Authorization includes the product's agreed terms of use (the 'label'), described in the Summary of Product Characteristics (SmPC). Prescribing a licensed product outside those terms is called 'off-label' prescribing. Products for which no-one holds a UK Marketing Authorization are unlicensed. Prescribers can prescribe authorized products according to the conditions described in the SmPC ('on-label') or outside those conditions ('off-label'). They can also prescribe unauthorized products, even if they are unlicensed in the UK, if they are licensed elsewhere or if they have been manufactured in the UK by a licensed manufacturer as a 'special'. The complexities of this system can be understood by considering the status of the manufacturer of the product, the company that markets it (which may or may not be the same), the product itself, and its modes of use, and by emphasizing the word 'authorized'. If a Marketing Authorization is granted to the supplier of a product, it will specify the authorized modes of use; the product will be prescribable as authorized (i.e. 'on-label') or in other modes of use, which will all be off-label. Unlicensed products with no authorized modes of use can be regarded as 'unauthorized products'. All 'specials' can be regarded as authorized products lacking authorized modes of use.


Assuntos
Rotulagem de Medicamentos/classificação , Controle de Medicamentos e Entorpecentes , Licenciamento/classificação , Uso Off-Label/classificação , Preparações Farmacêuticas/classificação , Terminologia como Assunto , Rotulagem de Medicamentos/legislação & jurisprudência , Rotulagem de Medicamentos/normas , Controle de Medicamentos e Entorpecentes/legislação & jurisprudência , Humanos , Licenciamento/legislação & jurisprudência , Licenciamento/normas , Uso Off-Label/legislação & jurisprudência , Uso Off-Label/normas , Preparações Farmacêuticas/normas , Guias de Prática Clínica como Assunto , Reino Unido
5.
Clin Infect Dis ; 61 Suppl 5: S428-33, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26553671

RESUMO

BACKGROUND: Through its normative and public health leadership roles, the World Health Organization (WHO) plays a key role in the availability of vaccine products in low-and middle-income countries. The recent introduction of a new group A meningococcal conjugate vaccine, PsA-TT (MenAfriVac), in Africa exemplifies this process. WHO requires that any new vaccine to be introduced in countries for public health reasons and supplied through United Nations centralized mechanisms be licensed by the national regulatory agency (NRA) in the producing country, then prequalified and given a marketing authorization in the user countries. METHODS: PsA-TT was manufactured by the Serum Institute of India, Ltd (SIIL), which submitted a license application in April 2009 to the Drug Controller General of India (DCGI), the Indian NRA responsible for licensing vaccines. WHO encouraged the DCGI to establish a collaboration with Health Canada's Centre for Vaccine Evaluation for the review. Through this collaborative effort, registration was facilitated and in December 2009 an export license was granted to SIIL, which subsequently submitted an application for WHO prequalification. RESULTS: Given the importance of the vaccine, WHO "fast tracked" the prequalification review, and after a detailed review and site visit, WHO prequalification was granted to PsA-TT in June 2010. Country use of the new vaccine could not occur until the vaccine was a registered product in each country seeking its use. WHO facilitated country reviews by conducting regulatory training exercises (in French and English) for country NRA staff, which used the PsA-TT registration as a case study. CONCLUSIONS: PsA-TT was gradually registered in African countries as vaccine introduction proceeded. The regulatory pathway for this new group A meningococcal conjugate vaccine proved to be a useful training opportunity both in India and Africa, because the availability of the vaccine was a high African public health priority, as well as for WHO as a case study to facilitate registration of vaccines based on reliance on other regulatory bodies.


Assuntos
Transmissão de Doença Infecciosa/prevenção & controle , Aprovação de Drogas , Meningite Meningocócica/prevenção & controle , Vacinas Meningocócicas/isolamento & purificação , Vacinas Meningocócicas/normas , Tecnologia Farmacêutica/normas , Vacinação/normas , África , Canadá , Humanos , Índia , Cooperação Internacional , Organização Mundial da Saúde
6.
Regul Toxicol Pharmacol ; 71(2): 251-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25591547

