Differences
This shows you the differences between two versions of the page.
| Both sides previous revisionPrevious revisionNext revision | Previous revisionBoth sides next revision | ||
| european_medicines_agency [2022/08/30 18:23] pamela | european_medicines_agency [2023/09/16 18:33] (current) liam [EMA COVID-19 Steering Group] | ||
|---|---|---|---|
| Line 1: | Line 1: | ||
| - | ===== European Medicines Agency | + | ====== European Medicines Agency |
| - | {{::ena_european_medicines_agency.png?600 |}} | + | {{ ::european_medicines_agency.png?200|}} |
| - | ==== History ==== | + | The **European Medicines Agency |
| - | + | ||
| - | The European Medicines Agency is an agency of the European Union in charge of the evaluation and supervision of medicinal products. | + | |
| Prior to 2004, it was known as the European Agency for the Evaluation of Medicinal Products or European Medicines Evaluation Agency. | Prior to 2004, it was known as the European Agency for the Evaluation of Medicinal Products or European Medicines Evaluation Agency. | ||
| - | The European Medicines Agency (EMA) is a decentralized agency of the [[:European Union]] (EU) whose goal is to promote and protect human and animal health. | + | ==== Regulatory information on veterinary medicines ==== |
| - | The EMA is the European Union’s | + | ===== Partners & networks ==== |
| - | ((https:// | + | {{ :: |
| + | The European Medicines Agency | ||
| + | ==== Pharmaceutical industry ==== | ||
| - | Overview | + | On this page, you will find information on the Agency’s activities that are most relevant to pharmaceutical industry, including news, and events. You can contribute to the Agency’s work by responding to public consultations. Learn more about how the pharmaceutical industry is actively involved in the work of the Agency. (()) |
| - | Role: EMA guarantees the scientific evaluation, supervision & safety monitoring of human & veterinary medicines in the EU. | ||
| - | Executive Director: Emer Cooke | ||
| - | Established in: 1995 | ||
| - | Number of staff: 897 | ||
| - | Location: Amsterdam (the Netherlands) | ||
| - | Website: EMA | ||
| - | The European Medicines Agency (EMA) protects and promotes human and animal health by evaluating and monitoring medicines within the European Union (EU) and the European Economic Area (EEA). | + | ==== EU Innovation Network ==== |
| - | What it does | + | |
| - | The Agency' | + | Innovation offices in national regulatory agencies have been working informally with the EMA's [[:Innovation Task Force]] (ITF) on matters relating to emerging therapies and technologies since 2011. In 2015, EMA and the EU national competent authorities (NCAs) strengthened their collaboration |
| - | The Agency fulfils its responsibilities by: | + | EMA and the HMAs adopted the mandate of the EU-Innovation Network in October 2016 |
| - | * facilitating the development | + | === Mandate |
| - | | + | {{ :: |
| - | | + | 2014 - The [[: |
| - | * | + | |
| - | Who benefits | + | The workshop provided an opportunity to discuss expectations with different stakeholders such as members of the European Medicines Agency (EMA) Healthcare Professionals' |
| - | The EMA's work benefits: | + | The aim of the network is to make the regulatory support for medicines developers currently available at national and EU levels more visible and attractive to innovators ((https:// |
| - | * patients | + | === Authorized Stakeholders === |
| - | * | + | |
| - | * | + | |
| - | * | + | |
| - | * | + | |
| - | * | + | |
| - | Through its scientific guidelines, scientific advice programme and incentives, it facilitates research into new medicines | + | List of eligible industry stakeholder organisations (as of 16 May 2018) With reference to the Criteria to be fulfilled by industry stakeholder organisations involved in EMA activities, (EMA/ |
| + | ==== Pharmacovigilance ==== | ||
| - | ==== Regulatory information on human medicines ==== | + | Pharmacovigilance is the science and activities relating to the detection, assessment, understanding and prevention of adverse effects or any other medicine-related problem. The European Medicines Agency (EMA) coordinates the European Union (EU) pharmacovigilance system and operates services and processes to support pharmacovigilance in the EU. |
| - | **Guideline on strategies to identify and mitigate risks for first-in-human | + | Before a medicine is authorised |
| - | {{ :: | + | |
