1 / 58

Drug Regulatory Overview

Drug Regulatory Overview. Milan Smid, MD, PhD Technical Officer Quality Assurance and Safety: Medicines Essential Medicines and Pharmaceutical Policies, World Health Organization smidm@who.int. Principles of drug regulation WHO regulatory support activities Harmonization and worksharing

kevinoliver
Download Presentation

Drug Regulatory Overview

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Drug Regulatory Overview Milan Smid, MD, PhD Technical Officer Quality Assurance and Safety: MedicinesEssential Medicines and Pharmaceutical Policies, World Health Organization smidm@who.int

  2. Principles of drug regulation • WHO regulatory support activities • Harmonization and worksharing • Assessment of regulatory functions • Trainings • Discussion

  3. Major health technologies Blood components Transplants Medicines and vaccines Medical devices including diagnostics Herbal and traditional medicines Functional food Nutraceuticals Advanced therapies

  4. Common features of health technologies • Used to prevent, diagnose or treat disease • Benefits from use must overweight the risks in order to improve health • To evaluate risk-benefit is frequently highly scientifically demanding • Not all health technologies bear comparable risks • Health technologies are subject of commerce and profit oriented behaviour

  5. Protection of public health • To achieve acceptable level of risk related to use of health technologies and maximize their benefit, governments have to intervene • Governmental interventions interfere with rights of persons and companies and with business practices • Regulatory systems and scope of regulation differ worldwide • Health products are split into different categories • Regulation of medicines is currently one of most sophisticated and complicated regulatory functions globally

  6. Medicines regulation • Medicines regulation is the totality of all measures - legal, administrative and technical - which governments take to ensure the safety, efficacy and quality of medicines, as well as the relevance and accuracy of product information • Medicines are special goods for which also prices are frequently regulated by governments to achieve affordability for patients • Medicines are needed and different government incentives are used to develop and produce them

  7. Medicines regulation Models for regulation of medicines are determined by: • Public health needs • Organization of the state and state administration • Size of the pharmaceutical market • Presence and type of pharmaceutical industry • Availability of resources (human, scientific, financial) • Maturity of stakeholders • Participation in regulatory networks

  8. Rationale for Government role Consequences of weak drug regulatory capacity • substandard, counterfeit, harmful, useless medicines on the market • biased information about medicines circulates • substandard practices in clinical testing, manufacture and supply of medicines • irrational prescription and consumption

  9. Rationale for Government role Consequences of over- or improper regulation • lack of needed medicines or delayed access • increased costs of medicines due to cost of regulatory system • lack of regulatory flexibility

  10. Regulatory basics Regulation should • Keep patient in its centre • Be evidence based • Be risk oriented • Bring added value • Respect interests of stakeholders and real possibilities • Be transparent but respect confidentiality • Be effective and flexible • Be part of broader drug policy

  11. Parties involved in regulatory system • Regulated subjects • Investigators and sponsors, members of ethics commitees • Manufacturers and importers • Wholesalers and distributors • Health professionals • Regulators • Regulatory authorities • Governments and political representations • Interested parties • General public and patients groups • Academia • Media

  12. Development of drug regulation • Quality • Safety • Efficacy • Information • Environment • Risk management and risk minimization

  13. Essential regulatory activities • R&D – data quality and ethics(GLP, GCP, bioethics) • Manufacture, import and supply – quality(GMP, GDP) • Market entry and existence on the market – Q, S, E, I, Env authorization/registration, maintenance, renewals • Special access schemes • Dispensation – safe and indicated use (GPhP) • Use – pharmacovigilance (GPhVP) • Promotion – unbiased, valid and understandable information (Conduct codes) • Environment – premarket review and environment monitoring • Regulatory and scientific advice

  14. Regulatory toolkit • Legislation • Standards (GXPs, Pharmacopoeias, Q,S,E guidelines) • Data assessment • Submitted data • Collected data • Inspections • Licensing (authorizations / registrations, products and operations) • Certification • Laboratory control • Provision of information • Regulatory intelligence and co-operation