RESUMO

INTRODUCTION: Different causes of drug shortages can be linked to the pharmaceutical legal framework, such as: parallel trade, quality requirements, economic decisions to suspend or cease production, etc. However until now no in-depth study of the different regulations affecting drug shortages is available. The aim of this paper is to provide an analysis of relevant legal and regulatory measures in the European pharmaceutical framework which influence drug shortages. METHODS: Different European and national legislations governing human medicinal products were analyzed (e.g. Directive 2001/83/EC and Directive 2011/62/EU), supplemented with literature studies. RESULTS: For patented drugs, external price referencing may encompass the largest impact on drug shortages. For generic medicines, internal or external reference pricing, tendering as well as price capping may affect drug shortages. Manufacturing/quality requirements also contribute to drug shortages, since non-compliance leads to recalls. The influence of parallel trade on drug shortages is still rather disputable. CONCLUSION: Price and quality regulations are both important causes of drug shortages or drug unavailability. It can be concluded that there is room for improvement in the pharmaceutical legal framework within the lines drawn by the EU to mitigate drug shortages.


Assuntos
Indústria Farmacêutica/legislação & jurisprudência , União Europeia , Acessibilidade aos Serviços de Saúde/legislação & jurisprudência , Preparações Farmacêuticas , Custos e Análise de Custo/economia , Custos e Análise de Custo/legislação & jurisprudência , Indústria Farmacêutica/economia , Medicamentos Genéricos/economia , Acessibilidade aos Serviços de Saúde/economia , Humanos , Preparações Farmacêuticas/economia , Mecanismo de Reembolso/legislação & jurisprudência , Reino Unido , Estados Unidos
7.
Health Aff Sch ; 2(8): qxae095, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39161949

RESUMO

Regulatory agencies worldwide have taken significant steps to expedite approval and market authorization of medicines based on their potential to address areas of significant unmet medical need and severe disease burden. However, initial approval of such medicines is often accompanied by limited evidence of benefit, posing a conundrum for payers and health systems who may desire greater certainty of their value. This paper describes a system of "accelerated access" to manage these tensions and coordinate activities across stakeholders, based on discussions held at a multi-stakeholder convening in June 2023. We focus on 6 core, near-term actions that can be taken to improve the current system: clarifying criteria for expedited regulatory approval, enhancing stakeholder coordination, creating expedited pathways in payer and health technology assessment settings, developing joint regulatory/payer/health technology assessment guidance on study design and data needs, linking pricing policy to data uncertainty, and improving patient and public understanding of the processes involved as well as the risks and benefits of the relevant medicines. Many of these actions will require additional resources and personnel, and some will necessitate unprecedented levels of coordination. Nevertheless, each action is designed to work with minimal adjustments to the current system rather than demanding an entirely new approach.

8.
Ther Innov Regul Sci ; 57(5): 987-996, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37211582

RESUMO

BACKGROUND: Cognizant of indispensable role as important health intervention tools, the global medical devices industry continues to bring new medical devices with varying degrees of technologies and complexities. Ensuring the safety, good performance and timely access of them have become challenging for regulatory authorities, particularly for developing countries including Ethiopia. In Ethiopia, the role of the regulatory authority is complicated further because of the lack of specific policies. Medical devices regulation is still being dealt under drug policy. OBJECTIVES: This study was aimed to assess the regulatory approval processes of medical devices in Ethiopia. METHODS: A mixed sequential explanatory study design was employed. Quantitative data were collected using a structured self-administered questionnaire and standard checklist; qualitative data were collected through in-depth interviews using a semi-structured guide. RESULTS: Retrospective trend analysis (2015 to 2018) indicated that 3,804 medical devices were registered in Ethiopia. Findings from the quantitative study indicated that 73.3% of regulatory experts had commendable knowledge on the medical devices regulatory system. However, gaps were identified in inspection and auditing (63.8%), practically understanding the system and procedures (24.3%), and having competencies in executing the critical core functions (6.9%). The top five challenges reported include (i) lack of capacity to assess dossiers (80.8%); (ii) lack of effective legislation (64.1%); (iii) provision of ambiguous feedback on deficiencies after dossier evaluations and delay in their communication (63.9%); (iv) long waiting time for approval (61.1%); and (v) lack of experienced and qualified staff (55.7%). In addition, the absence of a specific policy for medical device regulation presents a great hurdle. CONCLUSION: Basic functional systems and procedures for the regulation of medical devices in Ethiopia are present. However, there are still gaps that are impeding effective regulation of medical devices especially for those with advanced features and complex-monitoring modalities.


