Adaptive Licensing Orphan Cafe 12 juni 2014

Programma:
• Mignon van der Westerlaken
Voorzitter HollandBIO- expertgroep Orphan Drugs
• Bert Hiemstra
Hoofd afdeling Farmacologische, Toxicologische en
Biotechnologische Beoordeling bij het CBG
• Anthony Hall
Hoofd wetenschappelijke raad van PSR: the Orphan Drug
Experts,
Adaptive licensing
Adaptive licensing
Verantwoorde verkorting van de time to patient
Bert Hiemstra, College ter Beoordeling van Geneesmiddelen
Adaptive licensing
1) Waarom adaptive licensing?
2) Wat is adaptive licensing?
3) Wat doen de regulators?
4) Wat moet nog opgelost?
Adaptive licensing
Waarom adaptive licensing? - 1
Adaptive licensing
Waarom adaptive licensing? - 2
Adaptive licensing
Waarom adaptive licensing? - 3
Minister aan T.K.:
“Als het lukt om geneesmiddelen op een veilige manier
daadwerkelijk sneller op de markt te krijgen, hoeven
patiënten minder vaak een beroep te doen op de diensten van
organisaties als My Tomorrows.”
Adaptive licensing
Waarom adaptive licensing? - 4
Scenario‘s CBG 2030
Links en rechts ingehaald
Adaptive licensing
Wat is adaptive licensing? - 1
• Doel is goede geneesmiddelen z.s.m. bij de patiënt, op
verantwoorde manier
• Diverse synoniemen, o.a. MAPP (Medicines Adaptive
Pathways to Patients)
Adaptive licensing
Wat is adaptive licensing? - 2
•
•
•
•
•
Start met specifieke patiëntengroep met grote medical need
Geneesmiddel moet aansluiten bij die need
Er wordt onzekerheid geaccepteerd in benefit en / of risk
Onzekerheid in benefit en/of risk betekent niet slechte data
Niet meer data vragen dan nodig, maar ook geen regulation light
Uncertainty
Need
Adaptive licensing
Wat is adaptive licensing? - 3
• Afspraken over verdere data verzameling na registratie
• Primair om onzekerheid te verminderen
• Data na registratie kunnen ook bijdragen aan verbreding
indicatie
• Uiteindelijk worden meer data verzameld dan in regulier
proces
• Zijn bovendien grotendeels real life data
Adaptive licensing
Adaptive licensing
Wat is adaptive licensing? - 5
• Alle disciplines, ook quality (bijvoorbeeld omgang met nog
niet volledig gevalideerde full scale)
• Patiënt, behandelaar en andere stakeholders direct aan
tafel
• Every day life perspectief van patiënt (benefit & risk)
• Vroeg in ontwikkeltraject beginnen, belang van goede fase
1 en 2.
• Adaptieve prijsbepaling en vergoeding als onderdeel van
adaptive licensing
• Verschillende scenario’s ontwikkeltraject doornemen
• Belangrijke rol scientific advice, maar adaptive licensing
betreft meer dan advisering alleen
Adaptive licensing
Wat doen de regulators? - 1
• CBG Strategisch Business
Plan 2014 -2018
• Levenscyclus met drie
kernelementen
• Patiënt belangrijkste
stakeholder
• Samen met omgeving
Adaptive licensing
Wat doen de regulators? - 2
•
•
•
•
Samenwerking met ZIN, CCMO en IGZ
Advies op maat: vroeg, meerdere contactmomenten, laagdrempelig
Bevorderen maar ook onafhankelijk oordelen
Experimenteerruimte
Adaptive licensing
Wat doen de regulators? - 3
•
•
•
•
EMA pilot
Bedrijven melden zich
Actieve rol CBG bij validatie, prioritering en uitvoering pilots
Eerste pilot start binnenkort
Adaptive licensing
Wat doen de regulators? - 5
Uitgebreid palet aan wettelijke mogelijkheden:
•
•
•
•
•
•
Conditional approval
Exceptional circumstances
Compassionate use
Hospital exemption
Named patient
Prijs- / vergoedingsarrangementen
Adaptive licensing is een mindset, geen regelgeving
Maar gaat adaptive licensing werken binnen huidige regulering?
Adaptive licensing
Wat moet nog opgelost?
•
•
•
•
•
•
•
•
•
Risico off-label gebruik
Tien jaar bescherming gaat direct lopen
Communicatie over en omgang met onzekerheid
Zoeken naar rol adviseur versus regulator
Lukt adaptive licensing bininen huidige regels?