| + | After authorisation the medicine may be used in a large number of patients, for a long period of time and with other medicines. Certain side effects may emerge in such circumstances. | ||
| - | Executive summary | ||
| - | This is the first revision of the ‘Guideline on strategies to identify and mitigate risks for first-in-human clinical trials with investigational medicinal products’. It extends the existing EU guidance to address first-in-human (FIH) and early phase clinical trials (CTs) with integrated protocols. | ||
| + | It is therefore essential that the safety of all medicines is monitored throughout their use in healthcare practice. | ||
| + | {{:: | ||
| - | The revision is intended | + | **EU law therefore requires each marketing authorisation holder, national competent authority and EMA to operate a pharmacovigilance system.** The overall EU pharmacovigilance system operates through cooperation between the EU Member States, EMA and the [[:European Commission]]. In some Member States, regional centres |
| + | EMA's [[: | ||
| - | 1. Introduction (background) | + | EMA supports |
| - | The purpose of FIH trials is to evaluate an investigational medicinal product (IMP) in humans for the first time, to study the human pharmacology, | + | |
| - | Consequently, | + | The Agency |
| - | As IMPs are widely different | + | EudraVigilance is a single repository for reports of suspected adverse reactions seen in healthcare practice |
| - | 6.1. **Demonstration of relevance of the animal model** | + | The PRAC evaluates safety signals from [[: |
| - | The relevance of the selected animal model should be justified in the CT application. The demonstration of relevance of the animal model(s) may include comparison with humans | + | |
| - | 6.2. **Nature of the target** | + | EMA publishes |
| - | Beyond the mode of action, the nature of the target itself can impact on the potential risk inherent to the initial administrations to humans. Experimental and/or literature-data should be taken into account when defining | + | |
| - | 6.3. **Pharmacodynamics** | + | Users can view the total number |
| - | Primary PD studies should address | + | |
| - | derived material, and in vivo using animal models, as relevant. | + | |
| - | These studies might include target interactions preferably linked to functional response, e.g. receptor binding and occupancy, inhibition of | + | Reports for drug substances used in nationally authorised medicines are also available since October 2014. |
| - | enzymes, cellular response consequent to the interaction with the target, duration and (ir)reversibility of effect, dose-response relationships and physiological turn-over of the target. When defining the degree of uncertainty associated with the mode of action or effects, aspects to be | + | |
| - | considered may include | + | |
| - | 6.4.** Pharmaco- and toxicokinetics** | + | The Pharmacovigilance Risk Assessment Committee |
| - | PK and toxicokinetic | + | |
| - | Systemic exposures at pharmacodynamically active doses in the relevant animal models should be determined and considered especially when PD effects are suspected to contribute to potential safety concerns. Possible polymorphisms e.g. in metabolic enzymes should be taken into account. | + | The PRAC recommendation may include one or a combination of conclusions, |
| - | 6.5. **Safety pharmacology** | + | |
| - | Standard core battery | + | |
| + | | ||
| + | | ||
| + | | ||
| + | * a post-authorisation safety study conducted by the marketing authorisation holder; | ||
| + | * Need for regulatory action, such as: | ||
| + | * | ||
| + | * a referral procedure; | ||
| + | * | ||
| - | 6.6. **Toxicology** | ||
| - | The toxicology programme should be designed taking the characteristics of the IMP and the relevant ICH guidelines S6(R1), S9 and M3(R2) into account. | ||
| - | Toxicity can be the result of exaggerated pharmacological actions. These types of effects should not be ignored when establishing a safe starting dose for humans and the exposures, at which these toxicities are observed, should be considered for the definition of the dose escalation range to be investigated in humans. Primary and secondary PD data can support the generation of mechanistic hypotheses regarding the toxicities seen in vivo and help in the interpretation of the human relevance of these findings. | ||