  15. Incentives to develop, manufacture and supply needy medicines • Patents and supplementary patent protection • Data exclusivities • Market exclusivities • Limited data requirements • Regulatory advice • Regulatory fast tracks • Waivers of regulatory fees • Public-private partnership, stimulating price, guaranteed demand

  16. Regulatory environment Legislation Guidelines Courts Appeal bodies Regulated subjects Regulatory authorities Interested stakeholders (associations, professional bodies, NGOs, watch dogs)

  17. Responsibilities of parties involved in regulatory system Legislated • Investigators and sponsors • Manufacturers and importers • Wholesalers and distributors • Health professionals • Regulatory authorities Non-legislated • Governments and political representations • Patients • Media

  18. Registration Line extensions Postregistration commitments Renewals PSUR PSUR PSUR Drug Regulatory Cycle PSUR Variations Emergency and crisis management PSUR PSUR

  19. Principles applied in regulatory approvals • Benefits prevail the risks at a time of regulatory approval and nothing indicates that benefits will not prevail also during use of product in normal medical practice • Available data about quality (Dossier) • Available data about efficacy and safety or interchangeability (Dossier) • Available data are credible and were eticaly obtained • Good practices (GLP, GCP, GPhVP, …) • Existing reassurance about production in stable quality and quality assurance mechanisms • GMP • Way of use of medicine characterized for physicians and patients • Data sheets, SPCs, PILs, package labeling • Lack of knowledge is properly managed • Pharmacovigilance, risk management programmes, commitments

  20. Regulation of innovatory products and generics • For innovator products proof of QUALITY, SAFETY and EFFICACY is needed. Newly also plan of prospective risk-management. • For multisource products QUALITY, safety and efficacy data is referred to the originator, providing only evidence of INTERCHANGEABILITY (bioequivalence, clinical testing, in certain cases dissolution data).

  21. Data required for regulatory approval Innovative medicine Experimental data/ Literature Clinical data Generic medicine Multisource interchangeable Preclinical data Proof of interchangeability Pharmaceutical data Pharmaceutical data Administrative and summarizing data, including GMP

  22. Scientific and regulatory complexity concerning medicinal products is growing • New technologies • New therapeutic approaches • Evidence based medicine • Globalisation of commerce • Telematic instruments • Information and transparency requirements • Risk aversion in society • New tasks • Speed of change • Regulatory resources rarely reflect expectations of society and regulators themselves Paradigms of drug regulation are evolving

  23. Capacity to make regulatory decisions 193 WHO Member States:

  24. Challenges • Regulatory delays and backlogs • Lack of regulatory tools and guidelines • Lack of expertise • Lack of management skills • Regulatory pendulum

  25. Strategies used to cope with increasing demands? • Concentration on priority issues most relevant for public health • Co-operation with partners in order to increase regulatory capacity by elimination of duplicated activities • Facilitated by comparable standards and administrative requirements • Increased effectivity of internal operations • Quality systems, international benchmarking

  26. Sharing of expertise vs. recognition of decision • Acceptance of expertise is not equal to acceptance of decision • Acceptance of decision • is formal legal act, frequently requiring international treaties • may modify liabilities of involved parties and requires legal specification of acceptance and non-acceptance • Acceptance of expertise • is sovereign and complex regulatory decision of NRA based on scientific arguments and confidence • may be applied case to case • is followed by formal independent decision according to national legislation and mandate of NRA

  27. WHO approaches for consideration • Stimulating / initiating collaboration between regulators from various developing countries on various regulatory activities; • Employing internationally accepted guidelines adapted to suit local needs/circumstances; • Facilitating joint assessments between regulators through sub-regional approaches.