Dominante aandacht voor risico’s
Prijsbepaling / vergoeding nationale aangelegenheid
Opzetten van goede patientenregistraties ism stakeholders
Gezamelijke wil om stap terug te doen indien resultaten uit
de dagelijkse praktijk tegen vallen
Adaptive licensing
“Adaptive Licensing”
HollandBIO Orphan Café
Sociëteit De Vereeniging, Mariaplaats 14, Utrecht
12th June 2014
Dr Anthony Hall
Rationale for adaptive licensing
•
•
•
•
Too few drugs reaching market
Increasing costs of development
Earlier access for high unmet needs
Binary nature of drug authorisation
• EMA roadmap to 2015
New drugs launched in the UK from 1982 to 2011
Eroom’s Law: The number of new drugs approved by the FDA
per billion dollars (inflation- adjusted) spent on R&D has
halved roughly every 9 years
From Scannell JW et al. Nature Reviews Drug Disc. 2012; 11: 191-200
Earlier access for high unmet needs
•
•
•
•
Strong voice of patient groups
Halpin Protocol
Saatchi Medical Innovation Bill
Early Access to Medicines Scheme
Knowledge; Investment
Binary nature of drug authorisation
Current model of licensing
“The Magic Moment”
Experimental
therapy
Fully vetted, safe &
efficacious therapy
Time
Adapted from Eichler, EMA, London, March 2012
Definition of Adaptive Licensing
AL can be defined as a prospectively
planned, adaptive approach to bringing
drugs to market.
’staggered approval’, ’progressive licensing’, and
’adaptive licensing’ are often used interchangeably
Definition of Adaptive Licensing
• Start with an authorised indication (e.g. “niche”
indication), through iterative phases of
evidence gathering and progressive licensing
adaptations.
• Seeks to maximize the positive impact of new
drugs on public health by balancing timely
access for patients with the need to provide
adequate evolving information on benefits and
harms.
Knowledge; Investment
Adaptive licensing
Updated MA
Updated MA
Initial MA
Time
Adapted from Eichler, EMA, London, March 2012
Roadmap to 2015
Strategic vision for the operation of the
EMA from 2011 to 2015.
“…a key issue for regulators will be whether a more 'staggered'
approval (or progressive licensing) concept should be envisaged for
situations not covered by conditional marketing authorisations or
marketing authorisations under exceptional circumstances…”
“It should be emphasised that progressive licensing should not lead
to a reduced level of evidence for first-time marketing
authorisation.”
International proposals for AL
•
•
•
•
•
•
Europe: “staggered approval”
US: progressive reduction of uncertainty
Canada: progressive authorization
Singapore: test bed for adaptive regulation
MIT NEWDIGS: adaptive licensing project
CASMI: streamline the research, licensing and
market access processes
Adapted from Eichler, EMA, London, March 2012
CASMI
• Centre for the Advancement of Sustainable
Medical Innovation
• Partnership between Oxford University and UCL
• Created to develop new models for medical
innovation
• Aims to address the issues that have led to
current failures in the translation of basic
bioscience into affordable and widely adopted
new treatments
MIT-NEWDIGS
• Massachusetts Institute of Technology (MIT)
• New Drug Development ParadIGmS
• International multistakeholder collaborative
developing approaches to AL
• Currently testing new ideas & models for drug
development in “live” demonstration projects
using actual pipeline candidates from
pharmaceutical companies
Legal framework for AL
• Using existing legal framework
(at least for now)
•
•
•
•
•
Scientific advice (multi-stakeholder)
Conditional Marketing Authorisation
MA under Exceptional Circumstances
Registries
Pharmacovigilance tools
From HALL & CARLSON. The current status of orphan drug development in
Europe and the US. Intractible & Rare Diseases Research. 2014; 3(1): 1-7
Pharmacovigilance tools
• Risk Management Plans (EU)
• Risk Evaluation and Mitigation Strategies (US)
• Periodic Safety Update reports
• 5-year renewal of MA
Serious vs non-serious conditions
• Acceptable level of uncertainty may be
higher for serious conditions with no
treatments available vs less serious
conditions with treatments available.
• Highly applicable to orphan drugs.
• Currently 14 drugs have CMA, of which 7
are orphan drugs & 6 oncolgy drugs
• One size doesn’t fit all.
