EMA - Early Access. PEARRL Annual Meeting 2017-Regulatory Science Symposium. University College Cork, Ireland

Similar documents
PRIME. 7 th STAMP meeting Brussels, 27 th June Presented by Sonia Ribeiro Head of Regulatory Affairs Office, Human Medicines Evaluation Division

Priority Medicines (PRIME) scheme

PRIority MEdicines (PRIME)

EU scientific regulatory support mechanisms and initiatives for innovation in drug development: the EMA perspective

EU regulatory tools for expedited antibacterial development programmes

Regulatory update from Europe:

EMA/CAT support to ATMP developers

Conditional marketing authorisation

Regulatory point of view on clinical benefit assessment and parallel EMA/HTA advice

EMA Adaptive licensing: a tool concept for accelerated access to innovative medicines? Rob Hemmings, MHRA

Strengthening the prospective discussions on post-licensing evidence generation

EU Update on Regulatory Developments

Accelerated Approvals

Use of Real World Data in Development Programmes

PRIME: a two-year overview

Optimising early access tools: Revision of the guidelines on Accelerated Assessment and Conditional Marketing Authorisation

This video gives an overview of the centralised procedure at the European Medicines Agency

The role of patients at the EMA

Authorisation of ATMPs in EU: routes to facilitate prompt availability for patients.

Earlier Access to Medicines Early Access to Medicines Scheme and Adaptive Licensing pilot

Current Issues in EU Cell & Gene Therapy Regulation. Christiane Niederlaender, UK CAT Delegate, MHRA

Adaptive pathways workshop briefing book

Marketing Authorisation Routes in the EU

Final report on the adaptive pathways pilot

EUROPEAN COMMISSION DIRECTORATE-GENERAL FOR HEALTH AND FOOD SAFETY

Considerations on regulatory aspects

Use of real world data pre-authorisation what can it answer?

Orphan designation in the EU

Regulatory challenges and opportunities for the use of Real World Evidence for drug registration and labelling

Session 5: Dealing with Unmet Medical Needs and Support to Innovation

Office for support of INNOVATION and KNOWLEDGE of medicinal products. _ Spanish Agency of Medicines and Medical Devices

Supporting Innovation through Scientific Advice

Issues identified by stakeholders: follow-up from EMA s ATMP workshop

Orphan designation in the EU

Submission of comments on COMMISSION NOTICE ON THE APPLICATION OF ARTICLES 3, 5 AND 7 OF REGULATION (EC) NO 141/2000 ON ORPHAN MEDICINAL PRODUCTS

ORPHAN DESIGNATION BY THE EMA. Paillard Juliette M2 AREIPS 15/11/2016

Speed your time to market with FDA s expedited programs

HTAs and EMA working together: 23 parallel scientific advice procedures later - what have we learned?

Update on Real World Evidence Data Collection

Clinical Development and enabling Regulatory Steps: How to obtain ODD and Scientific Advice at EMA

Commission notice on the application of Articles 3, 5 and 7 of Regulation (EC) No 141/2000 on orphan medicinal products (2016/C 424/03)

Orphan Drug Designation within the Development Strategy

Engagement with stakeholders

Orphan Medicinal Products

Docket #: FDA-2018-D-3268

Overview of the Agency s role, activities and priorities for An agency of the European Union

Scientific advice and its impact on marketing authorisation application reviews

Use of RWE in a regulatory context: issues and examples. Rob Hemmings, MHRA

Accelerated Development of Appropriate Patient Therapies

6th EUROPEAN CONFERENCE ON RARE DISEASES & ORPHAN PRODUCTS ECRD 2012 Brussels

Applying for EU marketing authorisation. For medicinal products for human use. An agency of the European Union

The EU Risk Management Plan - a tool to address the uncertainties at the time of approval, and manage the risks of medicines

Personalised Medicine Regulatory Issues

Use of immunotherapy for cancer treatment

The compassionate use of medicinal products. An example: the French ATU system. 0ff label use in France