Assuntos
Política Pública , Humanos , Etiópia , Estudos Retrospectivos , Inquéritos e Questionários
9.
Eur J Pharm Sci ; 173: 106181, 2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35381330

RESUMO

There is a growing body of research about subcutaneously administered biologics, emphasizing the need for optimized bioavailability predictions. It is important to inform both translational and in silico models with properties of the drug products and compounds. However, the pharmaceutical, therapeutic and physicochemical properties of market authorized drug products for subcutaneous administration are currently not collated in the public domain. We provide an overview of subcutaneous administered drug products for humans and animals market authorized in EU, Canada, and the US. Data on the drug products were collected from the respective authorities, i.e. European Medicines Agency, Health Canada, and U.S. Food and Drug Administration. Physicochemical properties of active substances were gathered from DrugBank. Human drug products were often indicated for treatment of diabetes and anemia. EU veterinary drug products were often immunologicals. Canadian and US veterinary drug products often acted as antiinfectives for systemic use, on the genito-urinary system or as sex hormones. The final dataset with >1700 subcutaneous drug products is provided. In EU drug products, the majority of active substances were biologics. In the US, drug products most often contained small molecules. Solutions, emulsions and suspensions were the most common dosage forms. A minority of subcutaneous drug products were also registered for intramuscular or intravenous administration. The analysis presented here could aid further research, exploring formulation properties, prescription or sales of market authorized SC drug products and development of inclusive in silico models.


Assuntos
Produtos Biológicos , Drogas Veterinárias , Animais , Canadá , Preparações Farmacêuticas , Estados Unidos , United States Food and Drug Administration
10.
Therapie ; 77(3): 329-338, 2022.
Artigo em Francês | MEDLINE | ID: mdl-35012758

RESUMO

Analysis of off-label prescriptions of medicines in hospital in adult patients and study of feasibility of their detection by use of international disease classification, 10th version (IDC-10 codes). CONTEXT: In order to improve the appropriate use of medicines, a method of detection of off label prescriptions, especially in hospitalised patients, should be available. STUDY OBJECTIVES: Evaluate the performance of the detection of off-label prescriptions in hospitalised patients by use of IDC-10 codes. METHODS: Data prescriptions (excluding those directly taken in charge by the national health care system), clinical history and biological results were extracted from Assistance publique des Hôpitaux de Paris (AP-HP) data-warehouse for 108 in-hospital adults patients' journeys. An adjudication committee established the classification reference for the appropriate or off label drug prescriptions status after analysis of medical information for each patient. IDC-10 codification that is performed after every hospitalisation was crossed with those IDC-10 codes that were to be expected corresponding to the marketing authorisation labelling (section 4.1 of specifications of product characteristics [SPC]). Results of IDC-10 coding were compared to the reference for off label use identification. RESULTS: Out of the 1131 analysed prescriptions, 44 (3.9%) were classified as off label by the adjudication committee. Sensitivity of detection by IDC-10 coding was 87 (95% CI [0.73-0.96]) to 92% (95% CI [0.79-0.98]) and specificity 25 (95% CI [0.22-0.27]) to 41% (95% CI [0.38-0.44]) according to the number of characters of ICD-10 that could be used. CONCLUSIONS: Incidence of in-hospital off label use of drugs (restricted to within drug related groups prescriptions) appeared relatively low (3.9%). Its semi-automatic detection by IDC-10 coding appears feasible with a good sensitivity but a low specificity. Such method could be further assessed as a first step detection focusing on one pharmacological class or on one pathologic condition.


Assuntos
Uso Off-Label , Prescrições , Adulto , Atenção à Saúde , Estudos de Viabilidade , Hospitais , Humanos
11.
Front Public Health ; 9: 754482, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34900902