| - | An evaluation as to whether the target organs identified in the non-clinical studies warrant particular monitoring in the CT should be undertaken. Serious toxicity should lead to a more cautious approach when setting doses and applying risk mitigation strategies in the clinical setting. When serious toxicity or mortality is observed, these effects may require follow up studies to determine the cause of death or the mechanism of toxicity if this has not been possible to clarify within the studies undertaken, and if this information is relevant to the clinical trial design or safety monitoring plan. | ||
| - | 8.2.9. **Stopping rules** | ||
| - | {{ :: | ||
| - | The protocol should define unambiguous stopping rules which result in an immediate stop to dosing. It should further be specified in the rule if the stop is a final end of dosing or a temporary halt. Restart is possible without a substantial amendment if review leads to a conclusion which is fully within predefined conditions for the relevant stopping criterion. | ||
| - | Any submitted substantial amendment should include a rationale for the proposed dosing and for the continuation of the trial and details of any adjustments to the protocol including additional safety monitoring, if applicable. ((https:// | + | ==== Emergency Use ==== |
| + | The MHRA has been assured that acceptable standards of Good Manufacturing Practice (GMP) are in place for this product at all sites responsible for the manufacture, | ||
| - | ==== Regulatory information on veterinary medicines ==== | + | A Risk Management Plan (RMP) and a summary of the pharmacovigilance system have been provided with this application and are satisfactory. |
| - | ===== Partners & networks ==== | + | This batch, and any future batches, of COVID-19 mRNA Vaccine BNT162b2 are subject to Qualified Person (QP) certification and batch evaluation by an independent control laboratory before the vaccine is released into the UK. |
| + | The [[:COVID-19 Vaccine Benefit Risk Expert Working Group]] (Vaccine BR EWG) have met several times to review and discuss the quality, safety and efficacy aspects in relation to batches of COVID-19 mRNA Vaccine [[: | ||
| - | ==== Pharmaceutical industry ==== | + | The Vaccine BR EWG gave advice to the Commission of Human Medicines (CHM) on 11th September 2020, 8th October 2020, 27th October 2020, 28th November 2020 and 30th November 2020, regarding the requirements for authorisation for the temporary supply of COVID-19 [[:mRNA Vaccine]] BNT162b2. The requirements for quality, safety and efficacy were considered, taking into account the urgent public health need and risk to life, the pandemic situation and a lack of COVID-19 vaccines. As well as data on quality, safety and efficacy, specific mitigations and conditions on the product were discussed to ensure adequate standards of quality and safety are met. |
| - | On this page, you will find information on the Agency’s activities that are most relevant | + | The CHM concluded that the proposed supply of COVID-19 mRNA Vaccine BNT162b2 for active immunisation |
| + | Authorisation for the temporary supply of COVID-19 mRNA Vaccine BNT162b2 was granted in the UK on 1 December 2020. This report covers data received and reviewed for this authorisation only. This authorisation is valid until expressly withdrawn by MHRA or upon issue of a marketing authorisation. | ||
| - | ==== EU Innovation Network ==== | + | On 4 June 2021 the MHRA granted an extension of indication to ‘the active immunisation to prevent COVID-19 caused by the SARS-CoV-2 virus, in individuals 12 years of age and older’. ((https:// |
| - | Innovation offices in national regulatory agencies have been working informally with the EMA's [[: | ||
| - | EMA and the HMAs adopted the mandate of the EU-Innovation Network in October 2016 | + | ==== Publications ==== |
| - | === Mandate of the European Innovation Network | + | ==== EMA COVID-19 Steering Group ==== |
| - | {{ :: | + | |
| - | 2014 - The [[: | + | |
| - | The workshop provided an opportunity to discuss expectations with different stakeholders such as members | + | EMA’s [[:COVID-19 Steering Group]] provides strategic oversight at EMA of the evolving scientific |
| - | The aim of the network is to make the regulatory support for medicines developers currently available at national and EU levels more visible and attractive to innovators ((https:// | + | ===== External links ===== |
| - | ==== Publications | + | * [[https:// |
| + | |||
| + | ==== Links to sort ==== | ||
| + | |||
| + | * [[https:// | ||