Advanced Therapies in Europe

Content. Introduction: Approval of ATMPs. Support to ATMP developers. Role of EMA and National Competent Authorities

European contribution to the RWD/RWE debate. Alasdair Breckenridge July 2018

The Right Data for the Right Questions:

Regulatory utilization of Flexible Regulatory Pathways to meet unmet medical need

CVMP ad hoc veterinary expert group on novel therapies ADVENT

Recommendation on elements required to support the medical plausibility and the assumption of significant benefit for an orphan designation

Track III: International Clinical Trials: Global Compliance Norms and EU Focus

Work programme July 2016 EMA/92499/2016 Rev. 1 1 Executive Director. 30 Churchill Place Canary Wharf London E14 5EU United Kingdom

How CAT Works: Regulatory Process, Patient Contribution and Outcomes

ABPI response to European Commission consultation on advanced therapy medicinal products

Position Paper. Executive Summary

1. Council conclusions on strengthening the balance in the pharmaceutical systems in the EU and its Member States

Conditional Early Approval System for Innovative Medical Device Products (Fast-Break Scheme)

-Regulation (EC) No.1394/2007 -Regulation (EC) No. 668/2009

Summary of Product Characteristics Advisory Group (SmPC AG) activity report

ATMPs guideline on Safety and Efficacy follow-up and risk management

Initial Draft Discussion Document for A Canadian Orphan Drug Regulatory Framework

Stakeholder Consultation: Supplemental Process for Complex/Specialized Drugs Background Document

Regulatory Issues and Strategies: U.S. FDA

Szabolcs Barotfi Ph.D.

Orphan Drug Development Strategic considerations

Following Cancer Drug Pathways from Inception to Launch: Partnership, prospects and Pitfalls. Katie Pascoe (Value and Access, ABPI) 22 September 2016

Interested parties (organisations or individuals) that commented on the draft document as released for consultation.

This template is to be used by companies willing to submit an overview of relevant

Lessons learnt from the Ebola outbreak A regulatory position

Using local RWD to drive global therapeutic advancements.

EUROPEAN COMMISSION DIRECTORATE-GENERAL FOR HEALTH AND FOOD SAFETY

Coordination and Support Action: Enabling platform on medicines adaptive pathways to patients

New Pathways to Breakthroughs for Progress & Patient Access

Introduction to the European Medicines Agency

Early Access to Medicines Scheme (EAMS)

Update from the Agency s SME Office

AN INSIGHT ON ORPHAN DRUG DEVELOPMENT STRATEGY IN US AND EUROPE

Review of the ATMP Regulation

Work programme Adopted by the Management Board on 18 December December 2014 EMA/773839/2014 Rev. 1 Management Board

- OMICS IN PERSONALISED MEDICINE

EU health policy. Strategy for the pharmaceutical industry and biosimilars. Salvatore D'Acunto. DG Research. DG Internal Market. DG Health & Consumers

Moving HTA forward: The challenges of incorporating real world evidence into Health Technology Assessment

Pediatric drug development - do we need a PIP so early in development? American versus European approach

FDA from a Former FDAer: Secrets and insights into regulatory review and drug development

What are the real-world evidence tools and how can they support decision making?

AIFA s post-marketing registries and accelerated patient access. Opportunities and challenges in the context of MAPPs

Role of drug regulation (if any )

GMO Technology Conference

Transcription:

EMA - Early Access PEARRL Annual Meeting 2017-Regulatory Science Symposium University College Cork, Ireland Presented by Evangelos Kotzagiorgis Scientific Administrator, Quality of Medicines Office Specialised Scientific Disciplines Department An agency of the European Union

Agenda Early access tools Conditional marketing authorisation What is it? Experience EMA Adaptive Pathways Why do we need to be adaptive? - the concept Critical issues Conclusions EMA PRIME: priority medicines What is it? The concept - key points Conclusions 1