RESUMO

Some advanced therapy medicinal products (ATMPs) hold great promises for life-threatening diseases with high unmet needs. However, ATMPs are also associated with significant challenges in market access, which necessitates the joint efforts between all relevant stakeholders to navigate. In this review, we will elaborate on the importance of collaborations and harmonization across different stakeholders, to expedite the market access of promising ATMPs. Manufacturers of ATMPs should proactively establish collaborations with other stakeholders throughout the whole lifecycle of ATMPs, from early research to post-market activities. This covered engagements with (1) external developers (i.e., not-for-profit organizations and commercial players) to obtain complementary knowledge, technology, or infrastructures, (2) patient groups and healthcare providers to highlight their roles as active contributors, and (3) decision-makers, such as regulators, health technology assessment (HTA) agencies, and payers, to communicate the uncertainties in evidence package, where parallel consultation will be a powerful strategy. Harmonization between decision-makers is desired at (1) regulatory level, in terms of strengthening the international standardization of regulatory framework to minimize discrepancies in evidence requirements for market authorization, and (2) HTA level, in terms of enhancing alignments between regional and national HTA agencies to narrow inequity in patient access, and cross-border HTA cooperation to improve the quality and efficiency of HTA process. In conclusion, manufacturers and decision-makers shared the common goals to safeguard timely patient access to ATMPs. Collaboration and harmonization will be increasingly leveraged to enable the value delivery of ATMPs to all stakeholders.


Assuntos
Atenção à Saúde , Avaliação da Tecnologia Biomédica , Humanos , Incerteza
12.
Health Policy ; 124(7): 701-713, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32499078

RESUMO

BACKGROUND: A small number of regenerative medicines (RMs) have received market authorization (MA) worldwide, relative to the large number of clinical trials currently being conducted. Regulatory issues constitute one major challenge for the MA of RMs. OBJECTIVE: This study aimed to systematically review the regulation of RMs internationally, to identify the regulatory pathways for approved RMs, and to detail expedited programs to stimulate MA process. METHODS: Official websites of regulatory authorities in 9 countries (United States (US), Japan, South Korea, Australia, Canada, New Zealand, Singapore, China, and India) and the European Union (EU) were systematically browsed, and was complemented by a systematic literature review in Medline and Embase database. RESULTS: Specific RM legislation/frameworks were available in the EU, US, Japan, South Korea and Australia. A risk-based approach exempting eligible RMs from MA regulations were adopted in the EU and 6 countries. All investigated regions have established accelerated review or approval programs to facilitate the MA of RMs. 55 RMs have received MA in 9 countries and the EU. Twenty-three RMs received Priority Medicine designation, 32 RMs received Regenerative Medicine Advanced Therapy designation, and 11 RMs received SAKIGAKE (fore-runner initiative) designation. CONCLUSION: Regulators have adopted proactive strategies to facilitate RM approval. However, addressing the discrepancies in regulatory requirements internationally remains challenging.


Assuntos
Políticas , Medicina Regenerativa , Austrália , Canadá , China , Humanos , Japão , Nova Zelândia , República da Coreia , Singapura , Estados Unidos
13.
Therapie ; 75(2): 141-147, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32247678

RESUMO

Clinical development for orphan drugs is extremely demanding but fascinating. There is no single aspect that is really specific to it but instead it gathers most of the hurdles: design, outcomes, recruitment, ethics, cost, probability and predictability for success. To overcome these difficulties, there has to be a great collaboration between academic centers, small and large pharma companies, patients' representatives as well as health authorities to provide support and innovative approaches. The ultimate goal is to give access to patients with unmet medical needs to drugs with a favorable benefit-risk ratio. We review and discuss here the pillars for a successful clinical development for orphan drugs.


Assuntos
Produção de Droga sem Interesse Comercial , Doenças Raras/tratamento farmacológico , Ensaios Clínicos como Assunto , Análise Custo-Benefício , Aprovação de Drogas , Humanos , Produção de Droga sem Interesse Comercial/economia
14.
J Pharm Policy Pract ; 13: 45, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32695427

RESUMO

Drug repositioning is the scientific strategy of investigating existing drugs for additional clinical indications. The advantages of drug repositioning are that it benefits patients and that it adds new indications to existing drugs for lower costs compared to de novo drug development. Clinical research groups recognizing efficacy of these "old" drugs for a new indications often face an uphill struggle due to a lack of funding and support because of poor structural and regulatory support for clinical drug development. The current framework for drug repositioning allows "venture capital" companies to abuse loopholes in the legislation to gain long-term market authorization among with excessive high pricing. A new regulatory framework is needed to prevent abuse of the legislation and promote clinical investigator-driven drug repositioning. The COVID-19 pandemic has boosted funding and regulatory support for drug repositioning. The lessons learned from the COVID-19 pandemic should be implemented in a new clear blueprint for drug repositioning. This blueprint should guide clinicians through legislation for drug repositioning in the EU. This review summarizes the routes for registration and discusses the current state of drug repositioning in Europe.