  28. 13th ICDRA16-19 September 2008, Bern • Concentration on priority issues most relevant for public health • Risk-based approaches: regulation of clinical trials, GMP, product licensing, safety monitoring, market oversight • Increased effectivity and efficiency of internal operations • assessment of practices and performance by common toolkits

  29. 13th ICDRA16-19 September 2008, Bern • Co-operation with partners in order to increase regulatory capacity by elimination of duplicated work investments and optimising use of experts • Harmonization: standards: terminology, guidelines, good practices, pharmacopoeias, format and data content of applications, safety monitoring practices, • Collaboration and networking: inspections, market surveillance, counterfeits, • Information sharing and common data sets, information availability: trial registries, inspection outcomes and assessment reports, licensing outcomes, pharmacovigilance • Building mutual trust • New phenomenon: Major regulatory authorities (FDA, EMEA) recognize need of collaboration with NRA in exporting countries concerning oversight of manufacturers and information exchange

  30. Different initiatives to increase regulatory capacity and assemble needed scientific / inspection resources WHO – standards, international expert teams, pharmacovigilance UMC ICH – common standards for developed markets, involvement of regulators as well as industry experts EMEA - co-ordination of expertise available to EU/EEA Member States Australia and New Zealand – merger of NRAs (currently frozen) Monaco, Liechtenstein – recognition of regulatory decisions of France resp. Switzerland Canada, Switzerland, Japan, EU, Australia etc. – MRA on mutual acceptance of GMP standards FDA, EMEA, WHO, TGA … etc. – MoU on sharing of information PIC/S – share of standards and expertise on GMP, building confidence Regional and subregional initiatives (ASEAN, GCC, EAC) – different extent of co-operation National initiatives – involvement of external experts to support NRAs, recognition of expertise or decisions, co-operation with industry and learned societies

  31. Not to share expertise - If trustful expertise is available, not to consider it may be unjustified waste of resources needed elsewhere • Accepting the expertise of other regulators is complex regulatory and scientific decision, depending on • legislation, mandate • capacities and competence available for given issue • scientific arguments related to benefit/risk for public health • priorities for protection of public health • Understanding to limitations of transfer of expertise and its proper management are needed when considering expertise of others • Targeted, risk-based approach may be appropriate in some situations

  32. Expertise in support of developing countries • WHO PQP - HIV/AIDS, TB, malaria, RH, paediatric therapy • US PEPFAR - HIV/AIDS • EU Article 58 – assessment of products for non-EU teritories • Canada's Access to Medicines Regime – assessment of products according to WHO Model List of Essential Medicines • Other - orphans, paediatric therapy

  33. Faster access to medicines through sharing of regulatory information • WHO is working with regulators to find out how best to build confidence in regulatory decisions taken by other regulators, including -- how to facilitate exchange of consolidated information about assessments.

  34. WHO registration package Purpose: • For less-resourced agencies – in decision making process, to benefit from technical information developed by well-resourced ("mature") regulatory authorities; • To facilitate the "transfer of expertise"; • To create a format for exchange of regulatory information.

  35. Different initiatives to increase regulatory capacity WHO proposed a framework based on what is publicly available: • Summary Basis for Decision (Health Canada) • European Public Assessment Report (EMEA and EC) • Common Technical Document (US FDA) • WHO Public Assessment Report (WHOPAR) and Public Inspection Report (WHOPIR)

  36. WHO Pilot Project • Supported by European Commission; • With participation of 5 (7) countries: Ghana, Kenya, Nigeria, South Africa, Uganda, United Republic of Tanzania and Zimbabwe

  37. WHO Pilot Project Purpose: • To field-test a model technical registration package; • To finalize the content of the package and the guidance for its effective implementation in countries; • To ensure that the package is developed and implemented through a wide consultation with existing and potential stakeholders;

  38. WHO Pilot Project Field testing exercise • In order to have a maximum benefit from the field-test exercise, it was decided to provide all participating DRAs with the same drug application; • To ask them to develop their own registration package, based on the provided draft template; • One of the recently developed pharmaceutical products has served as a model application for the field-testing of the WHO model registration package.