Conservative vs transformative AL
Conservative
• Using exisiting legiaslation
• Small incremental steps
Transformative
• “Delinking” efficacy and safety populations
EMA Pilot Project on AL
• Launched 19 March 2014
• Purpose to provide a framework for
informal interactions by discussing ‘live
assets’ (medicines in early stage of
development)
• Sponsors invited to submit applicaitons
• Status as of 6 June 2014 (20 applications;
reviwed 9; accepted 2
For further information or to initiate a pilot case, contact
[email protected]
Involvement of all stakeholders
•
•
•
•
•
Regulators
Sponsor
HTA bodies
Groups responsible for guidelines
Patient organisations
Discusisons in a “safe harbour” environment
AL continues throughout life-cycle
Evidence generation
would encompass all
methodologies:
RCTs, other clinical trials,
observational studies,
registries & post-markeing
surveillance
Schematic reproduced from Lim. The
Path to Adaptive Drug Regulation.
IRGC International Conference 2013
Communication & perception
• New models of information for patients
• Interaction between patients, physicians &
pharmacists
• Differences in risk tolerance between
different cultures & social groups
Obstacles & disadvantages of AL
• Gradual market entry may lead to lower ROI
(but development time may be shorter / cheaper)
•
•
•
•
•
•
Concerns over reduced standards
Variable acceptability of increased uncertainty
Possible need for new legislation
Questions over industry commitment
Restricting prescriptions to the target popuation
Ethics of RCTs after MA
Obstacles & disadvantages of AL
• Difficulties with global launches
• Risk of litigation
• Need to collect data post-approval
•
•
•
Logistics
Costs
Need for CA / EC approval?
• Local reimbursement in EU
•
Problems of high-priced drugs with CMA
Comparison with Early Access to
Medicines Scheme (UK)
• UK launched scheme March 2014
• Promising Innovative Medicines (PIM)
designation
• Early Access to Medicines scientific opinon
• Late in development (close to launch)
• Drug must be provided free by company
• NICE has to assess after MA
Case studies
Case Study 1
Unlikely to be acceptable
• Open-label, non-inferiority study with soft
endpoints
• Approach lacks scientific rigour
Case Study 2
Broaden treatment-eligible population
• Novel anti-obesity drug
• Initially show benefit-risk in narrowly defined
group with high medical need
•
•
•
•
high expected responsiveness
acceptable side-effects
high treatment concordance
tightly controlled management
• Post CMA, further RCTs in wider population
Time course of evidence
generation and accrual rates of
patients treated with a new drug
under the current and adaptive
licensing (AL) scenarios.
(a) Current scenario; before the
license, nearly all patients
treated with the new drug are
enrolled in interventional studies.
Once the license is granted, the
treated population expands
rapidly.
(b) AL scenario; the number of
patients in prelicensing RCTs may
be smaller and the initial license
may be granted earlier than
under the current scenario.
Reproduced from Eichler et al. Clin.
Pharm. Ther. 2012; 91(3): 426-437
Case Study 3
Reduce statistical uncertainty
• Ideal for rare diseases
• Initial CMA based on small study with
larger alpha
• Inform patients of increased uncertainty
• Further RCTs likely impossible
• Knowledge expansion rely on uncontrolled
studies
The threshold approach to evidence generation when randomized controlled trials (RCTs) are
not practically or ethically feasible after an initial license.
The sponsor would initially conduct one (or possibly more) small RCTs. If successful, an initial
license may be granted on the basis of this limited information.
Subsequent uncontrolled observational studies would aim to show that the treatment
outcome under real-life conditions remains above a pre-agreed threshold.
Reproduced from Eichler et al. Clin. Pharm. Ther. 2012; 91(3): 426-437
Case Study 4
Enable new-new combination development
• May be unethical to compare combination
with individual components & SoC
• Initial licence based on combination vs SoC
• Post MA, contribution of individual
components assessed e.g. by randomised
withdrawal trials
Case Study 5
Ensure “real world” effectiveness
• e.g. Antimicrobial for multi-drug resistant
infections
• Initial MA based on non-clinical data plus
RCT in one of diseases
• “real world” assessment of larger number
of diseases
Case Study 6
Reduce uncertainty around surrogate EP
• Surrogate EPs are not always predictive
• “reasonably likely to predict a real clinical
benefit”
• Initial MA based on surrogate
• Further studies confirm effect on clinical EP
• CMA may close window for further RCTs
Case Study 6a
Nitisinone for alkaptonuria
• AKU is a metabolic disease affecting
tyrosine metabolism
• Accumluation of HGA leads to ochronosis
• Ochronotic symptoms usually appear in 4th
or 5th decade
• HGA is an obvious but unvalidated
surrogate
“Adaptive Licensing”
HollandBIO Orphan Café
Sociëteit De Vereeniging, Mariaplaats 14, Utrecht
12th June 2014
Dr Anthony Hall