15.
Ticks Tick Borne Dis ; 10(1): 63-71, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30197268

RESUMO

This study assessed cost-effectiveness of a potential anti-tick vaccine that would protect against both Lyme borreliosis (LB) and tick-borne encephalitis (TBE) in a highly endemic setting of Slovenia. A Markov model was developed to estimate cost-effectiveness of a vaccine with potential combined protection against LB and TBE from the societal perspective. The model expressed time in annual cycles, followed a target population through their lifetime, and applied an annual discounting of 3%. A target population entered the model in a susceptible state, with time dependent probabilities to acquire LB/TBE. Disease manifestations were either resolved within one cycle, or a patient developed LB/TBE sequelae. The vaccination consisted of initial immunization and one revaccination. Estimates of LB/TBE direct and indirect costs, and data on natural course of LB/TBE were obtained from Slovenian databases. Effectiveness of the vaccine with potential combined protection against LB/TBE was derived from studies on existing TBE and LB vaccines, while utility estimates were collected from various literature sources. A vaccine with potential combined protection against LB/TBE was predicted to have an incremental cost of €771,300 per 10,000 vaccinated persons, an incremental utility of 17QALYs and a base-case incremental cost-effectiveness ratio (ICER) of 46,061€/QALY. Vaccine cost, effectiveness and discount rates were identified as the most influential model parameters. A wholesale price for a vaccine shot of €9.13 would lead to cost savings followed by health gains for the vaccination strategy. The base-case ICER was below commonly accepted thresholds of cost-effectiveness, indicating that a combined LB/TBE vaccine might be a cost-effective option in Slovenia. With early Health Technology Assessment becoming increasingly important, this analysis still represents a rare example of cost-effectiveness assessment prior to market authorisation. Although obviously in such a situation some key parameters are unknown, our model sets up a tool to analyse pharmacoeconomic criteria that can help development of a cost-effective health technology, in this case a combined tick-borne diseases vaccine.


Assuntos
Vacinas Bacterianas/economia , Encefalite Transmitida por Carrapatos/prevenção & controle , Doença de Lyme/prevenção & controle , Vacinas Virais/economia , Análise Custo-Benefício , Eslovênia
16.
Front Pharmacol ; 10: 1009, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31619989

RESUMO

Objectives: Patient preference information (PPI) is gaining recognition among the pharmaceutical industry, regulatory authorities, and health technology assessment (HTA) bodies/payers for use in assessments and decision-making along the medical product lifecycle (MPLC). This study aimed to identify factors and situations that influence the value of patient preference studies (PPS) in decision-making along the MPLC according to different stakeholders. Methods: Semi-structured interviews (n = 143) were conducted with six different stakeholder groups (physicians, academics, industry representatives, regulators, HTA/payer representatives, and a combined group of patients, caregivers, and patient representatives) from seven European countries (the United Kingdom, Sweden, Italy, Romania, Germany, France, and the Netherlands) and the United States. Framework analysis was performed using NVivo 11 software. Results: Fifteen factors affecting the value of PPS in the MPLC were identified. These are related to: study organization (expertise, financial resources, study duration, ethics and good practices, patient centeredness), study design (examining patient and/or other preferences, ensuring representativeness, matching method to research question, matching method to MPLC stage, validity and reliability, cognitive burden, patient education, attribute development), and study conduct (patients' ability/willingness to participate and preference heterogeneity). Three types of situations affecting the use of PPS results were identified (stakeholder acceptance, market situations, and clinical situations). Conclusion: The factors and situation types affecting the value of PPS, as identified in this study, need to be considered when designing and conducting PPS in order to promote the integration of PPI into decision-making along the MPLC.