  39. WHO Pilot Project • The same application was submitted to EMEA experts for their opinion; • In the end of the project a follow up WHO consultative meeting was conducted; • The aim was to consider: • the results and lessons learnt from the field-testing exercise, • to finalize the contents of the model technical regulatory package, • to provide guidance for adoption of the package by national MRAs.

  40. Conclusions (1) • The main objective for DRAs should be to get the relevant information for the country, while avoiding collection of large amounts of the country-specific data that does not add value; • Sharing of information can help with best use of available resources, reduce workload and improve overall regulatory performance; • Available tools can be packaged for ease of reference and information exchange;

  41. Conclusions (2) • Procedures can be streamlined and processes can be improved (e.g., Fast Track or Priority Review for those products that address unmet medical needs); • Expert knowledge can be pooled and resources directed to functions that can improve public health and facilitate access to essential medicines.

  42. Regulatory support core functions • Developing evidence on the situation of Medicines Regulatory Systems worldwide • Providing Country support for strengthening medicines regulation • Providing Regional support for strengthening medicines regulation • Facilitate communication and promote harmonization among medicines regulatory authorities • Develop and continuously improve internal supporting tools, mechanism and capacities

  43. Developing evidence • To develop and maintain a comprehensive database on DRAs • To assess of medicine regulatory systems • Perform assessment to identify needs • Provide evidence for various supporting activities (Financial, Training, Consultancy,Equipment) • Develop institutional plan • To promote self-assessment as a tool for analysis and continuous improvement • Two training events performed for DRAs • Tool for harmonization purposes • 5 Pacific Island Countries / WPRO • 3 EAC Countries / AFRO

  44. Developing evidence • 44 Assessments performed on 40 Regulatory systems (with the involvement of HQ) • AFRO - 21 COUNTRIES / 24 ASSESSMENTS • EURO - 2 COUNTRIES / 2 ASSESSMENTS • EMRO - 4 COUNTRIES / 5 ASSESSMENTS • SEARO - 4 COUNTRIES / 4 ASSESSMENTS • WPRO - 7 COUNTRIES / 7 ASSESSMENTS • PAHO - 2 COUNTRIES / 2 ASSESSMENTS

  45. Developing evidence 2008 2007 2006 2004 2003 2003 No

  46. Country support • Develop and organize training opportunities • Strengthen information management capacity - SIAMED • Strengthen inspection capacity • Strengthen QC laboratory capacity • Strengthen marketing authorization capacities • Promoting good regulatory practices by providing guidelines, tools and technical assistance • In close collaboration with capacity building team within the PQ Program • In cooperation with IVB Vaccines on clinical trials

  47. Regional support • Provision of technical assistance to harmonization initiative and supporting participation of regulators • SADC, EAC, PIC, CARICOM,… • Financial support for technical secretariat (EAC, SADC and UEMOA) • WHO/EAC joined assessment of DRAs • Promoting and facilitating networking • Network of DRAS (EAC, SADC)

  48. Introduction to the grading model • Preliminary work made by Eshetu Wondemagegnehu • Possible approaches to developing drug regulation in: Effective Drug Regulation: what can countries do? • Based on Capability Maturity Model (CMM) • first described byW. Humphrey in Managing the Software Process • Maturity model for quality management system • introduced by P. Crosby in his book 'Quality is Free'. Generic approach of maturity of organization as a learning curve applied to medicines regulatory processes Not to give a mark

  49. For what purposes can it be used? • At country level • To build a strategy • To visualize the stage of development/maturity • To visualize and demonstrate progress • To assist process of harmonization • To identify areas of priority support • At sub-regional level • To organize sharing and exchange of expertise • To develop process of harmonization, cooperation or collaboration • At international level • To map regulatory systems • To build strategy/approach • To document results and planned activities

  50. Grading Model / MRA

More Related