Early access tools: Overview Other Compassionate Use, Marketing Authorisation (MA) under exceptional circumstances, etc. PRIME Major public health interest, unmet medical need. Dedicated and reinforced support. Enable accelerated assessment. Better use of existing regulatory & procedural tools. Adaptive Pathways Scientific concept of development and data generation. Iterative development with use of real-life data. Engagement with other healthcare-decision makers. Accelerated Assessment Major public health interest, unmet medical need. Reduce assessment time to 150 days. Parallel advice Conditional MA Unmet medical need, seriously debilitating or life-threatening disease, a rare disease or use in emergency situations. Early approval of a medicine on the basis of less complete clinical data. 2

Conditional marketing authorisation Eligibility for products in at least one of these categories: aimed at treating, preventing or diagnosing seriously debilitating or lifethreatening diseases; intended for use in emergency situations (also less comprehensive pharmaceutical and non-clinical data may be accepted for such products); designated as orphan medicines. CMA may be granted if all the following requirements are met: the benefit-risk (B/R) balance of the product is positive; it is likely that the applicant will be able to provide comprehensive data; unmet medical needs will be fulfilled; the benefit to public health of the medicinal product's immediate availability on the market outweighs the risks due to need for further data. 3

Conditional marketing authorisation CMA valid for one year - renewed annually. required to complete specific obligations (ongoing or new studies, and in some cases additional activities) objective to provide comprehensive data confirming that the B/R balance is positive. Once comprehensive data obtained, the marketing authorisation may be converted into a standard marketing authorisation 4

Conditional marketing authorisation Authorisation before comprehensive data are available in order to address unmet medical needs, when benefits of early access outweigh the risks due to limited data CMA exceptional circumstances (comprehensive data cannot be obtained even after authorisation). Access Pharmaceutical + nonclinical Phase I and II Confirmatory phase III Assessment and Approval Reimbursement and launch A key tool for early access: Product can be authorised several years earlier Comprehensive data are still generated after authorisation 5

Conditional Marketing Authorisations Apart from 2016, there is no clear trend in number of CMAs, which remain an exceptional authorisation route On average within 4 years a conditional MA is converted into a standard MA 6 Data updated on Dec 2016

Key findings of 10 year analysis Apart from 2016, there is no clear trend in overall number On average within 4 years a conditional MA is converted into a standard MA Reluctance in pro-active use by industry room for improvement in prospective planning CMAs successful only in few therapeutic areas Most specific obligations did not have any change 7 Conditional marketing authorisation - 10 year analysis

Agenda Early access tools Conditional marketing authorisation EMA Adaptive Pathways Why do we need to be adaptive? - the concept Critical issues Conclusions EMA PRIME: priority medicines 8

Please, allow me to introduce you to Jane, late fifties, recent diagnosis of advanced cancer, life expectancy: ~2 years John, late fifties, in good health, family history of cancer, life expectancy: ~20 years There is a promising treatment out there; but it s still early days Should our healthcare systems* cater for the needs of A: only Jane? B: only John? C: Jane and John? An iterative, life-span approach to learning and on-market access, a.k.a. Adaptive Pathways http://www.ema.europa.eu/ema/index.jsp?curl=pages/regulation/general/general_content_000 9 601.jsp&mid=WC0b01ac05807d58ce presented at the PEARRL Regulatory symposium 2017 for personal use only

Access vs evidence : an ethical and scientific conundrum Need to reduce (unavoidable) uncertainties fast Adaptive Pathways a solution to inevitable problems? Development of nonconventional products (e.g. ATMPs) Sustainability of costs Need to enlarge the toolbox for evidence generation (where RCTs cannot answer the questions) presented at the PEARRL Regulatory symposium 2017 for personal use only 10

Adaptive Pathways - component parts Leverage multi-stakeholder collaboration Manage on-market utilisation Focus on high unmet need (sub-)population first, and on products likely to have major impact for patients Use entire tool box for knowledge generation Reduce uncertainty as fast as possible; react to incoming data (iterative development; rapid cycle analysis) Pre-plan, across entire life span (incl. postmarketing) presented at the PEARRL Regulatory symposium 2017 for personal use only 11

Adaptive Pathways harnessing existing tools Conditional marketing authorisation (in EU legislation) Post-marketing commitments; Risk Management Plans (in PharmacoVigilance Regulation) Multi-stakeholder scientific advice Registries, other data sources Adaptive pricing/reimbursement (managed entry agreements) presented at the PEARRL Regulatory symposium 2017 for personal use only 12

Agenda Early access tools Conditional marketing authorisation EMA Adaptive Pathways Why do we need to be adaptive? - the concept Critical issues o Need and unmet need? o Lowering the standards? o Randomised Controlled Trials (RCT) vs Real World Data (RWD) o Promises, compliance, exits" o On-market utilisation Conclusions EMA PRIME: priority medicines 13

Need and unmet need Early access is it worth it? Addressing unmet need ; focus on: Conditions with major impact on quality of life / life-shortening / debilitating Credible promise of relevant improvements in patientrelevant outcome(s) an acceptably high probability of a relevant effect size presented at the PEARRL Regulatory symposium 2017 for personal use only 14

Lowering the standards? Access vs evidence conundrum has always been acknowledged: where, the benefit to public health of the immediate availability on the market [ ] outweighs the risk inherent in the fact that additional data are still required [Regulation (EC) No 507/2006] presented at the PEARRL Regulatory symposium 2017 for personal use only 15

Lowering the standards? Benefit-Risk (B/R) must be positive for treatmenteligible population. Same standards for the evaluation of B/R or the requirement to demonstrate a positive B/R balance to obtain marketing authorisation. Challenges regarding some particular aspects, e.g. sufficient stability data? Manage uncertainty. presented at the PEARRL Regulatory symposium 2017 for personal use only 16

Randomised Controlled Trials (RCTs) and Real World Data (RWD) 1 RCTs are the methodology with the highest internal validity ( gold standard, not black & white) RCTs & RWD 2 3 For efficient increase of knowledge of benefits and risks: embrace the full evidence spectrum (RCTs, pragmatic trials, observational studies) RWD complements rather than replaces RCTs. The right study type for the right question where feasible 4 Pre and post-licensing evidence generation are not two different lives, it s one continuous life presented at the PEARRL Regulatory symposium 2017 for personal use only 17

Promises, compliance, exits Promised data may not be forthcoming, post marketing commitments might not be honoured Compliance with legally binding post marketing studies generally good (but start of studies slow). Regulatory system is robust*; supported by recent experience (post 2012) * http://ec.europa.eu/health/files/pharmacovigilance/pharmacovigilance-report-2012-2014_en.pdf presented at the PEARRL Regulatory symposium 2017 for personal use only 18

Promises, compliance, exits Subsequent data may not confirm initial promise of high effect size For regulators, not a new scenario For payers, plan exit (or adaptive disengagement ) scenarios upfront Payers can get incentives right: limited initial label with prospect of widening, flexible conditions of reimbursement presented at the PEARRL Regulatory symposium 2017 for personal use only 19

On-market utilisation Regulators can provide some (!) steer on appropriate prescribing (Risk Management Plans) Right incentives (for companies) will help Payer action will be helpful but heterogeneity across EU member states is acknowledged Access to local healthcare data / drug utilisation review will facilitate appropriate utilisation where feasible presented at the PEARRL Regulatory symposium 2017 for personal use only 20

Conclusion Adaptive Pathways is an attempt to solve inevitable problems and challenges in an imperfect world These can be successfully addressed by way of adequate pre-planning, and collaboration of stakeholders EMA will continue to explore the potential of the Adaptive Pathways concept. Scientific concept of development and data generation. 21

Agenda Early access tools Conditional marketing authorisation EMA Adaptive Pathways Why do we need to be adaptive? - the concept Critical issues Conclusions EMA PRIME: priority medicines What is it? The scheme- key points Conclusions 22

Features of the PRIME scheme Early access tool, supporting patient access to innovative medicines 23 LAUNCHED in March 2016 Written confirmation of PRIME eligibility and potential for accelerated assessment; Early CHMP Rapporteur appointment during development; Kick off meeting with multidisciplinary expertise from EU network; Enhanced scientific advice at key development milestones/decision points; EMA dedicated contact point; Fee incentives for SMEs and academics on Scientific Advice requests.

PRIME scheme - Goal & Scope To foster the development of medicines with major public health interest.?! Enable accelerated assessment Promote generation of high quality data Facilitated by knowledge gained throughout development Reinforce scientific and regulatory advice Foster and facilitate early interaction Raise awareness of requirements earlier in development Building on existing framework; Eligibility according to existing Accelerated Assessment criteria Optimise development for robust data generation Focus efficient development Promote generation of robust and high quality data

Eligibility to PRIME scheme Based on Accelerated Assessment criteria Medicinal products of major public health interest and in particular from the viewpoint of therapeutic innovation. Potential to address to a significant extent an unmet medical need Scientific justification, based on data and evidence available from nonclinical and clinical development No satisfactory method or if method exists, bring a major therapeutic advantage Introducing new methods or improving existing ones Meaningful improvement of efficacy (impact on onset, duration, improving morbidity, mortality) 25

What do we expect to grant eligibility? Unmet medical need No treatment or clear limitations of existing therapies Nonclinical data supporting pharmacological rationale (e.g. gene therapy) Clinical exploratory data on relevant endpoint If uncontrolled, use comparable historical control i.e. need sufficient information on baseline characteristics Magnitude of the effect size supporting major therapeutic advantage 26

Entry points PRIME eligibility and required evidence Nonclinical Phase I Exploratory Confirmatory SMEs Academia Confirmation Any sponsor Proof of principle (For SMEs and academia only) Sound pharmacological rationale, convincing scientific concept Relevant nonclinical effects of sufficiently large magnitude and duration Tolerability in first in man trials Proof of concept Sound pharmacological rationale Clinical response efficacy and safety data in patients (exploratory trials) Substantial improvement Magnitude, duration, relevance of outcomes to be judged on a case by case basis 27

To take place shortly after eligibility confirmation, at EMA Kick-off meeting Multi disciplinary meeting with relevant experts from SAWP and CHMP and other committees; Facilitate initial interaction between applicant and EU regulatory network; Discuss the overall development plan and regulatory strategy; Provide recommendation on milestones and topics for scientific advice. Introduction of product and development status by applicant; 28

Early Rapporteur appointment Opportunity for knowledge gain on the product Identification of relevant expertise and build adequate team Opportunity to influence development Very positive views on the kick-off meeting Importance of preparation and tailored agenda Facilitate interactions across committees and with EMA Timing of PRIME eligibility is critical for fruitful engagement Involvement in follow-up scientific advice and workload Need to improve follow-up communications/updates 29

Enhanced scientific advice 7 products 11 SA requests following kick-off meetings Multi-stakeholder 1 EMA/HTA parallel advice 2 with patients involved Rapporteur involvement through one of SAWP coordinator Scientific advice All aspects covered Quality, nonclinical, clinical Flexibility Shorter pre-submission 3 adopted in 40 days 30

Requests covering wide range of therapeutic areas and product type 70 % in oncology/haematology 34% of requests for ATMPs 31

In summary, Eligibility review: robust, short time, in writing Majority of PRIME products in rare diseases Iterative scientific advices with opportunity for multi-stakeholders involvement, including patients 32

Acknowledgements Hans-Georg Eichler - Senior Medical Officer Zahra Hanaizi - Scientific officer, PRIME coordinator Jordi Llinares - Head of Scientific and Regulatory Management Department Zigmars Sebris - Scientific Officer Regulatory Affairs 33

Thank you for your attention Further information European Medicines Agency 30 Churchill Place London E14 5EU www.ema.europa.eu info@ema.europa.eu Follow us on @EMA_News