17.
Health Policy ; 122(6): 599-606, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29605527

RESUMO

The early benefit assessment of pharmaceuticals in Germany and their preceding market authorization pursue different objectives. This is reflected by the inclusion of varying confirmatory endpoints within the evaluation of oncology drugs in early benefit assessment versus market authorization, with both relying on the same evidence. Data from assessments up to July 2015 are used to estimate the impact of explorative in comparison to confirmatory endpoints on market authorization and early benefit assessment by contrasting the benefit-risk ratio of EMA and the benefit-harm balance of the HTA jurisdiction. Agreement between market authorization and early benefit assessment is examined by Cohen's kappa (k). 21 of 41 assessments were considered in the analysis. Market authorization is more confirmatory than early benefit assessment because it includes a higher proportion of primary endpoints. The latter implies a primary endpoint to be relevant for the benefit-harm balance in only 67% of cases (0.078). Explorative mortality endpoints reached the highest agreement regarding the mutual consideration for the risk-benefit ratio and the benefit-harm balance (0.000). For explorative morbidity endpoints (-0.600), quality of life (-0.600) and side effects (-0.949) no agreement is ascertainable. To warrant a broader confirmatory basis for decisions supported by HTA, closer inter-institutional cooperation of approval authorities and HTA jurisdictions by means of reliable joint advice for manufacturers regarding endpoint definition would be favorable.


Assuntos
Antineoplásicos/provisão & distribuição , Tomada de Decisões , Aprovação de Drogas , Medição de Risco , Avaliação da Tecnologia Biomédica/métodos , Alemanha , Humanos , Qualidade de Vida
18.
J Tissue Eng Regen Med ; 12(7): 1579-1593, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29702746

RESUMO

Modern regenerative medicine research has expanded well past the development of traditional drugs and medical devices with many promising new therapies encompassing an increasingly diverse range of substances, notably cell-based therapies. These substantial recent developments and the progress in the health care and therapeutics fields necessitate a new regulatory framework agile enough to accommodate these unique therapies and acknowledge their differences with traditional pharmaceuticals. In the United States, recent proposed changes in the regulatory framework for autologous human cells, tissues, and cellular and tissue-based products (HCT/Ps) and their perceived risk-benefit analysis for patients remain controversial in the scientific field. To provide perspective on of the current status of the most recent attempts to redefine and conceptualize these changes in the United States, we will examine 4 draft guidance documents implemented by the Food and Drug Administration in interpreting relevant concepts and terminology pertaining to HCT/Ps: the Bipartisan Policy Center think tank report, "Advancing Regenerative Cellular Therapy: Medical Innovation for Healthier Americans," the proposed REGROW Act for HCT/Ps, and the current 24 Food and Drug Administration-approved HCT/Ps and related products in the United States.


Assuntos
Produtos Biológicos/normas , Terapia Baseada em Transplante de Células e Tecidos/normas , Aprovação de Drogas/legislação & jurisprudência , United States Food and Drug Administration , Produtos Biológicos/uso terapêutico , Humanos , Estados Unidos
19.
Eur J Pharmacol ; 719(1-3): 9-15, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23891846

RESUMO

Regulatory science as a discipline has evolved over the past years with the object to boost and promote scientific rationale behind benefit/risk and decision making by regulatory authorities. The European Medicines Agency, EMA, the Food and Drug Administration, FDA, and the Japanese Pharmaceutical and Medical Devices Agency, PMDA, highlighted in their distinct ways the importance of regulatory science as a basis of good quality assessment in their strategic plans. The Medicines Evaluation Board, MEB, states: 'regulatory science is the science of developing and validating new standards and tools to evaluate and assess the benefit/risk of medicinal products, facilitating sound and transparent regulatory decision making'. Through analysis of regulatory frameworks itself and their effectiveness, however, regulatory science can also advance knowledge of these systems in general. The comprehensive guidance that is issued to complete an application dossier for regulatory product approval has seldomly been scrutinized for its efficiency. Since it is the task of regulatory authorities to protect and promote public health, it is understood that they take a cautious approach in regulating drugs prior to market access. In general, the authorities are among the first to be blamed if dangerous or useless drugs were allowed to the market. Yet, building a regulatory framework that is not challenged continuously in terms of deliverables for public health and cost-effectiveness, might be counterproductive in the end. Regulatory science and research can help understand how and why regulatory decisions are made, and where renewed discussions may be warranted. The MEB supports regulatory science as an R&D activity to fuel primary regulatory processes on product evaluation and vigilance, but also invests in a 'looking into the mirror' approach. Along the line of the drug life-cycle, publicly available data are reviewed and their regulatory impact highlighted. If made explicit, regulatory research can open the door to evidence based regulatory practice and serve the regulator's contribution to innovative drug licensing today.


Assuntos
Preparações Farmacêuticas , Controle Social Formal/métodos , Animais , Prática Clínica Baseada em Evidências , Órgãos Governamentais , Humanos , Segurança
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA